Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Prostate ; 84(6): 539-548, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38173301

RESUMEN

BACKGROUND: Data on the utilization and effects of prebiopsy prostate multiparametric magnetic resonance imaging (mpMRI) to support its routine use in real-world setting are still scarce. OBJECTIVE: To evaluate the change of clinical practice of prebiopsy mpMRI over time, and assess its diagnostic accuracy. DESIGN, SETTING, AND PARTICIPANTS: We retrospectively analyzed data from 6168 patients who underwent primary prostate biopsy (PBx) between January 2011 and December 2021 and had prostate-specific antigen (PSA) values ranging from 3 to 100 ng/mL. INTERVENTION: Prebiopsy MRI at the time of PBx. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS: We performed general linear regression and to elucidate trends in the annual use of prebiopsy mpMRI and conducted multivariable logistic regression to evaluate the potential benefits of incorporating prebiopsy mpMRI for prostate cancer (PCa) detection. RESULTS AND LIMITATIONS: The utilization of prebiopsy mpMRI significantly increased from 9.2% in 2011 to 75.0% in 2021 (p < 0.001). In addition, prebiopsy mpMRI significantly reduced negative PBx by 8.6% while improving the detection of clinically significant PCa (csPCa) by 7.0%. Regression analysis showed that the utilization of prebiopsy mpMRI was significantly associated with a 48% (95% confidence interval [CI]: 1.19-1.84) and 36% (95% CI: 1.12-1.66) increased PCa detection rate in the PSA 3-10 ng/mL and 10-20 ng/mL groups, respectively; and a 34% increased csPCa detection rate in the PSA 10-20 ng/mL group (95% CI: 1.09-1.64). The retrospective design and the single center cohort constituted the limitations of this study. CONCLUSIONS: Our study demonstrated a notable rise in the utilization of prebiopsy mpMRI in the past decade. The adoption of this imaging technique was significantly associated with an increased probability of detecting prostate cancer. PATIENT SUMMARY: From 2011 to 2021, we demonstrated a steady increase in the utilization of prebiopsy mpMRI among biopsy-naïve men. We also confirmed the positive impact of prebiopsy mpMRI utilization on the detection of prostate cancer.


Asunto(s)
Imágenes de Resonancia Magnética Multiparamétrica , Neoplasias de la Próstata , Masculino , Humanos , Antígeno Prostático Específico , Próstata/diagnóstico por imagen , Próstata/patología , Estudios Retrospectivos , Neoplasias de la Próstata/diagnóstico por imagen , Neoplasias de la Próstata/patología , Imagen por Resonancia Magnética/métodos , Biopsia Guiada por Imagen/métodos
2.
Exp Cell Res ; 432(2): 113781, 2023 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-37722551

RESUMEN

The progression of cholestasis is characterized by excessive accumulation of bile acids (BAs) in the liver, which leads to oxidative stress (OS), inflammation and liver injury. There are currently limited treatments for cholestasis. Therefore, appropriate drugs for cholestasis treatment need to be developed. Dimethyl fumarate (DMF) has been widely used in the treatment of various diseases and exerts antioxidant and anti-inflammatory effects, but its effect on cholestatic liver disease remains unclarified. We fed mice 3,5-diethoxycarbonyl-1,4-dihydrocollidine or cholic acid to induce cholestatic liver injury and treated these mice with DMF to evaluate its protective ability. Alanine aminotransferase, aspartate aminotransferase, and total liver BAs were assessed as indicators of liver function. The levels of OS, liver inflammation, transporters and metabolic enzymes were also measured. DMF markedly altered the relative ALT and AST levels and enhanced the liver antioxidant capacity. DMF regulated the MST/NRF2 signaling pathway to protect against OS and reduced liver inflammation through the NLRP3/GSDMD signaling pathway. DMF also regulated the levels of BA transporters by promoting FXR protein expression. These findings provide new strategies for the treatment of cholestatic liver disorders.

3.
Cancer Sci ; 114(4): 1378-1395, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36519785

RESUMEN

The histone methyltransferase enhancer of zeste homolog 2 (EZH2) is overexpressed in a variety of malignancies including prostate cancer (PCa) and may play important roles in tumor progression. Gene copy number gains, enhanced transcription, and a few circRNAs have been reported to upregulate EZH2. It was not known whether EZH2 itself generates circRNAs that promote its own expression. We here report the identification of circEZH2E2/E3 that is derived from exons 2 and 3 of the EZH2 gene and overexpressed in PCa. We show that circEZH2E2/E3 functions as a dual inhibitor for both miR363 and miR708 that target the EZH2 3'UTR and CDS, respectively, resulting in the upregulation of EZH2 expression and hence the downregulation of EZH2-repressed genes (e.g., CDH1 and DAB2IP), and enhancement of PCa cell proliferation, migration, invasion, and xenograft PCa growth. Overexpression of circEZH2E2/E3 is significantly correlated with higher tumor grade, tumor progression, and unfavorable progression-free and disease-specific survival in PCa patients. These findings show a novel autoenhancing EZH2-circEZH2E2/E3 -miR363/miR708-EZH2 regulatory loop, by which circEZH2E2/E3 plays important roles in PCa tumorigenesis and progression by upregulating EZH2, and may have potential diagnostic, prognostic, and therapeutic uses in PCa management.


Asunto(s)
MicroARNs , Neoplasias de la Próstata , Masculino , Humanos , Proteína Potenciadora del Homólogo Zeste 2/genética , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Complejo Represivo Polycomb 2/genética , Complejo Represivo Polycomb 2/metabolismo , ARN Circular , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Neoplasias de la Próstata/patología , Proliferación Celular/genética , Proteínas Activadoras de ras GTPasa/metabolismo , MicroARNs/genética , MicroARNs/metabolismo
4.
Prostate ; 83(5): 440-453, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36541373

RESUMEN

BACKGROUND: The homeodomain-containing transcription factor NANOG is overexpressed in prostate adenocarcinoma (PCa) and predicts poor prognosis. The SOX family transcription factor SOX9, as well as the transcription co-activator HMGB3 of the HMGB family, are also overexpressed and may play pivotal roles in PCa. However, it is unknown whether SOX9 and HMGB3 interact with each other, or if they regulate NANOG gene transcription. METHODS: We identified potential SOX9 responsive elements in NANOG promoter, and investigated if SOX9 regulated NANOG transcription in co-operation with HMGB3 by experimental analysis of potential SOX9 binding sites in NANOG promoter, reporter gene transcription assays with or without interference or artificial overexpression of SOX9 and/or HMGB3, and protein-binding assays of SOX9-HMGB3 interaction. Clinicopathologic and prognostic significance of SOX9-HMGB3 overexpression in PCa was analyzed. RESULTS: SOX9 activated NANOG gene transcription by preferentially binding to a highly conserved consensus cis-regulatory element (-573 to -568) in NANOG promoter, and promoted the expression of NANOG downstream oncogenic genes. Importantly, HMGB3 functioned as a partner of SOX9 to co-operatively enhance transactivation of NANOG by interacting with SOX9, predominantly via the HMG Box A domain of HMGB3. Overexpression of SOX9 and/or HMGB3 enhanced PCa cell survival and cell migration and were significantly associated with PCa progression. Notably, Cox proportional regression analysis showed that co-overexpression of both SOX9 and HMGB3 was an independent unfavorable prognosticator for both CRPC-free survival (relative risk [RR] = 3.779,95% confidence interval [CI]: 1.159-12.322, p = 0.028) and overall survival (RR = 3.615,95% CI: 1.101-11.876, p = 0.034). CONCLUSIONS: These findings showed a novel SOX9/HMGB3/NANOG regulatory mechanism, deregulation of which played important roles in PCa progression.


Asunto(s)
Proteína HMGB3 , Proteína Homeótica Nanog , Neoplasias de la Próstata , Factor de Transcripción SOX9 , Humanos , Masculino , Regulación de la Expresión Génica , Proteína HMGB3/genética , Proteína HMGB3/metabolismo , Proteína Homeótica Nanog/genética , Proteína Homeótica Nanog/metabolismo , Procesos Neoplásicos , Próstata/metabolismo , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Factores de Transcripción/genética
5.
Mod Pathol ; 36(11): 100303, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37580017

RESUMEN

Fumarate hydratase (FH)-deficient renal cell carcinoma (RCC) is a rare and distinct subtype of renal cancer caused by FH gene mutations. FH negativity and s-2-succinocysteine (2SC) positivity on immunohistochemistry can be used to screen for FH-deficient RCC, but their sensitivity and specificity are not perfect. The expression of AKR1B10, an aldo-keto reductase that catalyzes cofactor-dependent oxidation-reduction reactions, in RCC is unclear. We compared AKR1B10, 2SC, and FH as diagnostic biomarkers for FH-deficient RCC. We included genetically confirmed FH-deficient RCCs (n = 58), genetically confirmed TFE3 translocation RCCs (TFE3-tRCC) (n = 83), clear cell RCCs (n = 188), chromophobe RCCs (n = 128), and papillary RCCs (pRCC) (n = 97). AKR1B10, 2SC, and FH were informative diagnostic markers. AKR1B10 had 100% sensitivity and 91.4% specificity for FH-deficient RCC. The nonspecificity of AKR1B10 was shown in 26.5% of TFE3-tRCCs and 21.6% of pRCCs. 2SC showed 100% sensitivity and 88.9% specificity. However, nonspecificity for 2SC was evident in multiple RCCs, including pRCC, TFE3-tRCC, clear cell RCCs, and chromophobe RCCs. FH was 100% specific but 84.5% sensitive. AKR1B10 served as a highly sensitive and specific diagnostic biomarker. Our findings suggest the value of combining AKR1B10 and 2SC to screen for FH-deficient RCC. AKR1B10+/2SC+/FH- cases can be diagnosed as FH-deficient RCC. Patients with AKR1B10+/2SC+/FH+ are highly suspicious of FH-deficient RCC and should be referred for FH genetic tests.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Humanos , Carcinoma de Células Renales/patología , Fumarato Hidratasa/genética , Fumarato Hidratasa/metabolismo , Neoplasias Renales/patología , Factores de Transcripción , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice , Aldo-Ceto Reductasas
6.
J Transl Med ; 21(1): 840, 2023 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-37993879

RESUMEN

Fibroblast growth factor receptor 1 (FGFR1) is a core component of the FGFs/FGFR pathway that activates multiple signalling pathways, including ERK1/2, PI3K/AKT, PLCγ, and NF-κB. Aberrant expression of FGFR1 due to gene amplification, chromosome rearrangement, point mutation, and epigenetic deregulations, have been reported in various cancers. FGFR1 overexpression has also been reported in prostate cancer (PCa), but the underlining mechanisms are not clear. Here we report a novel circular RNA, circFGFR1int2, derived from intron 2 of FGFR1 gene, which is overexpressed in PCa and associated with tumor progression. Importantly, we show that circFGFR1int2 facilitates FGFR1 transcription by recruiting transcription activators P65/FUS and by interacting with FGFR1 promoter. Moreover, we show that circFGFR1int2 suppresses post-transcriptional inhibitory effects of miR-4687-5p on FGFR1 mRNA. These mechanisms synergistically promote PCa cell growth, migration, and invasion. Overexpression of circFGFR1int2 is significantly correlated with higher tumor grade, Gleason score, and PSA level, and is a significant unfavorable prognosticator for CRPC-free survival (CFS) (RR = 3.277, 95% confidence interval: 1.192-9.009; P = 0.021). These findings unravelled novel mechanisms controlling FGFR1 gene expression by intronic circRNA and its potential clinicopathological utility as a diagnostic or therapeutic target.


Asunto(s)
MicroARNs , Neoplasias de la Próstata , Masculino , Humanos , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , ARN Circular/genética , Intrones/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Neoplasias de la Próstata/patología , Proliferación Celular/genética , MicroARNs/genética , MicroARNs/metabolismo , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Proteína FUS de Unión a ARN/genética , Proteína FUS de Unión a ARN/metabolismo
7.
BJU Int ; 129(3): 345-355, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34185954

RESUMEN

OBJECTIVES: To investigate the genetic alterations of patients with prostate cancer (PCa) with and without intraductal carcinoma of the prostate (IDC-P). PATIENTS AND METHODS: We performed targeted sequencing of plasma cell-free DNA on 161 patients with prostate adenocarcinoma (PAC) with IDC-P and 84 without IDC-P. Genomic alterations were compared between these two groups. The association between genetic alterations and patients' survival outcomes was also explored. RESULTS: We identified that 29.8% (48/161) and 21.4% (18/84) of patients with and without IDC-P harboured genomic alterations in DNA repair pathways, respectively (P = 0.210). Pathogenic germline DNA repair alterations were frequently detected in IDC-P carriers compared to IDC-P non-carriers (11.8% [19/161] vs 2.4% [two of 84], P = 0.024). Germline BReast CAncer type 2 susceptibility protein (BRCA2) and somatic cyclin-dependent kinase 12 (CDK12) defects were specifically identified in IDC-P carriers relative to PAC (BRCA2: 8.7% [14/161] vs 0% and CDK12: 6.8% [11/161] vs 1.2% [one of 84]). Patients with IDC-P had a distinct androgen receptor (AR) pathway alteration, characterised by an enrichment of nuclear receptor corepressor 2 (NCOR2) mutations compared with patients with pure PAC (21.1% [34/161] vs 6.0% [five of 84], P = 0.004). Increased AR alterations were detected in patients harbouring tumours with an IDC-P proportion of ≥10% vs those with an IDC-P proportion of <10% (6.4% [five of 78] vs 18.1% [15/83], P = 0.045). For IDC-P carriers, tumour protein p53 (TP53) mutation was associated with shorter castration-resistant-free survival (median 10.9 vs 28.9 months, P = 0.026), and BRCA2 alteration was related to rapid prostate-specific antigen progression for those receiving abiraterone treatment (median 9.1 vs 11.9 months, P = 0.036). CONCLUSION: Our findings provide genomic evidence explaining the aggressive phenotype of tumours with IDC-P, highlighting the potential therapeutic strategies for this patient population.


Asunto(s)
Carcinoma Intraductal no Infiltrante , ADN Tumoral Circulante , Neoplasias de la Próstata , Carcinoma Intraductal no Infiltrante/genética , Carcinoma Intraductal no Infiltrante/patología , ADN Tumoral Circulante/genética , Humanos , Masculino , Fenotipo , Próstata/patología , Neoplasias de la Próstata/patología
8.
BMC Pulm Med ; 22(1): 468, 2022 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-36476475

RESUMEN

BACKGROUND: Extra-pulmonary multi-organ failure in patients with severe acute respiratory distress syndrome (ARDS) is a major cause of high mortality. Our purpose is to assess whether airway pressure release ventilation (APRV) causes more multi-organ damage than low tidal volume ventilation (LTV). METHODS: Twenty one pigs were randomized into control group (n = 3), ARDS group (n = 3), LTV group (n = 8) and APRV group (n = 7). Severe ARDS model was induced by repeated bronchial saline lavages. Pigs were ventilated and monitored continuously for 48 h. Respiratory data, hemodynamic data, serum inflammatory cytokines were collected throughout the study. Histological injury and apoptosis were assessed by two pathologists. RESULTS: After severe ARDS modeling, pigs in ARDS, LTV and APRV groups experienced significant hypoxemia and reduced lung static compliance (Cstat). Oxygenation recovered progressively after 16 h mechanical ventilation (MV) in LTV and APRV group. The results of the repeated measures ANOVA showed no statistical difference in the PaO2/FiO2 ratio between the APRV and LTV groups (p = 0.54). The Cstat showed a considerable improvement in APRV group with statistical significance (p < 0.01), which was significantly higher than in the LTV group since 16 h (p = 0.04). Histological injury scores showed a significantly lower injury score in the middle and lower lobes of the right lung in the APRV group compared to LTV (pmiddle = 0.04, plower = 0.01), and no significant increase in injury scores for extra-pulmonary organs, including kidney (p = 0.10), small intestine (p = 1.0), liver (p = 0.14, p = 0.13) and heart (p = 0.20). There were no significant differences in serum inflammatory cytokines between the two groups. CONCLUSION: In conclusion, in the experimental pig models of severe ARDS induced by repetitive saline lavage, APRV improved lung compliance with reduced lung injury of middle and lower lobes, and did not demonstrate more extra-pulmonary organ injuries as compared with LTV.


Asunto(s)
Presión de las Vías Aéreas Positiva Contínua , Síndrome de Dificultad Respiratoria , Porcinos , Animales , Apoptosis , Síndrome de Dificultad Respiratoria/terapia
9.
Prostate ; 81(15): 1191-1201, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34435696

RESUMEN

BACKGROUND: To explore whether metastatic castration-resistant prostate cancer (mCRPC) patients with distinct intraductal carcinoma of the prostate (IDC-P) subtypes respond differently to abiraterone and docetaxel treatment. METHODS: We retrospectively analyzed data of 170 mCRPC patients receiving abiraterone or docetaxel as first-line therapy. PSA response, PSA progression-free survival (PSA-PFS), radiographic progression-free survival (rPFS), and overall survival (OS) were analyzed based on the presence of IDC-P and its subpatterns. RESULTS: IDC-P was confirmed in 91/170 (53.5%) patients. Among them 36/91 (39.6%) and 55/91 (60.4%) harbored IDC-P patterns 1 and 2, respectively. Patients with IDC-P pattern 1 shared similar clinical outcomes to those without IDC-P in both abiraterone and docetaxel treatment. However, against cases without IDC-P or with IDC-P pattern 1, patients with IDC-P pattern 2 had markedly poorer prognosis in either abiraterone (mPSA-PFS: 11.9 vs. 11.1 vs. 6.1 months, p < 0.001; mrPFS: 18.9 vs. 19.4 vs. 9.6 months, p < 0.001) or docetaxel (mPSA-PFS: 6.2 vs. 6.6 vs. 3.0 months, p < 0.001; mrPFS: 15.1 vs. 12.6 vs. 5.5 months, p < 0.001) treatment. For patients without IDC-P, docetaxel had comparable therapeutic efficacy with abiraterone. However, the efficacy of docetaxel was significantly inferior to abiraterone in patients with either IDC-P pattern 1 (mPSA-PFS: 6.6 vs. 11.1 months, p = 0.021; mrPFS: 12.6 vs. 19.4 months, p = 0.027) or pattern 2 (mPSA-PFS: 3.0 vs. 6.1 months, p = 0.003; mrPFS: 5.5 vs. 9.6 months, p = 0.007). CONCLUSION: Compared to docetaxel, abiraterone exhibited better efficacy in patients with IDC-P of either pattern. However, IDC-P pattern 2 responded unsatisfactorily to either abiraterone or docetaxel therapy. Novel therapeutic strategies for IDC-P pattern 2 need further investigations.


Asunto(s)
Androstenos/uso terapéutico , Antineoplásicos/uso terapéutico , Docetaxel/uso terapéutico , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata/tratamiento farmacológico , Anciano , Supervivencia sin Enfermedad , Humanos , Masculino , Persona de Mediana Edad , Supervivencia sin Progresión , Próstata/patología , Neoplasias de la Próstata/mortalidad , Neoplasias de la Próstata/patología , Neoplasias de la Próstata Resistentes a la Castración/mortalidad , Neoplasias de la Próstata Resistentes a la Castración/patología , Estudios Retrospectivos , Tasa de Supervivencia , Resultado del Tratamiento
10.
Neuropathology ; 40(6): 599-605, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32761642

RESUMEN

Glioma is the most common intracranial malignant tumor, with poor prognosis. The new World Health Organization (WHO) integrated classification (2016) for diffuse glioma is mainly based on the status of the isocitrate dehydrogenase (IDH) gene (IDH) mutation and 1p/19q codeletion, with diffuse glioma separated into three distinct molecular categories: chromosome 1p/19q codeletion/IDH mutant, 1p/19q intact /IDH mutant, and IDH wild-type. Gliomas harboring 1p/19q codeletion but without IDH mutation are rare and cannot be classified according to the new revision of the WHO classification. Here we report three high-grade gliomas with this atypical molecular phenotype, and describe their histological and immunohistochemical features, the status of mutations in TERT promopter, H3F3A, HIST1H3B, and BRAF, as well as MGMT promoter methylation, and prognosis. Considering morphology, molecular parameters, and patients prognosis, we found that high-grade gliomas harboring 1p/19q codeletion but without IDH mutation were not typical glioblastoma multiforme (GBM) but were more likely to be GBM than anaplastic oligodendroglioma.


Asunto(s)
Neoplasias Encefálicas/genética , Cromosomas Humanos Par 1/genética , Glioma/genética , Adulto , Neoplasias Encefálicas/patología , Aberraciones Cromosómicas , Femenino , Glioma/patología , Humanos , Isocitrato Deshidrogenasa , Masculino , Persona de Mediana Edad
11.
Prostate ; 79(8): 819-825, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30900303

RESUMEN

BACKGROUND: Overexpression of ERG protein resulting from TMPRSS2:ERG rearrangement is highly specific for prostate cancer (PCa). However, the biological function of this fusion protein and its relationship with clinicopathological features still remain controversial. METHOD: In this study, we evaluated ERG protein expression/gene rearrangement and heterogeneity by immunohistochemistry (IHC) and fluorescence in situ hybridization (FISH) in a cohort of 633 consecutive PCa initially diagnosed by core-needle biopsy in the West China Hospital. RESULT: Overall, ERG protein expression was detected in 16.7% (106 of 633) cases, and frequently observed in PCa patients less than 60 years of age (31.9% vs 15.5%, P = 0.004) and in PCa with Gleason score less than 8 (20.0% vs 13.4%, P = 0.027), but infrequently observed in cases with intraductal carcinoma of the prostate (IDC-P) (10.0% vs 18.6%, P = 0.012). Follow-up analysis found that patients who progressed to castration-resistant prostate cancer (CRPC) have a lower frequency of ERG protein expression at initial biopsies compared to androgen deprivation therapy (ADT)-sensitive cases (14.1% vs 23.5%, P = 0.042), but Kaplan-Meier curve showed that ERG protein expression was not an independent prognostic marker. Of all the 106 ERG-positive cases, eight cases (7.5%) exhibited heterogeneous expression of ERG protein, in which ERG was only positive in tumors with Gleason pattern 3, but negative in Gleason pattern 4. The FISH analysis was consistent with IHC in six of these cases. In the other two cases, ERG rearrangement was detected in tumors with both Gleason pattern 3 and 4 by FISH, despite the negative protein expression in Gleason pattern 4. In case 1, a repeated biopsy was performed when the disease progressed to CRPC, and no ERG-positive cells were identified neither by IHC nor FISH. CONCLUSION: This was by far the largest series of ERG expression and heterogeneity analysis in Chinese PCa. The ERG rearrangement seemed to be frequently expressed in patients with relatively younger age and lower Gleason score and infrequently expressed in PCa with the IDC-P. PCa with positive ERG were less frequently to progress to CRPC, but there was no prognostic significance of ERG expression. In heterogeneous cases, ERG protein was detectable only in tumors with Gleason pattern 3 but not in pattern 4. Tumor cells with positive ERG expression/rearrangement seemed easily response to ADT.


Asunto(s)
Carcinoma Intraductal no Infiltrante/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Neoplasias de la Próstata/metabolismo , Biopsia con Aguja Gruesa/métodos , Carcinoma Intraductal no Infiltrante/genética , Carcinoma Intraductal no Infiltrante/patología , Estudios de Cohortes , Expresión Génica , Reordenamiento Génico , Heterogeneidad Genética , Humanos , Biopsia Guiada por Imagen/métodos , Inmunohistoquímica , Hibridación Fluorescente in Situ , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Pronóstico , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/patología , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Regulador Transcripcional ERG/biosíntesis , Regulador Transcripcional ERG/genética , Carga Tumoral
12.
Prostate ; 79(14): 1673-1682, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31433509

RESUMEN

BACKGROUND: The prognostic value of ductal adenocarcinoma of the prostate (DA) in nonmetastatic prostate cancer (PCa) has been identified in many studies. However, it remains unknown whether DA is an adverse prognostic factor in metastatic PCa (mPCa). METHOD: Data from 634 mPCa patients histopathologically documented with DA or/and acinar adenocarcinoma of the prostate in our center between 2012 and 2018 were retrospectively analyzed. Propensity score matching (PSM) was used to balance the baseline features. Data from the Surveillance, Epidemiology, and End Results (SEER) database were utilized to validate our findings. Castration-resistant PCa-free survival (CFS), overall survival (OS), and cancer-specific survival (CSS) were set as endpoints. RESULTS: DA was confirmed in 35 of 634 (5.5%) patients. Among the DA-positive patients, 7 of 35 (20%) and 28 of 35 (80%) harbored high (DA ≥ 50%) and low (DA < 50%) DA components, respectively. DA was not associated with poorer median CFS (mCFS) or median OS (mOS) either before PSM (mCFS: 16.9 vs 18.4 month, P = .814; mOS: 42.0 vs 70.1 month, P = .796) or after PSM (mCFS: 16.9 vs 16.9 month, P = .949; mOS: 42.0 vs 79.9 month, P = .960). Likewise, in the SEER data, DA-positive patients (n = 15 153) shared similar median CSS (25.0 vs 28.0 month, P = .206) and OS (26.0 vs 35.0 month, P = .095) with DA-negative patients (n = 70). No prognostic difference was found between patients with high and low DA components. CONCLUSION: We conducted the first study investigating the prognostic value of DA in de novo mPCa. DA was not associated with adverse clinical outcomes in mPCa patients. These findings are helpful for prognostic evaluation, treatment decision making and counseling in mPCa patients.


Asunto(s)
Carcinoma Ductal/patología , Metástasis de la Neoplasia/patología , Neoplasias de la Próstata/patología , Anciano , Humanos , Masculino , Metástasis de la Neoplasia/terapia , Pronóstico , Neoplasias de la Próstata/mortalidad , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/mortalidad , Neoplasias de la Próstata Resistentes a la Castración/patología , Estudios Retrospectivos , Programa de VERF , Tasa de Supervivencia
13.
Prostate ; 79(13): 1553-1562, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31294486

RESUMEN

BACKGROUND: Previous studies had demonstrated that aldo-keto reductase family 1 member C3 (AKR1C3), a crucial enzyme in the steroidogenic pathway, played an important role in abiraterone (ABI)-resistance in metastatic castration-resistant prostate cancer (mCRPC) by increasing intratumoral androgen synthesis. However, its value in predicting treatment response in patients with mCRPC is unknown. METHOD AND MATERIALS: Data of 163 patients with metastatic prostate cancer between 2016 and 2018 were retrospectively analyzed. All patients received androgen deprivation therapy plus bicalutamide after initial diagnosis. After mCRPC, either ABI or docetaxel (DOC) treatment was used. No patient had the experience of therapy to the primary tumor. AKR1C3 protein was detected by immunohistochemical staining from rebiopsy (re-Bx) of primary prostate lesions at mCRPC. Kaplan-Meier curves and Cox regression were used to analyze the association between AKR1C3 and treatment outcomes. RESULTS: AKR1C3 was positive in 58 of 163 (35.6%) cases. AKR1C3 was associated with significantly shorter median prostate-specific antigen progression-free survival (mPSA-PFS, 5.6 mo vs 10.7 mo; P < .001), median radiographic progression-free survival (mrPFS, 11.1 mo vs 18.0 mo; P = .018), and numerically shorter median overall survival (mOS, 20.4 mo vs 26.4 mo; P = .157). Notably, AKR1C3-positive patients treated with ABI, but not DOC, had shorter mPSA-PFS and mrPFS compared with AKR1C3-negative men, (mPSA-PFS, 5.7 mo vs. 11.2 mo; P < .001; mrPFS, 12.4 mo vs 23.3 mo; P = .048). However, AKR1C3 expression had no correlation to PSA response or OS. Multivariate Cox regression indicated that AKR1C3 was independently accompanied with rapid PSA progression (hazard ratio [HR], 3.64; 95% confidence interval [CI], 2.10-6.31; P < 0.001) and radiological progression (HR, 2.08; 95% CI, 1.05-4.11; P = .036) in the ABI-treated subgroup. CONCLUSION: This study demonstrated that AKR1C3 detection in tissues from prostate re-Bx at mCRPC was associated with early resistance to ABI but not DOC. These results will help to make optimal personalized treatment decisions for patients with mCRPC, facilitate physicians predicting the effectiveness of ABI.


Asunto(s)
Miembro C3 de la Familia 1 de las Aldo-Ceto Reductasas/metabolismo , Androstenos/uso terapéutico , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/enzimología , Anciano , Miembro C3 de la Familia 1 de las Aldo-Ceto Reductasas/biosíntesis , Androstenos/administración & dosificación , Androstenos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Docetaxel/administración & dosificación , Resistencia a Antineoplásicos , Humanos , Biopsia Guiada por Imagen , Inmunohistoquímica , Masculino , Metástasis de la Neoplasia , Prednisona/administración & dosificación , Neoplasias de la Próstata Resistentes a la Castración/patología , Estudios Retrospectivos
14.
J Urol ; 201(4): 759-768, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30652989

RESUMEN

PURPOSE: Intraductal carcinoma of the prostate is an adverse prognosticator of prostate cancer. However, the roles of proportion and architectural patterns of intraductal prostate carcinoma in patient outcomes remain unclear. MATERIALS AND METHODS: We retrospectively analyzed data on 644 patients with de novo metastatic prostate cancer between 2010 and 2017. Intraductal carcinoma of the prostate was identified from 12-core prostate biopsy. We calculated the proportion of intraductal prostate carcinoma and identified patterns according to the 2016 WHO classification. Propensity score matching was performed to balance baseline characteristics between patients with and without intraductal prostate carcinoma. Kaplan-Meier curves and Cox regression were used for survival analyses. The end points were castration resistant prostate cancer-free survival and overall survival. RESULTS: Of the 644 patients 180 (28.0%) harbored intraductal carcinoma of the prostate. A 10% or greater incidence of the carcinoma was independently associated with worse prognosis (castration resistant prostate cancer-free survival HR 2.06, 95% CI 1.51-2.81, p <0.001, and overall survival HR 2.52, 95% CI 1.52-4.16, p <0.001), as was pattern 2 intraductal carcinoma of the prostate (HR 1.86, 95% CI 1.40-2.49, p <0.001, and HR 2.12, 95% CI 1.29-3.46, p = 0.003, respectively). Based on these 2 risk factors all men were classified into 5 groups. Patients in group 0 (no intraductal carcinoma of the prostate) and prostate intraductal carcinoma group 1 (less than 10% intraductal carcinoma, pattern 1) had favorable median castration resistant prostate cancer-free survival (18.0 vs 16.9 months, p = 0.871) and median overall survival (neither reached, p = 0.698). Men in intraductal carcinoma of the prostate group 4 (10% or greater intraductal carcinoma, pattern 2) harbored the worst outcomes (median castration resistant prostate cancer-free and overall survival 8.4 and 29.9 months, respectively). Group 2 (less than 10% intraductal carcinoma, pattern 2, with median castration resistant prostate cancer-free and overall survival 14.2 and 45.9 months) and group 3 (10% or less prostate intraductal carcinoma, pattern 1, with median castration resistant prostate cancer-free and overall survival 11.9 and 39.7 months, respectively) had an intermediate prognosis. CONCLUSIONS: A 10% or greater proportion of intraductal carcinoma of the prostate and pattern 2 were 2 unfavorable prognosticators of metastatic prostate cancer. Pathological reporting criteria based on intraductal carcinoma of the prostate could improve the prediction of patient outcomes and optimize treatment decisions.


Asunto(s)
Carcinoma Intraductal no Infiltrante/patología , Neoplasias de la Próstata/patología , Anciano , Humanos , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia , Pronóstico , Estudios Retrospectivos
15.
Prostate ; 78(5): 343-352, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29341215

RESUMEN

BACKGROUND: The ERK signaling pathway is frequently deregulated in tumorigenesis, mostly by classical mechanisms such as gene mutation of its components (eg, RAS and RAF). However, whether and how multiple key components of ERK pathway are regulated by microRNAs are not clear. METHODS: We firstly predicted post-transcriptional regulation of multiple key components of the ERK signaling pathway by miR181c through bioinformatics analysis, and then confirmed the post-transcriptional regulation by dual luciferase reporter gene assays and Western blot analysis. The biological effects of miR181c on prostate cancer cell proliferation, apoptosis, migration, and invasion were measured by CCK-8 assay, flow cytometry, wound scratch assay, transwell cell migration, and invasion assays. RESULTS: miR181c post-transcriptionally regulated multiple key members of the ERK signaling pathway, including extracellular signal-regulated kinase 2 (ERK2), ribosomal S6 kinase 2 (RSK2), serum response factor (SRF), and FBJ murine osteosarcoma viral oncogene homolog (c-Fos). Ectopic expression of miR181c mimics effectively suppressed prostate cancer cell proliferation, migration, and invasion, but promoted cell apoptosis. Furthermore, miR181c treatment combined with the multi-kinase inhibitor sorafenib significantly enhanced these anti-tumor effects. CONCLUSIONS: Downregulation of miR181c results in deregulated ERK signaling and promotes prostate cancer cell growth and metastasis.


Asunto(s)
Sistema de Señalización de MAP Quinasas , MicroARNs/metabolismo , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Regulación hacia Abajo , Humanos , Masculino , MicroARNs/genética , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Invasividad Neoplásica , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/patología , Proteínas Quinasas S6 Ribosómicas 90-kDa/antagonistas & inhibidores , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo , Factor de Respuesta Sérica/antagonistas & inhibidores , Factor de Respuesta Sérica/metabolismo , Sorafenib/farmacología
16.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 35(2): 160-164, 2018 Apr 10.
Artículo en Zh | MEDLINE | ID: mdl-29652983

RESUMEN

OBJECTIVE: To explore the role of HIF1α gene in prostate cancer cell line DU145 by knocking it out with a novel gene-editing tool CRISPR/cas9 system. METHODS: A CRISPR/cas9 system with two sgRNAs targeting exon 1 of the HIF1α gene was constructed for the knock out experiment. CCK8 assay and transwell experiment were carried out to assess the effect of the knock out on the proliferation, migration and invasiveness of DU145 cells. RESULTS: The efficiency of gene-targeting was measured through a T7E1 assaying and sequence analysis, which confirmed that the partial knock out was successful and has led to a significant decrease in the expression of HIF1α and inhibition of cell proliferation, migration and invasiveness. CONCLUSION: A CRISPR/cas9 system for the knock out of HIF1α has been successfully constructed, which could inhibit the proliferation and migration of DU145 cells. The system can facilitate further studies of the HIF1α gene and its roles in tumorigenesis.


Asunto(s)
Sistemas CRISPR-Cas/genética , Edición Génica , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Neoplasias de la Próstata/patología , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Masculino , Invasividad Neoplásica
17.
Mol Cancer ; 16(1): 142, 2017 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-28830551

RESUMEN

BACKGROUND: Emerging studies show that long noncoding RNAs (lncRNAs) play important roles in carcinogenesis and cancer progression. The lncRNA ZEB1 antisense 1 (ZEB1-AS1) derives from the promoter region of ZEB1 and we still know little about its expressions, roles and mechanisms. METHODS: RACE was used to obtain the sequence of ZEB1-AS1. RNA interference was used to decrease ZEB1-AS1 expression. Adenovirus expression vector was used to increase ZEB1-AS1 expression. CHIP and RIP were used to detect the epigenetic mechanisms by which ZEB1-AS1 regulated ZEB1. CCK8 assay, wound healing assay and transwell assay were used to measure proliferation and migration of prostate cancer cells. RESULTS: In this study, in prostate cancer cells, we found that RNAi-mediated downregulation of ZEB1-AS1 induced significant ZEB1 inhibition while artificial overexpression of ZEB1-AS1 rescued ZEB1 expression, which means that ZEB1-AS1 promotes ZEB1 expression. Also, ZEB1-AS1 indirectly inhibited miR200c, the well-known target of ZEB1, and upregulated miR200c's target BMI1. Mechanistically, ZEB1-AS1 bound and recruited histone methyltransferase MLL1 to the promoter region of ZEB1, induced H3K4me3 modification therein, and activated ZEB1 transcription. Biologically, ZEB1-AS1 promoted proliferation and migration of prostate cancer cells. CONCLUSIONS: Collectively, ZEB1-AS1 functions as an oncogene in prostate cancer via epigenetically activating ZEB1 and indirectly regulating downstream molecules of ZEB1.


Asunto(s)
Neoplasias de la Próstata/metabolismo , ARN Largo no Codificante/metabolismo , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Epigénesis Genética/genética , Humanos , Masculino , MicroARNs/metabolismo , Neoplasias de la Próstata/genética , ARN Largo no Codificante/genética , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/genética
18.
Environ Sci Technol ; 50(2): 643-52, 2016 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-26713547

RESUMEN

Deepwater Horizon was the largest marine oil spill in U.S. waters, oiling large expanses of coastal wetland shorelines. We compared marsh periwinkle (Littoraria irrorata) density and shell length at salt marsh sites with heavy oiling to reference conditions ∼16 months after oiling. We also compared periwinkle density and size among oiled sites with and without shoreline cleanup treatments. Densities of periwinkles were reduced by 80-90% at the oiled marsh edge and by 50% in the oiled marsh interior (∼9 m inland) compared to reference, with greatest numerical losses of periwinkles in the marsh interior, where densities were naturally higher. Shoreline cleanup further reduced adult snail density as well as snail size. Based on the size of adult periwinkles observed coupled with age and growth information, population recovery is projected to take several years once oiling and habitat conditions in affected areas are suitable to support normal periwinkle life-history functions. Where heavily oiled marshes have experienced accelerated erosion as a result of the spill, these habitat impacts would represent additional losses of periwinkles. Losses of marsh periwinkles would likely affect other ecosystem processes and attributes, including organic matter and nutrient cycling, marsh-estuarine food chains, and multiple species that prey on periwinkles.


Asunto(s)
Contaminación por Petróleo/efectos adversos , Caracoles/efectos de los fármacos , Caracoles/fisiología , Contaminantes Químicos del Agua/toxicidad , Distribución por Edad , Animales , Golfo de México , Louisiana , Densidad de Población , Humedales
19.
Prostate ; 75(8): 855-62, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25683382

RESUMEN

BACKGROUND: The downregulation of the tumor suppressor miR200c plays important roles in many malignant tumors. This study aims to show that miR200c is a posttranscriptional regulator of insulin receptor substrate 1 (IRS1) and over-expression of miR200c suppresses prostate cancer cell growth. METHODS: Bioinformatics analysis was used to show potential post-translational regulation of IRS1 by miR200c. Dual reporter gene assays were chosen to test the binding of miR200c to the potential seed sequences in IRS1 3'UTR. RT-PCR, Q-PCR and western blot were applied to determine the regulation effect of miR200c on IRS1. CCK8 assay, soft agar assay, trypan blue exclusion assay and flow cytometric analysis were used to measure the biological effects of miR200c on prostate cancer cell proliferation and apoptosis. RESULTS: The 449-455 nt, 3061-3067 nt, and 3096-3102 nt of the IRS1 3'-UTR were identified as three potential seed sequences for miR200c. MiR200c directly binds to IRS1 through the seed sequences in IRS1 3'-UTR. Artificial overexpression of miR200c significantly downregulated the mRNA and protein levels of IRS1, together with decreased cell proliferation and increased cell death of PC3 and DU145 cells. CONCLUSIONS: Our results suggest that miR200c plays crucial roles in prostate cancer by post-transcriptional regulation of IRS1. The mir200c/IRS1 pathway may be a potential therapeutic target to prevent prostate cancer cell growth.


Asunto(s)
Aumento de la Célula , Proteínas Sustrato del Receptor de Insulina/biosíntesis , MicroARNs/biosíntesis , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/prevención & control , Apoptosis/fisiología , Muerte Celular/fisiología , Línea Celular Tumoral , Humanos , Masculino , Neoplasias de la Próstata/patología , ARN Mensajero/biosíntesis
20.
Analyst ; 140(17): 6054-60, 2015 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-26194058

RESUMEN

Cisplatin is a widely used anti-cancer agent, which was believed to trigger apoptosis of cancer cells by forming DNA adducts. However, recent studies evidenced a cisplatin-induced extrinsic apoptotic pathway through interaction with plasma membranes. We present quantitative time-course imaging of cisplatin-induced permeation of ferrocenemethanol to single live human bladder cancer cells (T24) using scanning electrochemical microscopy (SECM). Simultaneous quantification of cellular topography and membrane permeability was realized by running SECM in the depth scan mode. It was demonstrated that the acute addition of cisplatin to the outer environment of T24 cells immediately induced membrane permeability change in 5 min, which indicated a loosened structure of the cellular membrane upon cisplatin dosage. The cisplatin-induced permeation of T24 cells might be a one-step action, an extrinsic mechanism, since the cell response was quick, and no continuous increase in the membrane permeability was observed. The time-lapse SECM depth scan method provided a simple and facile way of monitoring cisplatin-induced membrane permeability changes. Our study is anticipated to lead to a methodology of screening anti-cancer drugs through their interactions with live cells.


Asunto(s)
Antineoplásicos/toxicidad , Permeabilidad de la Membrana Celular/efectos de los fármacos , Cisplatino/toxicidad , Microscopía Electroquímica de Rastreo , Línea Celular Tumoral , Compuestos Ferrosos/química , Compuestos Ferrosos/metabolismo , Humanos , Imagen de Lapso de Tiempo , Neoplasias de la Vejiga Urinaria/metabolismo , Neoplasias de la Vejiga Urinaria/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA