Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
BMC Med ; 21(1): 453, 2023 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-37993934

RESUMEN

BACKGROUND: The epididymis is crucial for post-testicular sperm development which is termed sperm maturation. During this process, fertilizing ability is acquired through the epididymis-sperm communication via exchange of protein and small non-coding RNAs (sncRNAs). More importantly, epididymal-derived exosomes secreted by the epididymal epithelial cells transfer sncRNAs into maturing sperm. These sncRNAs could mediate intergenerational inheritance which further influences the health of their offspring. Recently, the linkage and mechanism involved in regulating sperm function and sncRNAs during epididymal sperm maturation are increasingly gaining more and more attention. METHODS: An epididymal-specific ribonuclease T2 (RNase T2) knock-in (KI) mouse model was constructed to investigate its role in developing sperm fertilizing capability. The sperm parameters of RNase T2 KI males were evaluated and the metabolic phenotypes of their offspring were characterized. Pandora sequencing technology profiled and sequenced the sperm sncRNA expression pattern to determine the effect of epididymal RNase T2 on the expression levels of sperm sncRNAs. Furthermore, the expression levels of RNase T2 in the epididymal epithelial cells in response to environmental stress were confirmed both in vitro and in vivo. RESULTS: Overexpression of RNase T2 caused severe subfertility associated with astheno-teratozoospermia in mice caput epididymis, and furthermore contributed to the acquired metabolic disorders in the offspring, including hyperglycemia, hyperlipidemia, and hyperinsulinemia. Pandora sequencing showed altered profiles of sncRNAs especially rRNA-derived small RNAs (rsRNAs) and tRNA-derived small RNAs (tsRNAs) in RNase T2 KI sperm compared to control sperm. Moreover, environmental stress upregulated RNase T2 in the caput epididymis. CONCLUSIONS: The importance was demonstrated of epididymal RNase T2 in inducing sperm maturation and intergenerational inheritance. Overexpressed RNase T2 in the caput epididymis leads to astheno-teratozoospermia and metabolic disorder in the offspring.


Asunto(s)
Enfermedades Metabólicas , ARN Pequeño no Traducido , Ratones , Animales , Masculino , Epidídimo/metabolismo , Semen , Espermatozoides/metabolismo , ARN Pequeño no Traducido/genética , ARN Pequeño no Traducido/metabolismo
2.
J Urol ; 206(3): 630-637, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33904759

RESUMEN

PURPOSE: We aimed to explore the association between genomic status and clinical outcome of platinum-based chemotherapy among patients with metastatic castration-resistant prostate cancer (mCRPC). MATERIALS AND METHODS: We conducted a retrospective study of 55 patients with mCRPC who received platinum-based chemotherapy after the progression to docetaxel chemotherapy and underwent genomic profiling of 14 homologous recombination (HR) pathway genes. Progression-free survival (PFS) was analyzed using the Kaplan-Meier method. RESULTS: Of 55 patients, 23 harbored genomic defects in HR pathway genes. Median prostate specific antigen (PSA)-PFS for the HR defect group was 6.7 months compared with 2.6 months for the no HR defect group (p=0.001). The patients harboring somatic HR defect displayed shorter PSA-PFS than those harboring germline HR defect (4.5 months vs not available; p=0.066). The PSA50 (patients who survived for 12 weeks and had a PSA decline over 50% from baseline) response rate displayed higher in patients harboring BRCA2 or ATM defect (6/8, 75.0%) than in those harboring CDK12 defect (2/9, 22.2%; p=0.06). Patients harboring BRCA2 or ATM defect displayed prolonged PSA-PFS, compared with those harboring CDK12 defect and those harboring other HR defect (p=0.038). In multivariate Cox regression analysis, HR defect and BRCA2 or ATM defect were independent significant factors associated with superior PAS-PFS to platinum-based chemotherapy. CONCLUSIONS: The patients with mCRPC harboring alterations in different HR genes displayed distinct response to platinum-based chemotherapy. Patients with mCRPC harboring genomic defects in crucial HR genes either in the germline or somatic, especially BRCA2 and ATM, might experience superior outcomes to platinum-based chemotherapy, compared with those harboring CDK12 defect.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Biomarcadores de Tumor/genética , Carboplatino/uso terapéutico , Cisplatino/uso terapéutico , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteína BRCA2/genética , Biopsia , Carboplatino/farmacología , Cisplatino/farmacología , Quinasas Ciclina-Dependientes/genética , Análisis Mutacional de ADN , Docetaxel/farmacología , Docetaxel/uso terapéutico , Resistencia a Antineoplásicos/genética , Recombinación Homóloga/genética , Humanos , Calicreínas/sangre , Masculino , Persona de Mediana Edad , Mutación , Supervivencia sin Progresión , Próstata/patología , Antígeno Prostático Específico/sangre , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/mortalidad , Neoplasias de la Próstata Resistentes a la Castración/secundario , Estudios Retrospectivos
3.
J BUON ; 23(4): 1097-1102, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30358217

RESUMEN

PURPOSE: To detect the expression of VEGF and EGFR in peripheral blood and cancer tissues of patients with renal cell carcinoma (RCC), and to explore the correlations with clinical stage, pathological grade and prognosis of disease. METHODS: A total of 64 patients with RCC who were diagnosed and treated from June 2016 to August 2017 in our hospital were enrolled. Patients were divided into different clinical stages and pathological grades, and ELISA and immunohistochemistry were used to detect the expression of VEGF and EGFR in peripheral blood. Peripheral blood was also taken from 24 healthy individuals to serve as control group. Real-time fluorescence quantitative PCR (qRT-PCR) was used to detect the expression of VEGF and EGFR in RCC tissues and paracancer tissues. All patients were followed up after discharge to record their survival. RESULTS: Significant differences in the expression levels of VEGF and EGFR were found between stage III and IV (p<0.05), but not between stage I and II. Expressions level of VEGF and EGFR in serum of well-differentiated, moderatelydifferentiated, and poorly-differentiated RCC were all higher than those in the healthy control group, and significant differences were found between different pathological grades (p<0.05). Patients with higher expression levels of VEGF and EGFR showed shorter survival compared to patients with lower expression levels (p<0.05). CONCLUSION: VEGF and EGFR in peripheral blood can be used as one of the effective indicators of prognosis of RCC. Our study provided reference for clinical treatment and prediction of prognosis of RCC.


Asunto(s)
Carcinoma de Células Renales/sangre , Carcinoma de Células Renales/patología , Neoplasias Renales/sangre , Neoplasias Renales/patología , Factor A de Crecimiento Endotelial Vascular/sangre , Adulto , Anciano , Carcinoma de Células Renales/mortalidad , Estudios de Casos y Controles , Receptores ErbB/biosíntesis , Receptores ErbB/sangre , Femenino , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Neoplasias Renales/mortalidad , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Estadificación de Neoplasias , Pronóstico , Factor A de Crecimiento Endotelial Vascular/biosíntesis
4.
Prostate ; 77(12): 1233-1241, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28752926

RESUMEN

OBJECTIVE: To determine if prognostic nutritional index (PNI) and its variation could predict initial response to treatment and prognosis in metastatic castration-resistant prostate cancer (mCRPC) patients treated with Abiraterone (AA). PATIENTS AND METHODS: One-hundred-twelve chemotherapy pretreated or chemotherapy-naive patients were scheduled for systemic treatment with AA. PNI levels were measured before and after one month of AA treatment. Univariate and multivariate logistic regression analyses were used to identify predictive factors of initial response to AA treatment. Univariable and Multivariable Cox regression analyses were performed to determine prognostic factors that were associated with PSA progression-free survival (PSA-PFS), radiographic PFS (rPFS) and overall survival (OS). The Harrell concordance index with variables only or combined PNI data were used to evaluate the prognostic accuracy. RESULTS: Eighty-one (72.3%) of 112 patients showed initial response to AA treatment, in which 15 experienced PSA flare during AA treatment. In multivariate logistic regression analyses, high baseline PNI level, PSA level decrease during the first month of AA treatment and PNI level elevation during the first month of AA treatment were significantly correlated with initial response to AA treatment. In multivariate Cox regression analysis, low PNI level remained significant predictors of OS, rPFS and PSA-PFS. The estimated c-index of the multivariate model for OS increased from 0.82 without PNI to 0.83 when PNI added. CONCLUSION: Independent of PSA level variation, PNI level elevation during the first month of AA treatment and high baseline PNI level were significantly correlated with initial response to AA treatment. In addition, low pretreatment PNI level is a negative independent prognosticator of survival outcomes in mCRPC treated with AA and also increases the accuracy of established prognostic model.


Asunto(s)
Androstenos/uso terapéutico , Antineoplásicos/uso terapéutico , Evaluación Nutricional , Neoplasias de la Próstata Resistentes a la Castración/diagnóstico , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Anciano , Estudios de Cohortes , Estudios de Seguimiento , Humanos , Masculino , Pronóstico , Estudios Retrospectivos , Resultado del Tratamiento
5.
Prostate ; 77(13): 1373-1380, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28804908

RESUMEN

BACKGROUND: To determine the influence of abiraterone Acetate (AA) on neuroendocrine differentiation (NED) in patients with chemotherapy-naive metastatic castration-resistant prostate cancer (mCRPC). METHODS: We conducted an analysis in 115 chemotherapy-naïve mCRPC patients who would be treated with chemotherapy. The serum levels of chromogranin A (CgA), neurone-specific enolase (NSE) were measured in 67 mCRPC patients without AA treatment and 48 patients after the failure of AA treatment, in which these markers were also measured in 34 patients before and after 6 months of AA treatment. Comparative t-test was used to evaluate the serial changes of serum NED markers during AA treatment and univariate and multivariate analyses were performed to test the influence of AA treatment on NED. RESULTS: Serum CgA were NSE were evaluated to be above the upper limit of normal (ULN) in 56 (48.7%) and 29 (25.2%) patients before chemotherapy. In 34 patients with serial evaluation, serum CgA level of 14 patients and NSE of 14 patients increased after the failure of AA treatment. There was no significant difference of NED markers (CgA or NSE variation (P = 0.243) between at baseline and after the failure of AA treatment. Compared with the CgA elevation group in the first 6 months of AA treatment and baseline supranormal CgA group, the CgA decline group, and baseline normal CgA group has a much longer median PSA PFS (14.34 vs 10.00 months, P < 0.001, and 14.23 vs 10.30 months, P = 0.02) and rPFS, respectively (18.33 vs 11.37 months, P < 0.001, and 17.10 vs 12.07 months, P = 0.03). In logistic univariate analysis, AA treatment and its duration were not independent factors influencing NED. CONCLUSIONS: We hypothesized that AA might not significantly lead to progression of NED of mCRPC in general. Furthermore, we found there was heterogeneity in changes of NED markers in different mCRPC patients during AA treatment. Serial CgA and NSE evaluation might help clinicians guide clinical treatment of mCRPC patients.


Asunto(s)
Acetato de Abiraterona , Adenocarcinoma , Biomarcadores , Cromogranina A , Neurosecreción , Próstata , Neoplasias de la Próstata Resistentes a la Castración , Acetato de Abiraterona/administración & dosificación , Acetato de Abiraterona/farmacocinética , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Anciano , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Biomarcadores/sangre , Biomarcadores/metabolismo , China , Cromogranina A/sangre , Cromogranina A/metabolismo , Monitoreo de Drogas/métodos , Humanos , Masculino , Persona de Mediana Edad , Neurosecreción/efectos de los fármacos , Neurosecreción/fisiología , Próstata/metabolismo , Próstata/patología , Antígeno Prostático Específico/sangre , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/patología , Estudios Retrospectivos
6.
BJU Int ; 120(2): 226-232, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28107595

RESUMEN

OBJECTIVE: To determine the prognostic utility of serum chromogranin A (CgA) and neurone-specific enolase (NSE) variations during the first 3 months of abiraterone acetate (AA) treatment in patients with metastatic castration-resistant prostate cancer (mCRPC). PATIENTS AND METHODS: The serum levels of CgA, NSE were measured at baseline and after 3 months of AA treatment in 40 patients with mCRPC. Outcome measures were prostate-specific antigen progression-free survival (PSA-PFS), radiographic PFS (rPFS), and overall survival (OS). RESULTS: CgA levels were not correlated with NSE levels (P = 0.296). In multivariate analysis the combination of CgA and NSE (≥1 marker positive vs both markers negative) and the combination of CgA and NSE elevation during the first 3 months of AA treatment (≥1 marker positive vs both markers negative) remained significant predictors of OS, rPFS, and PSA-PFS. CONCLUSION: We found that CgA and NSE elevation during the first 3 months of AA treatment and elevated baseline CgA and NSE levels were independent prognostic factors for OS, rPFS and PSA-PFS in patients with mCRPC treated with AA. This suggests that serial CgA and NSE evaluation may help clinicians in distinguishing patients with mCRPC who would obtain the best survival benefit from AA treatment.


Asunto(s)
Acetato de Abiraterona/uso terapéutico , Antineoplásicos/uso terapéutico , Biomarcadores de Tumor/sangre , Cromogranina A/sangre , Fosfopiruvato Hidratasa/sangre , Neoplasias de la Próstata Resistentes a la Castración/sangre , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Anciano , Humanos , Masculino , Metástasis de la Neoplasia , Pronóstico , Antígeno Prostático Específico/sangre , Neoplasias de la Próstata Resistentes a la Castración/mortalidad , Neoplasias de la Próstata Resistentes a la Castración/patología , Estudios Retrospectivos
7.
Cell Tissue Res ; 366(3): 623-637, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27655233

RESUMEN

Diabetes mellitus (DM) is an important risk factor for erectile dysfunction. Adipose-derived stem cells (ADSCs) are essential for maintaining erectile function but their function is impaired during hyperglycemia. To evaluate the effects of pigment epithelium-derived factor (PEDF)-transfected ADSCs on the restoration of erectile function ADSCs and PEDF-transfected ADSCs were exposed to normal or high glucose levels for 72 h and the effects on cell survival and protein expression were determined. For in vivo studies, rats with streptozocin-induced DM were intravenously injected with ADSCs or PEDF-transfected ADSCs. Two weeks later, the intracavernosal pressure (ICP) and mean arterial pressure (MAP) were measured to assess erectile function, and penile tissues were harvested for further evaluation. PEDF overexpression in ADSCs protected cells against hyperglycemia-induced apoptosis (as determined by a TUNEL assay), increasing the expression of neurotrophic factors and decreasing the expression of inflammatory cytokines and oxidative stress-related proteins (as determined by western blotting and ELISA). In DM rats, transplantation with PEDF-transfected ADSCs effectively restored erectile function, as determined by the ICP/MAP ratio, compared with untreated ADSCs. PEDF overexpression also resulted in higher survival rates and decreased apoptosis of ADSCs. Promotion of neurotrophic factor expression and suppression of inflammatory cytokines and oxidative stress-related proteins were also observed after transplantation of PEDF-transfected ADSCs. Thus, our results demonstrate that transplantation of ADSCs restored erectile function in a rat model of DM, attenuating the negative effects of hyperglycemia. These findings indicate the therapeutic potential of ADSCs for treating erectile dysfunction and the additional benefits of PEDF overexpression.


Asunto(s)
Tejido Adiposo/citología , Diabetes Mellitus Experimental/complicaciones , Disfunción Eréctil/complicaciones , Disfunción Eréctil/terapia , Proteínas del Ojo/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Serpinas/metabolismo , Trasplante de Células Madre , Células Madre/citología , Transfección , Animales , Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Citocinas/metabolismo , Diabetes Mellitus Experimental/patología , Modelos Animales de Enfermedad , Disfunción Eréctil/patología , Proteínas del Ojo/farmacología , Glucosa/toxicidad , Mediadores de Inflamación/metabolismo , Masculino , Factores de Crecimiento Nervioso/farmacología , Óxido Nítrico Sintasa de Tipo I/metabolismo , Estrés Oxidativo/efectos de los fármacos , Pene/inervación , Pene/patología , Ratas Sprague-Dawley , Serpinas/farmacología , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Estreptozocina
8.
BMC Urol ; 16(1): 24, 2016 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-27267922

RESUMEN

BACKGROUND: Intestinal metaplasia of the bladder is an uncommon glandular proliferation. We examined a large series of intestinal metaplasia for the clinicopathological features and discuss the significance of this lesion. METHODS: All cases of intestinal metaplasia diagnosed in our institution between 1990 and 2014 were retrospectively reviewed. Patients with a history of urothelial carcinoma or concurrent adenocarcinoma were excluded. Patient characteristics, pathological features, and follow-up outcomes were obtained. RESULTS: We identified 89 patients with intestinal metaplasia during this period. Sixty seven were men and 22 were women. Mean age at diagnosis was 57 years (range 23-81). Common presenting complaints included haematuria (73 cases), mucosuria (13 cases), and irritative voiding symptoms (seven cases). The majority of intestinal metaplasias located on or near the trigone (67 cases). Eighty-two patients underwent transurethral resection of their lesions. Partial cystectomy was performed in the remaining seven patients. The mean follow-up of 78 patients was 105 months (range 6-255). One case of bladder adenocarcinoma was indentified 6 months later. The initial histologic findings had revealed intestinal metaplasia with severe dysplasia. Four patients presented recurrence during the follow-up, and this occurred 9, 13, 17 and 24 months after the surgery. CONCLUSIONS: Although intestinal metaplasia can be treated effectively by transurethral resection in most cases, its potential malignancy need to be taken into consideration after the evidence of recurrences and its association with bladder adenocarcinoma. Therefore, it is necessary to perform close surveillance following the surgery, particularly in patients with dysplastic changes.


Asunto(s)
Lesiones Precancerosas/patología , Lesiones Precancerosas/cirugía , Neoplasias de la Vejiga Urinaria/patología , Neoplasias de la Vejiga Urinaria/cirugía , Vejiga Urinaria/patología , Vejiga Urinaria/cirugía , Adulto , Distribución por Edad , Anciano , Anciano de 80 o más Años , China/epidemiología , Femenino , Humanos , Estudios Longitudinales , Masculino , Metaplasia/patología , Persona de Mediana Edad , Lesiones Precancerosas/epidemiología , Prevalencia , Estudios Retrospectivos , Factores de Riesgo , Distribución por Sexo , Resultado del Tratamiento , Neoplasias de la Vejiga Urinaria/epidemiología
9.
Nat Commun ; 14(1): 7794, 2023 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-38016952

RESUMEN

Neuroendocrine prostate cancer is a rapidly progressive and lethal disease characterized by early visceral metastasis, poor prognosis, and limited treatment options. Uncovering the oncogenic mechanisms could lead to the discovery of potential therapeutic avenues. Here, we demonstrate that the RNA-binding protein ELAVL3 is specifically upregulated in neuroendocrine prostate cancer and that overexpression of ELAVL3 alone is sufficient to induce the neuroendocrine phenotype in prostate adenocarcinoma. Mechanistically, ELAVL3 is transcriptionally regulated by MYCN and subsequently binds to and stabilizes MYCN and RICTOR mRNA. Moreover, ELAVL3 is shown to be released in extracellular vesicles and induce neuroendocrine differentiation of adenocarcinoma cells via an intercellular mechanism. Pharmacological inhibition of ELAVL3 with pyrvinium pamoate, an FDA-approved drug, effectively suppresses tumor growth, reduces metastatic risk, and improves survival in neuroendocrine prostate cancer mouse models. Our results identify ELAVL3 as a critical regulator of neuroendocrine differentiation in prostate cancer and propose a drug repurposing strategy for targeted therapies.


Asunto(s)
Adenocarcinoma , Neoplasias de la Próstata , Humanos , Masculino , Animales , Ratones , Proteína Proto-Oncogénica N-Myc/genética , Retroalimentación , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Fenotipo , Adenocarcinoma/genética , Línea Celular Tumoral , Proteína 3 Similar a ELAV/genética
10.
J Clin Invest ; 133(24)2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-38099497

RESUMEN

Cell lineage plasticity is one of the major causes for the failure of targeted therapies in various cancers. However, the driver and actionable drug targets in promoting cancer cell lineage plasticity are scarcely identified. Here, we found that a G protein-coupled receptor, ADORA2A, is specifically upregulated during neuroendocrine differentiation, a common form of lineage plasticity in prostate cancer and lung cancer following targeted therapies. Activation of the ADORA2A signaling rewires the proline metabolism via an ERK/MYC/PYCR cascade. Increased proline synthesis promotes deacetylases SIRT6/7-mediated deacetylation of histone H3 at lysine 27 (H3K27), and thereby biases a global transcriptional output toward a neuroendocrine lineage profile. Ablation of Adora2a in genetically engineered mouse models inhibits the development and progression of neuroendocrine prostate and lung cancers, and, intriguingly, prevents the adenocarcinoma-to-neuroendocrine phenotypic transition. Importantly, pharmacological blockade of ADORA2A profoundly represses neuroendocrine prostate and lung cancer growth in vivo. Therefore, we believe that ADORA2A can be used as a promising therapeutic target to govern the epigenetic reprogramming in neuroendocrine malignancies.


Asunto(s)
Neoplasias Pulmonares , Neoplasias de la Próstata , Sirtuinas , Animales , Humanos , Masculino , Ratones , Línea Celular Tumoral , Epigénesis Genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Prolina/metabolismo , Prolina/uso terapéutico , Próstata/metabolismo , Próstata/patología , Neoplasias de la Próstata/patología , Sirtuinas/metabolismo
11.
Comput Math Methods Med ; 2022: 4182401, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35770119

RESUMEN

Prostate cancer (PCa) is the most frequent cancer in men. Developing new treatment methods for CRPC will be a significant challenge in the clinical treatment of PCa. In conclusion, the results of this study show that NRF2 is downregulated in untreated PCa samples compared to normal PCa samples; however, it was upregulated in mCRPC samples compared to HSPC samples. These results demonstrated that NRF2 may serve as a tumor suppressor in tumorigenesis but promote PCa androgen-independent transferring after ADT treatment. Bioinformatics analysis showed that NRF2 was related to multiple signaling, such as the AGE-RAGE pathway, MAPK pathway, NF-kappa B signaling, PI3K-Akt signaling pathway, and VEGF signaling pathway. Moreover, we revealed that the NRF2 inhibitor significantly inhibited tumorigenicity of CRPC cells in vitro. Of note, combination of the NRF2 inhibitor and autophagy inhibitor had a more significantly suppressive role than either ML385 or CQ, indicating that combination of CQ (autophagy inhibitor) and ML385 (NRF2 inhibitor) is a potential treatment of CRPC. Finally, we conformed that high levels of autophagy regulators LC3B, ULK1, and beclin1 significantly correlated to longer PSA recurrence-free survival time. We think that this study could provide more evidence to confirm that NRF2 is a crucial regulator and targeting NRF2 and autophagy is a potential therapy option for CRPC.


Asunto(s)
Neoplasias de la Próstata Resistentes a la Castración , Autofagia , Línea Celular Tumoral , Humanos , Masculino , Factor 2 Relacionado con NF-E2/genética , Fosfatidilinositol 3-Quinasas , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/metabolismo
12.
Theranostics ; 12(11): 4965-4979, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35836810

RESUMEN

Background: Prostate cancer is usually considered as immune "cold" tumor with poor immunogenic response and low density of tumor-infiltrating immune cells, highlighting the need to explore clinically actionable strategies to sensitize prostate cancer to immunotherapy. In this study, we investigated whether docetaxel-based chemohormonal therapy induces immunologic changes and potentiates checkpoint blockade immunotherapy in prostate cancer. Methods: We performed transcriptome and histopathology analysis to characterize the changes of prostate cancer immune microenvironment before and after docetaxel-based chemohormonal therapy. Furthermore, we investigated the therapeutic benefits and underlying mechanisms of chemohormonal therapy combined with anti-PD1 blockade using cellular experiments and xenograft prostate cancer models. Finally, we performed a retrospective cohort analysis to evaluate the antitumor efficacy of anti-PD1 blockade alone or in combination with docetaxel-based chemotherapy. Results: Histopathology assessments on patient samples confirmed the enrichment of tumor-infiltrating T cells after chemohormonal therapy. Moreover, we found that docetaxel activated the cGAS/STING pathway in prostate cancer, subsequently induced IFN signaling, resulting in lymphocytes infiltration. In a xenograft mouse model, docetaxel-based chemohormonal therapy prompted the intratumoral infiltration of T cells and upregulated the abundance of PD1 and PD-L1, thereby sensitizing mouse tumors to the anti-PD1 blockade. To determine the clinical significance of these results, we retrospectively analyzed a cohort of 30 metastatic castration-resistant prostate cancer patients and found that docetaxel combined with anti-PD1 blockade resulted in better prostate-specific antigen progression-free survival when compared with anti-PD1 blockade alone. Conclusions: Our study demonstrates that docetaxel activates the antitumoral immune response and facilitates T cell infiltration in a cGAS/STING-dependent manner, providing a combination immunotherapy strategy that would improve the clinical benefits of immunotherapy.


Asunto(s)
Inmunoterapia , Neoplasias de la Próstata , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Docetaxel/uso terapéutico , Humanos , Factores Inmunológicos , Inmunoterapia/métodos , Masculino , Ratones , Nucleotidiltransferasas , Neoplasias de la Próstata/tratamiento farmacológico , Estudios Retrospectivos , Microambiente Tumoral
13.
Front Oncol ; 12: 955166, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36033483

RESUMEN

The androgen receptor (AR) signaling is a key contributor to tumorigenesis and the progression of prostate cancer. A subset of patients may develop neuroendocrine (NE) features, resulting in resistance to androgen deprivation therapy and poor prognosis. In this study, we combined immunostaining and bulk and single-cell transcriptome analyses to better characterize the status of AR in prostate cancer with neuroendocrine differentiation. The exploration of online datasets indicated the existence of ARHIGH/NEHIGH prostate cancer and revealed that these double-high cases are majorly present in castration-resistant prostate cancer with a less neuroendocrine-transdifferentiated state. We then reviewed 8,194 prostate cancer cases with available immunohistochemistry reports and found 2.3% cases (n = 189) that showed at least one of the NE markers (chromogranin A, synaptophysin, and neural cell adhesion molecule 1) being positive in at least 5% of epithelial cells. Within these 189 cases, we observed that 81.0% cases (n = 153) showed AR positive and 19.0% (n = 36) showed AR negative. Patients with AR loss tumors demonstrated a correlation with adverse clinical stages, indicating a trade-off between AR and advanced disease in neuroendocrine differentiation. Using multiplex immunofluorescence staining, we observed the co-localization of AR and NE markers in prostate cancer cells. In addition, data mining of single-cell transcriptome further confirmed the existence of ARHIGH/NEHIGH prostate cancer cells in castration-resistant samples and suggested that AR still exerts its androgen response and anti-apoptotic effect in these double-high cells. Thus, our study provides a better understanding of AR signaling in the cellular plasticity of prostate cancer with neuroendocrine differentiation and allows new insights into the therapeutic development.

14.
Nat Cancer ; 3(5): 565-580, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35624341

RESUMEN

Among the greatest hurdles in clinical management of prostate cancer (PCa) are the progression to lethal castration-resistant prostate cancer (CRPC) and the lack of suitable targeted therapies for advanced disease. Here we identify Gremlin1 as a ligand for fibroblast growth factor receptor 1 (FGFR1), which promotes lineage plasticity and drives castration resistance. Importantly, we generate a specific anti-Gremlin1 therapeutic antibody and demonstrate synergistic effect with androgen deprivation therapy (ADT) in CRPC. GREM1 transcription is suppressed by androgen receptor (AR) and released following ADT. We show that Gremlin1 binds to FGFR1 and activates downstream MAPK signaling. Gremlin1 interacts with FGFR1 differently to its canonical ligand FGF1, as revealed through protein structure docking and mutagenesis experiments. Altogether, our data indicate Gremlin1 as a promising candidate therapeutic target for CRPC.


Asunto(s)
Neoplasias de la Próstata Resistentes a la Castración , Antagonistas de Andrógenos/farmacología , Castración , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Ligandos , Masculino , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Transducción de Señal
15.
Clin Genitourin Cancer ; 19(4): e223-e234, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33824096

RESUMEN

BACKGROUND: The study aimed to investigate the efficacy and safety outcomes in hormone-sensitive oligometastatic prostate cancer (OMPC) patients treated with docetaxel-based neoadjuvant chemohormonal therapy (NCHT) prior to radical prostatectomy (RP) compared with direct RP and standard androgen deprivation therapy (ADT) alone using propensity score match (PSM) analysis. PATIENTS AND METHODS: A single-center, prospective, three-arm study was conducted with hormone-sensitive OMPC patients. Eligible patients (N = 130) were divided into three groups-NCHT, RP, and standard treatment (ST)-and received their respective treatments. The primary endpoint was progression-free survival (PFS), and the secondary endpoints were pathological response rate, radiographic progression-free survival (rPFS), and overall survival (OS). Further, propensity scores were calculated and group-wise comparisons were carried out: NCHT versus RP, ST versus RP, and ST versus NCHT. RESULTS: After PSM, in the NCHT group, two patients (11.76%) and four patients (23.52%) had complete and partial pathological responses, respectively. Univariate and multivariate Cox regression analysis showed that PFS and rPFS were significantly higher in the NCHT group. For NCHT versus RP, the PFS hazard ratio (HR) = 0.11 (95% confidence interval [CI], 0.02-0.51; P = .004) and HR = 0.016 (95% CI, 0.0015-0.17; P < .001); the rPFS HR = 0.088 (95% CI, 0.011-0.71; P = .023) and HR = 0.03 (95% CI, 0.0025-0.36; P = .006). Further, the median OS of the ST group was 44.6 months for ST versus RP, and it was 49.3 months for ST versus NCHT; it was not reached in either the NCHT or RP group. Furthermore, 17.65% and 47.06% patients had positive surgical margins in the NCHT and RP groups, respectively, and no therapy-related deaths were observed during the study period. CONCLUSIONS: PSM analysis revealed NCHT before RP in OMPC patients has potential therapeutic benefits with acceptable toxicities and lower incidence of postoperative positive surgical margins.


Asunto(s)
Terapia Neoadyuvante , Neoplasias de la Próstata , Antagonistas de Andrógenos/uso terapéutico , Hormonas , Humanos , Masculino , Puntaje de Propensión , Estudios Prospectivos , Prostatectomía , Neoplasias de la Próstata/tratamiento farmacológico
16.
Cancer Med ; 10(15): 5358-5374, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34128342

RESUMEN

BACKGROUND: Tumor-infiltrating immune cells participate in the initiation and progression of prostate adenocarcinoma (PRAD). However, it is not fully known how immune infiltration affects the development of PRAD and its clinical presentation. METHODS: Herein, we investigated the immune infiltration phenotypes in PRAD based on transcriptome profiles, methylation profiles, somatic mutation, and copy number variations. We also developed an immune prognostic model (IPM) to identify unfavorable prognosis. To verify this model, immunohistochemistry staining was performed on a cohort of PRAD samples. Moreover, we constructed a nomogram to assess the survival of PRAD incorporating immune infiltration and other clinical features. RESULTS: We categorized PRAD patients into high and low-level clusters based on immune infiltration phenotypes. The patients in the high-level clusters had worse survival than their low-level counterparts. Gene set enrichment analysis indicated that both anti- and pro-tumor terms were enriched in high-level cluster. Moreover, we identified a positive correlation between anti- and pro-tumor immune cells in PRAD microenvironment. Notably, Somatic mutation analysis showed patients in high-level cluster had a higher somatic mutation burden of KMT2D, HSPA8, CHD7, and MAP1A. In addition, we developed an IPM with robust predictive ability. The model can distinguish high-risk PRAD patients with poor prognosis from low-risk PRAD patients in both training and another three independent validation datasets. Besides, we constructed a nomogram incorporating Gleason score, pathological T stage, and IPM for the prognosis prediction of PRAD patients, which displayed robust predictive ability and might contribute to clinical practice. CONCLUSION: Our work illustrated the immune infiltration phenotypes strongly related to the poor prognosis of PRAD patients, and highlighted the potential of the IPM to identify unfavorable tumor features.


Asunto(s)
Adenocarcinoma/inmunología , Linfocitos Infiltrantes de Tumor/citología , Neoplasias de la Próstata/inmunología , Adenocarcinoma/genética , Adenocarcinoma/mortalidad , Adenocarcinoma/patología , Estudios de Cohortes , Variaciones en el Número de Copia de ADN , ADN Helicasas/genética , Metilación de ADN , Proteínas de Unión al ADN/genética , Conjuntos de Datos como Asunto , Progresión de la Enfermedad , Perfilación de la Expresión Génica , Proteínas del Choque Térmico HSC70/genética , Humanos , Inmunofenotipificación , Masculino , Metilación , Proteínas Asociadas a Microtúbulos/genética , Persona de Mediana Edad , Mutación , Tasa de Mutación , Clasificación del Tumor , Proteínas de Neoplasias/genética , Nomogramas , Pronóstico , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/mortalidad , Neoplasias de la Próstata/patología , Transcriptoma , Microambiente Tumoral/inmunología
17.
Int J Nanomedicine ; 16: 5193-5209, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34354353

RESUMEN

BACKGROUND: Recently, nanocatalyst-induced endoplasmic reticulum (ER) stress for cancer therapy has been attracting considerable attention. However, cancer cells are often able to overcome ER stress-induced death by activating the unfolded protein response (UPR), making nanocatalytic monotherapy a poor defense against cancer progression. PURPOSE: In this study, to improve the nanocatalytic treatment efficacy, a phase change material (PCM) was used to encapsulate the upstream ER stress initiator, iron oxide nanoparticles (Fe3O4 NPs), and the downstream UPR modulator, PR-619. Subsequently, the tumor-homing peptide tLyP-1 was coupled to it to form tLyP-1/PR-619/Fe3O4@PCM (tPF@PCM) theranostic platform. MATERIALS AND METHODS: tPF@PCM was synthesized using nanoprecipitation and resolidification methods followed by the EDC/NHS cross-linking method. The targeting capacity of tPF@PCM was evaluated in vitro and in vivo using flow cytometry and magnetic resonance imaging, respectively. The therapeutic efficacy of tPF@PCM was investigated in a renal cell carcinoma mouse model. Moreover, we explored the synergistic anti-tumor mechanism by examining the intracellular reactive oxygen species (ROS), aggregated proteins, ER stress response levels, and type of cell death. RESULTS: tPF@PCM had excellent tumor-targeting properties and exhibited satisfactory photothermal-enhanced tumor inhibition efficacy both in vitro and in vivo. Specifically, the phase transition temperature (45 °C) maintained using 808 nm laser irradiation significantly increased the release and catalytic activity of the peroxidase mimic Fe3O4 NPs. This strongly catalyzed the generation of hydroxyl radicals (•OH) via the Fenton reaction in the acidic tumor microenvironment. The redox imbalance subsequently resulted in an increase in the level of damaged proteins in the ER and initiated ER stress. Moreover, the pan-deubiquitinase inhibitor PR-619 blocked the "adaptive" UPR-mediated degradation of these damaged proteins, exacerbating the ER burden. Consequently, irremediable ER stress activated the "terminal" UPR, leading to apoptosis in cancer cells. CONCLUSION: This ER stress-exacerbating strategy effectively suppresses tumorigenesis, offering novel directions for advances in the treatment of conventional therapy-resistant cancers.


Asunto(s)
Retículo Endoplásmico , Neoplasias , Animales , Apoptosis , Retículo Endoplásmico/metabolismo , Estrés del Retículo Endoplásmico , Humanos , Ratones , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo , Respuesta de Proteína Desplegada
18.
Cell Prolif ; 54(7): e13056, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34021647

RESUMEN

OBJECTIVES: In contrast to extensive studies on bone metastasis in advanced prostate cancer (PCa), liver metastasis has been under-researched so far. In order to decipher molecular and cellular mechanisms underpinning liver metastasis of advanced PCa, we develop a rapid and immune sufficient mouse model for liver metastasis of PCa via orthotopic injection of organoids from PbCre+ ; rb1f/f ;p53f/f mice. MATERIALS AND METHODS: PbCre+ ;rb1f/f ;p53f/f and PbCre+ ;ptenf/f ;p53f/f mice were used to generate PCa organoid cultures in vitro. Immune sufficient liver metastasis models were established via orthotopic transplantation of organoids into the prostate of C57BL/6 mice. Immunofluorescent and immunohistochemical staining were performed to characterize the lineage profile in primary tumour and organoid-derived tumour (ODT). The growth of niche-labelling reporter infected ODT can be visualized by bioluminescent imaging system. Immune cells that communicated with tumour cells in the liver metastatic niche were determined by flow cytometry. RESULTS: A PCa liver metastasis model with full penetrance is established in immune-intact mouse. This model reconstitutes the histological and lineage features of original tumours and reveals dynamic tumour-immune cell communication in liver metastatic foci. Our results suggest that a lack of CD8+ T cell and an enrichment of CD163+ M2-like macrophage as well as PD1+ CD4+ T cell contribute to an immuno-suppressive microenvironment of PCa liver metastasis. CONCLUSIONS: Our model can be served as a reliable tool for analysis of the molecular pathogenesis and tumour-immune cell crosstalk in liver metastasis of PCa, and might be used as a valuable in vivo model for therapy development.


Asunto(s)
Neoplasias Hepáticas/patología , Neoplasias de la Próstata/patología , Animales , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Comunicación Celular , Modelos Animales de Enfermedad , Neoplasias Hepáticas/secundario , Macrófagos/citología , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfohidrolasa PTEN/deficiencia , Fosfohidrolasa PTEN/genética , Neoplasias de la Próstata/metabolismo , Microambiente Tumoral , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/genética
19.
Nat Commun ; 12(1): 1812, 2021 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-33753739

RESUMEN

Human hexokinase 2 is an essential regulator of glycolysis that couples metabolic and proliferative activities in cancer cells. The binding of hexokinase 2 to the outer membrane of mitochondria is critical for its oncogenic activity. However, the regulation of hexokinase 2 binding to mitochondria remains unclear. Here, we report that SUMOylation regulates the binding of hexokinase 2 to mitochondria. We find that hexokinase 2 can be SUMOylated at K315 and K492. SUMO-specific protease SENP1 mediates the de-SUMOylation of hexokinase 2. SUMO-defective hexokinase 2 preferably binds to mitochondria and enhances both glucose consumption and lactate production and decreases mitochondrial respiration in parallel. This metabolic reprogramming supports prostate cancer cell proliferation and protects cells from chemotherapy-induced cell apoptosis. Moreover, we demonstrate an inverse relationship between SENP1-hexokinase 2 axis and chemotherapy response in prostate cancer samples. Our data provide evidence for a previously uncovered posttranslational modification of hexokinase 2 in cancer cells, suggesting a potentially actionable strategy for preventing chemotherapy resistance in prostate cancer.


Asunto(s)
Carcinogénesis/metabolismo , Hexoquinasa/metabolismo , Mitocondrias/metabolismo , Neoplasias de la Próstata/metabolismo , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Carcinogénesis/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cisteína Endopeptidasas/genética , Cisteína Endopeptidasas/metabolismo , Docetaxel/farmacología , Hexoquinasa/genética , Humanos , Masculino , Ratones , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , Unión Proteica , Sumoilación , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
20.
J Cancer Res Clin Oncol ; 146(3): 687-694, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31745702

RESUMEN

OBJECTIVE: To assess the use of the cell cycle progression (CCP) score versus actual risk stratification practice in making treatment decisions for prostate cancer patients with locally adverse pathology after radical prostatectomy (RP). PATIENTS AND METHODS: Men with adverse pathologic features, pT3 or positive surgical margins who underwent RP in 2010-2014 at Renji hospital were retrospectively analyzed. The primary outcome was biochemical recurrence (BCR) after RP. RNA was quantified from paraffin-embedded RP specimens. The CCP score was calculated as average expression of 31 CCP genes, normalized to 15 housekeeper genes. The prognostic utility of the CCP score was assessed using Kaplan-Meier analysis and multivariable Cox proportional hazards model. RESULTS: Among the 100 men identified, 5-year BCR-free survival for the low- (< 0), intermediate- (0-1) and high- (> 1) CCP score groups was 89.3%, 38.8%, and 12.9%, respectively. In multivariable models adjusting for clinical and pathological variables with the cancer of the prostate risk assessment post-surgical (CAPRA-S) score, both continuous CCP score [hazard ratio (HR) 1.373 per unit score, 95% confidence interval (CI) 1.006-1.874; p = 0.046) and the categorized CCP score (p < 0.001)were independent predictors of BCR. CONCLUSIONS: The present study provides insights into the role the CCP score plays in risk stratification of this cohort and in determining candidacy for deferred secondary treatment. From our perspective, the CCP score allows better stratification and can help identifying patients at lower risk of disease recurrence who could benefit from a wait-and-see policy.


Asunto(s)
Ciclo Celular/fisiología , Neoplasias de la Próstata/patología , Anciano , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/patología , Pronóstico , Prostatectomía , Neoplasias de la Próstata/cirugía , Estudios Retrospectivos , Medición de Riesgo/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA