Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Drug Resist Updat ; 70: 100990, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37478518

RESUMEN

This study investigated cellular mechanisms in steroidogenesis responsible for treatment resistance to the novel antiandrogen agent darolutamide in prostate cancer. HSD3B1 was overexpressed in darolutamide-resistant cells and induced by darolutamide treatment and AR knockdown. Inversely, HSD3B1 knockdown increased cellular sensitivity to darolutamide. Similarly, its upstream regulator NR5A2 was up-regulated in darolutamide-resistant cells and induced by darolutamide treatment and AR knockdown. Inversely, NR5A2 knockdown and NR5A2 inhibitor ML180 decreased expression of various steroidogenic enzymes including HSD3B1, leading to increased cellular sensitivity to darolutamide. The NR5A2/HSD3B1 pathway promoted cellular resistance to darolutamide and targeting NR5A2/HSD3B1 pathway is a promising therapeutic strategy to overcome darolutamide resistance.


Asunto(s)
Antagonistas de Andrógenos , Neoplasias de la Próstata Resistentes a la Castración , Humanos , Masculino , Antagonistas de Andrógenos/farmacología , Antagonistas de Andrógenos/uso terapéutico , Antagonistas de Receptores Androgénicos/farmacología , Antagonistas de Receptores Androgénicos/uso terapéutico , Complejos Multienzimáticos/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo
2.
Int J Urol ; 30(4): 340-346, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36478611

RESUMEN

OBJECTIVES: To investigate the impact of extended pelvic lymph node dissection (ePLND) on urinary incontinence (UI) at early post-surgery robot-assisted radical prostatectomy (RARP). METHODS: Patients who underwent RARP without cavernous nerve sparing were included between 2014 and 2019. Patient data were obtained prospectively. The associations between ePLND and postoperative urinary continence were defined as a maximum of one daily pad use. International prostate symptom score (IPSS) was examined. Expression of synaptophysin and tyrosine hydroxylase (TH) in perilymph node adipose tissue (PLA) was evaluated by immunohistochemistry. RESULTS: In total, 186 and 163 patients underwent RARP with and without ePLND. Urinary continence rate at 1 month postoperatively among patients with ePLND was lower than those without ePLND (24.1% vs. 35.1%, p < 0.05), however, not significantly different at 3, 6, and 12 months after RARP (57.4 vs. 62.6%, 73.1 vs. 74.2%, and 83.0 vs. 81.2%, respectively). Total and voiding plus postvoiding IPSS scores at 1 month were higher in patients with ePLND than in those without ePLND (14.5 ± 0.5 vs. 13.6 ± 0.6, 7.0 ± 0.3 vs. 6.2 ± 0.4, respectively, p < 0.05). In univariate and multivariate analyses, larger prostate volume and ePLND were factors associated with an increased UI rate. Among patients who underwent ePLND, synaptophysin and TH-positive nerve fibers were detected in PLA. CONCLUSIONS: Detection of synaptophysin and TH-immunopositive nerves suggested denervation of sympathetic and peripheral nerves caused by ePLND might be associated with a higher UI rate and poor urinary symptoms at an early stage after RARP.


Asunto(s)
Neoplasias de la Próstata , Procedimientos Quirúrgicos Robotizados , Robótica , Incontinencia Urinaria , Masculino , Humanos , Próstata/cirugía , Próstata/patología , Sinaptofisina , Neoplasias de la Próstata/patología , Escisión del Ganglio Linfático/efectos adversos , Procedimientos Quirúrgicos Robotizados/efectos adversos , Prostatectomía/efectos adversos , Incontinencia Urinaria/epidemiología , Incontinencia Urinaria/etiología , Poliésteres
3.
Cancer Sci ; 113(5): 1731-1738, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35118769

RESUMEN

Ribosomal S6 kinase has been shown to play a key role in cellular resistance to endocrine therapy in prostate cancer through its regulation of YB-1/androgen receptor (AR) signaling. PMD-026, an oral first-in-class small molecule kinase inhibitor, is the first identified ribosomal S6 kinase inhibitor. This study investigated the effect of PMD-026 on YB-1/AR signaling and its antitumor effect in prostate cancer in vitro and in vivo. Castration-resistant prostate cancer 22Rv1 cells that express high-level AR variants were used in this study. The effect of PMD-026 on YB-1/AR signaling was investigated by quantitative real-time PCR and western blot analysis. The effects of PMD-026 on prostate cancer cells were investigated by cytotoxicity analysis, apoptosis assay, and cell cycle assay in vitro and a mouse castration model in vivo. PMD-026 decreased YB-1 phosphorylation as well as AR V7 mRNA and AR variant expressions in 22Rv1 cells. PMD-026 suppressed cell proliferation alone and in combination with the second-generation antiandrogens enzalutamide and darolutamide by inducing cellular apoptosis and G2/M arrest. In a mouse xenograft model, PMD-026 suppressed tumor growth, and the combination of PMD-026 and enzalutamide inhibited tumor growth more prominently than single treatments. Our results demonstrate an excellent antitumor effect of the novel ribosomal S6 kinase inhibitor PMD-026 and the combination effect with the antiandrogen enzalutamide in castration-resistant prostate cancer. These findings warrant a clinical trial of PMD-026 in prostate cancer patients.


Asunto(s)
Antineoplásicos , Neoplasias de la Próstata Resistentes a la Castración , Antagonistas de Andrógenos/uso terapéutico , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis , Línea Celular Tumoral , Resistencia a Antineoplásicos , Puntos de Control de la Fase G2 del Ciclo Celular , Humanos , Masculino , Ratones , Nitrilos/farmacología , 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 , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Proteínas Quinasas S6 Ribosómicas
4.
Andrologia ; 54(1): e14307, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34747051

RESUMEN

This study aimed to investigate the significance of HSD3B1 gene status including germline polymorphism and somatic alterations in prostate cancer. Patients with prostate cancer treated with androgen-deprivation therapy, as well as tissues from metastatic prostate cancer, were included. Genomic DNA was extracted from cancer tissues and whole blood samples, and HSD3B1 (rs1047303, 1245C) was genotyped by Sanger sequencing. The association of HSD3B1 genotype with progression-free survival according to metastatic volume was examined. Copy number alteration and gene expression of HSD3B1 were examined in prostate cancer cells and public datasets. Among 194 patients, 121 and 73 patients were categorized into low- and high-volume diseases respectively. In multivariate analysis, the adrenal-permissive genotype (AC/CC) was significantly associated with increased risk of progression compared with the adrenal-restrictive genotype (AA) in low volume, but not high-volume diseases. Somatic mutation in HSD3B1 was detected at least in two cases of castration-resistant prostate cancer tissues. HSD3B1 amplification and overexpression were detected in castration-resistant prostate cancer cells and tissues. The current findings suggest that both germline and somatic alterations of HSD3B1 may cooperatively promote castration resistance in prostate cancer and HSD3B1 as a promising biomarker for precision medicine, warranting further investigations.


Asunto(s)
Antagonistas de Andrógenos , Neoplasias de la Próstata Resistentes a la Castración , Genotipo , Humanos , Masculino , Complejos Multienzimáticos/genética , Polimorfismo Genético
5.
Cancer Sci ; 112(1): 323-330, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33064355

RESUMEN

Although Y-box binding protein-1 (YB-1) is known to be overexpressed in prostate cancer, especially castration-resistant prostate cancer (CRPC), the mechanism of its overexpression remains unclear. We aimed to elucidate the mechanism of YB-1 overexpression in CRPC. Gene amplification in CRPC cells and tissues was examined by public database analysis, and digital PCR. The significance of YB-1 amplification for the YB-1/androgen receptor (AR) axis and prognosis was examined by public database analysis and immunohistochemistry. YB-1 amplification was mainly observed in CRPC tissues by public database analysis and confirmed in CRPC cells and tissues by digital PCR. Expression of YB-1 was increased in CRPC tissues compared with treatment-naïve tissues. Furthermore, YB-1 and phosphorylated YB-1 levels were associated with AR and AR V7 expression levels. Finally, YB-1 amplification was associated with poor outcomes in CRPC. Taken together, the present findings suggest that YB-1 amplification contributes to progression to CRPC through regulation of AR and AR V7 expressions, and that YB-1 is a promising therapeutic target in CRPC.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/genética , Neoplasias de la Próstata Resistentes a la Castración/genética , Receptores Androgénicos/biosíntesis , Proteína 1 de Unión a la Caja Y/genética , Amplificación de Genes , Humanos , Masculino , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Proteína 1 de Unión a la Caja Y/metabolismo
6.
Prostate ; 80(11): 885-894, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32483877

RESUMEN

BACKGROUND: Eradication of persistent androgen receptor (AR) activity in castration-resistant prostate cancer may be a promising strategy to overcome castration resistance. We aimed to identify novel compounds that inhibit AR activity and could be potential therapeutic agents for prostate cancer. METHODS: A high-throughput screening system involving cell lines stably expressing AR protein and AR-responsive luciferase was employed for the 1260 compound library. Molecular and antitumor effects on candidate pathways that interacted with AR signaling were examined in prostate cancer cells expressing AR. RESULTS: The high-throughput screening identified various potential compounds that interfered with AR signaling through known and novel pathways. Among them, a 5-hydroxytryptamine 5A (5-HT5A) receptor antagonist suppressed AR activity through protein kinase A signaling, which was confirmed by 5-HT5A receptor knockdown. Consistently, 5-HT5A receptor inhibitors showed cytotoxic effects toward prostate cancer cells. CONCLUSIONS: Taken together, this study identifies 5-HT5A receptor as a promising therapeutic target for prostate cancer via its interaction with AR signaling.


Asunto(s)
Antagonistas de Receptores Androgénicos/farmacología , Andrógenos/farmacología , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata/tratamiento farmacológico , Línea Celular Tumoral , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Ensayos de Selección de Medicamentos Antitumorales , Ensayos Analíticos de Alto Rendimiento/métodos , Humanos , Masculino , Células PC-3 , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/metabolismo
7.
Prostate ; 79(10): 1147-1155, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31077419

RESUMEN

BACKGROUND: Cigarette smoking is associated with worse outcomes in prostate cancer, whose growth is dependent on androgen receptor (AR) signaling. We aimed to elucidate the biological effect of cigarette smoking on AR signaling and its clinical influence on oncological outcome. METHODS: Gene expression levels after exposure to tobacco smoke condensate (TSC) were evaluated by quantitative real-time polymerase chain reaction and Western blot analysis in prostate cancer cells. Cellular sensitivities to enzalutamide and docetaxel after TSC exposure were evaluated using a prostate cancer cell proliferation assay. Prognosis was compared between current smokers and nonsmokers when treated with AR-axis-targeting (ARAT) agent enzalutamide and docetaxel. RESULTS: Expression of AR variants as well as prostate-specific antigen was augmented after TSC exposure, which occurred after Akt phosphorylation. These inductions were suppressed by Akt inhibitor LY294002 as well as antioxidant N-acetylcysteine. Consistently, TSC exposure augmented cellular resistance to enzalutamide. In clinical data, cigarette smoking was associated with worse progression-free survival and cancer-specific survival when patients with prostate cancer were treated with ARAT agents but not docetaxel. CONCLUSIONS: It was suggested that cigarette smoking leads to detrimental oncological outcome when prostate cancer patients are treated with ARAT agents through induction of aberrant AR signaling. Accordingly, we recommend that patients with advanced prostate cancer should refrain from cigarette smoking.


Asunto(s)
Antagonistas de Andrógenos/farmacología , Fumar Cigarrillos/patología , Resistencia a Antineoplásicos/efectos de los fármacos , Neoplasias de la Próstata/patología , Receptores Androgénicos/metabolismo , Antagonistas de Andrógenos/uso terapéutico , Androstenos/farmacología , Androstenos/uso terapéutico , Benzamidas , Línea Celular Tumoral , Fumar Cigarrillos/metabolismo , Humanos , Masculino , Nitrilos , Feniltiohidantoína/análogos & derivados , Feniltiohidantoína/farmacología , Feniltiohidantoína/uso terapéutico , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/metabolismo
8.
J Biol Chem ; 292(6): 2191-2202, 2017 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-28028174

RESUMEN

DOCK8 is an atypical guanine nucleotide exchange factor for Cdc42, and its mutations cause combined immunodeficiency in humans. Accumulating evidence indicates that DOCK8 regulates the migration and activation of various subsets of leukocytes, but its regulatory mechanism is poorly understood. We here report that DOCK8-deficient macrophages exhibit a migration defect in a 2D setting. Although DOCK8 deficiency in macrophages did not affect the global Cdc42 activation induced by chemokine stimulation, rescue experiments revealed that the guanine nucleotide exchange factor activity of DOCK8 was required for macrophage migration. We found that DOCK8 associated with LRAP35a, an adaptor molecule that binds to the Cdc42 effector myotonic dystrophy kinase-related Cdc42-binding kinase, and facilitated its activity to phosphorylate myosin II regulatory light chain. When this interaction was disrupted in WT macrophages, they showed a migration defect, as seen in DOCK8-deficient macrophages. These results suggest that, during macrophage migration, DOCK8 links Cdc42 activation to actomyosin dynamics through the association with LRAP35a.


Asunto(s)
Movimiento Celular/fisiología , Factores de Intercambio de Guanina Nucleótido/fisiología , Macrófagos/metabolismo , Proteína de Unión al GTP cdc42/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Células Cultivadas , Factores de Intercambio de Guanina Nucleótido/metabolismo , Macrófagos/citología , Ratones , Ratones Noqueados , Fosforilación , Unión Proteica
9.
Nat Commun ; 15(1): 5004, 2024 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-38902223

RESUMEN

The differentiation of B cells into plasma cells is associated with substantial transcriptional and epigenetic remodeling. H3.3 histone variant marks active chromatin via replication-independent nucleosome assembly. However, its role in plasma cell development remains elusive. Herein, we show that during plasma cell differentiation, H3.3 is downregulated, and the deposition of H3.3 and chromatin accessibility are dynamically changed. Blockade of H3.3 downregulation by enforced H3.3 expression impairs plasma cell differentiation in an H3.3-specific sequence-dependent manner. Mechanistically, enforced H3.3 expression inhibits the upregulation of plasma cell-associated genes such as Irf4, Prdm1, and Xbp1 and maintains the expression of B cell-associated genes, Pax5, Bach2, and Bcl6. Concomitantly, sustained H3.3 expression prevents the structure of chromatin accessibility characteristic for plasma cells. Our findings suggest that appropriate H3.3 expression and deposition control plasma cell differentiation.


Asunto(s)
Diferenciación Celular , Histonas , Células Plasmáticas , Diferenciación Celular/genética , Histonas/metabolismo , Células Plasmáticas/metabolismo , Células Plasmáticas/citología , Animales , Ratones , Cromatina/metabolismo , Cromatina/genética , Humanos , Ratones Endogámicos C57BL , Factores Reguladores del Interferón/metabolismo , Factores Reguladores del Interferón/genética , Linfocitos B/metabolismo , Linfocitos B/citología , Regulación de la Expresión Génica
10.
Free Radic Biol Med ; 221: 81-88, 2024 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-38762061

RESUMEN

Androgen receptor (AR)-targeting therapy induces oxidative stress in prostate cancer. However, the mechanism of oxidative stress induction by AR-targeting therapy remains unclear. This study investigated the mechanism of oxidative stress induction by AR-targeting therapy, with the aim to develop novel therapeutics targeting oxidative stress induced by AR-targeting therapy. Intracellular reactive oxygen species (ROS) was examined by fluorescence microscopy and flow cytometry analysis. The effects of silencing gene expression and small molecule inhibitors on gene expression and cytotoxic effects were examined by quantitative real-time PCR and cell proliferation assay. ROS induced by androgen depletion co-localized with peroxisomes in prostate cancer cells. Among peroxisome-related genes, PPARA was commonly induced by AR inhibition and involved in ROS production via PKC signaling. Inhibition of PPARα by specific siRNA and a small molecule inhibitor suppressed cell proliferation and increased cellular sensitivity to the antiandrogen enzalutamide in prostate cancer cells. This study revealed a novel pathway by which AR inhibition induced intracellular ROS mainly in peroxisomes through PPARα activation in prostate cancer. This pathway is a promising target for the development of novel therapeutics for prostate cancer in combination with AR-targeting therapy such as antiandrogen enzalutamide.


Asunto(s)
Benzamidas , Proliferación Celular , Resistencia a Antineoplásicos , Nitrilos , Estrés Oxidativo , PPAR alfa , Peroxisomas , Feniltiohidantoína , Neoplasias de la Próstata , Especies Reactivas de Oxígeno , Receptores Androgénicos , Masculino , Humanos , Feniltiohidantoína/farmacología , Nitrilos/farmacología , Peroxisomas/metabolismo , Peroxisomas/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Benzamidas/farmacología , Receptores Androgénicos/metabolismo , Receptores Androgénicos/genética , Especies Reactivas de Oxígeno/metabolismo , PPAR alfa/metabolismo , PPAR alfa/genética , Proliferación Celular/efectos de los fármacos , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/genética , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Antagonistas de Receptores Androgénicos/farmacología , ARN Interferente Pequeño/genética
11.
Low Urin Tract Symptoms ; 13(2): 299-307, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33089671

RESUMEN

OBJECTIVE: To develop a new mouse model of underactive bladder (UAB) caused by chronic bladder outlet obstruction (BOO). METHODS: BOO was created in 6-week-old male C57BL/6 mice using surgery to loosely place a silver jump ring around the bladder neck of each mouse. Micturition behavior (assessed with a metabolic cage) and cystometry were used to evaluate bladder function at 8 and 16 weeks after BOO. Following completion of the functional studies, the bladders of the mice were excised, weighed, and subjected to histological analysis. RESULTS: Micturition behavior analysis showed that mice subjected to BOO for 16 weeks had a lower frequency of micturition (7.3 ± 1.1 vs 12.5 ± 3.0 times/d, P < .05) and volume per void (106.0 ± 0.1 vs 133.9 ± 3.2 µL, P < .05) than mice subjected to BOO for 8 weeks. Cystometry revealed that mice subjected to BOO for 16 weeks had lower baseline pressure (8.4 ± 0.6 vs 14.0 ± 0.7 cmH2 O, P < .01) and micturition pressure (13.9 ± 1.1 vs 42.8 ± 1.7 cmH2 O, P < .05) than mice subjected to BOO for 8 weeks. BOO caused progressive increases in bladder mass and collagen deposition over time. CONCLUSIONS: We successfully established a novel mouse model of UAB using surgery to place a silver jump ring loosely on the bladder neck. BOO initially induced bladder overactivity but subsequently resulted in UAB due to deterioration of detrusor smooth muscle contractility and progressive deposition of collagen in the bladder wall.


Asunto(s)
Obstrucción del Cuello de la Vejiga Urinaria , Vejiga Urinaria de Baja Actividad , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Obstrucción del Cuello de la Vejiga Urinaria/etiología , Micción
12.
Front Oncol ; 11: 697955, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34113577

RESUMEN

Transforming growth factor-ß1 (TGF-ß1) plays a dual role in cancer, acting as a tumor suppressor in the early stage of cancer development and as a tumor promoter in the later stage of cancer progression in various cancers. In this study, we investigated the association between genetic polymorphisms in TGFB1 and clinicopathological characteristics or oncological outcome in prostate cancer cases treated with androgen-deprivation therapy (ADT) according to metastasis status. Japanese male patients with hormone-sensitive prostate cancer treated with ADT from 1993 to 2005 were included in this study. Genomic DNA was obtained from whole blood samples, and genotyping of TGFB1 (rs2241716 and rs4803455) was performed by PCR-based technique. No significant association between genetic polymorphisms in TGFB1 (rs2241716 and rs4803455) and clinicopathological parameters or prognosis was observed in patients with non-metastatic disease. In patients with metastatic disease, Gleason score in CT/TT carriers (rs2241716) and CA/AA carriers (rs4803455) was unfavorable compared with CC carriers. In addition, the CT/TT alleles in rs2241716 (hazard ratio, 1.82; 95% confidence interval, 1.12-2.94; P = 0.015) and the CA/AA alleles in rs4803455 (hazard ratio, 1.75; 95% confidence interval, 1.03-2.98; P = 0.040) were associated with a higher risk of progression during ADT compared with the CC allele in patients with metastatic disease. TGFB1 genetic variations were associated with adverse characteristics and progression risk in ADT among patients with metastatic disease, but not those with non-metastatic disease, supporting a distinct role of TGF-ß signaling between non-metastatic and metastatic prostate cancer.

13.
Am J Transl Res ; 12(6): 3033-3045, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32655828

RESUMEN

Adipocytokines such as leptin and adiponectin have functions in metabolism as well as the development and progression of various types of malignancies. However, little is known about their role in bladder cancer. In this study, we investigated whether leptin, adiponectin, and their receptors have an impact on bladder cancer outgrowth and the mechanisms involved. We performed immunohistochemistry for leptin, leptin receptor (Ob-R), adiponectin, and adiponectin receptors (AdipoR1, AdipoR2) in bladder cancer tissue microarrays. Wound healing assay and western blot were then performed in human bladder cancer lines. The positive rates (0 vs 1+/2+/3+) of Ob-R (P=0.004), adiponectin (P<0.001), AdipoR1 (P=0.016), and AdipoR2 (P<0.001) expression were significantly higher in bladder tumors than in benign urothelial tissues. Strong (3+) leptin expression tended to be present more often in tumors (10.2%; P=0.079) than in benign tissues (3.2%). Multivariate analysis revealed a lower risk of recurrence (hazard ratio [HR]=0.432; 95% confidence interval [CI]=0.198-0.942; P=0.034) in patients with an adiponectin-positive non-muscle-invasive tumor and a higher risk of progression (HR=5.148, 95% CI=1.190-22.273; P=0.028) in patients with a leptin-positive muscle-invasive tumor. Treatment of two bladder cancer cell lines with a synthetic adiponectin inhibited their migration and the expressions of phospho-NF-κB, NF-κB, snail, slug, Y-box-binding protein 1, and COX-2, whereas leptin showed reverse effects. Downregulation of adiponectin expression and upregulation of leptin expression were independent predictors for the recurrence of non-muscle-invasive bladder tumors and progression of muscle-invasive bladder tumors, respectively. In summary, synthetic adiponectin might exhibit antitumor activity against bladder cancer.

14.
Urol Oncol ; 38(11): 849.e11-849.e18, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32712140

RESUMEN

OBJECTIVE: Androgen metabolism is a key component in therapeutic resistance to androgen deprivation therapy (ADT). This study aimed to reveal the significance of genetic polymorphisms in genes involved in androgen metabolism, including CYP17A1, AKR1C3, and HSD17B, on serum testosterone levels during ADT, as well as the prognosis of men undergoing ADT for metastatic prostate cancer (CaP). MATERIALS AND METHODS: This study included 104 Japanese patients with metastatic CaP, for whom serum testosterone data during ADT were available for 80 patients. The association of CYP17A1 (rs743572), AKR1C3 (rs12529), HSD17B1 (rs605059), HSD17B3 (rs2066479), and HSD17B4 (rs7737181) with serum testosterone levels during ADT and prognosis (progression-free survival and overall survival) was examined. Enzymatic activity in AKR1C3 H5Q was examined using recombinant protein. RESULTS: Homozygous wild-type (GG allele; median [interquartile range], 12.0 ng/ml [8.0-19.0 ng/ml]) AKR1C3 rs12529 was associated with higher serum testosterone levels during ADT compared with variant-type (GC/CC alleles; median [interquartile range], 9.0 ng/ml [6.4-10.8 ng/ml]). Consistently, variant-type (GC/CC alleles) AKR1C3 rs12529 showed significantly lower risk of progression (hazard ratio [95% confidence interval], 0.47 [0.24-0.96], P = 0.039) compared with homozygous wild-type (GG allele) on multivariate analysis. Meanwhile, other genetic variations were associated with neither serum testosterone during ADT nor prognosis. Enzyme activity of wild-type AKR1C3 was comparable to the H5Q mutant. CONCLUSIONS: Taken together, this study demonstrated that AKR1C3 polymorphism, which was associated with serum testosterone levels during ADT, may be a prognostic factor of the progression to castration-resistant prostate cancer in Japanese men with metastatic CaP.


Asunto(s)
Antagonistas de Andrógenos/uso terapéutico , Andrógenos/metabolismo , Neoplasias de la Próstata/sangre , Neoplasias de la Próstata/tratamiento farmacológico , Testosterona/sangre , Anciano , Andrógenos/genética , Humanos , Masculino , Polimorfismo Genético , Pronóstico , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Estudios Retrospectivos
15.
Cancer Chemother Pharmacol ; 83(5): 933-938, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30868236

RESUMEN

PURPOSE: Inadequate suppression of testosterone during androgen-deprivation therapy impairs its efficacy. This study investigated the significance of genetic polymorphism in CYP19A1, which encodes aromatase that catalyzes androgens into estrogens, among men treated with primary ADT for metastatic prostate cancer. METHODS: This study included 80 Japanese patients with metastatic prostate cancer whose serum testosterone levels during ADT were available. The association of CYP19A1 gene polymorphism (rs1870050) with clinicopathological parameters including serum testosterone levels during ADT as well as progression-free survival and overall survival was examined. RESULTS: Serum testosterone levels during ADT of men carrying homozygous wild-type (AA) in the CYP19A1 gene [median (interquartile range); 11.6 (8.3-20.3) ng/dl] were higher than those in men carrying the heterozygous/homozygous variant (AC/CC) [median (interquartile range); 10.0 (6.4-12.8) ng/dl]. When adjusted by Gleason score, initial PSA, M-stage and serum testosterone level during ADT, heterozygous/homozygous variant (AC/CC) in the CYP19A1 gene was associated with a lower risk of progression to castration resistance [hazard ratio (95% confidence interval), 0.53 [0.29-0.92], p = 0.025], but not to any-cause death [hazard ratio (95% confidence interval), 0.74 [0.36-1.49], p = 0.40]. CONCLUSIONS: These findings suggest that genetic variation in CYP19A1 (rs1870050) might affect the prognosis of patients with metastatic prostate cancer when treated with ADT by regulating serum testosterone levels.


Asunto(s)
Antagonistas de Andrógenos/administración & dosificación , Aromatasa/genética , Neoplasias de la Próstata/tratamiento farmacológico , Testosterona/sangre , Anciano , Supervivencia sin Enfermedad , Humanos , Japón , Masculino , Metástasis de la Neoplasia , Polimorfismo Genético , Pronóstico , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Estudios Retrospectivos , Tasa de Supervivencia
16.
Clin Genitourin Cancer ; 17(3): e387-e393, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31036465

RESUMEN

INTRODUCTION: Testosterone suppression in serum during androgen deprivation therapy (ADT) can affect the oncologic outcome of ADT. Although genetic variants in sex hormone-binding globulin (SHBG) were reported to be correlated with serum testosterone level, the association with serum testosterone during ADT remains unclear. Therefore, this study investigated the impact of a missense polymorphism in the SHBG gene among men treated with primary ADT for metastatic prostate cancer. PATIENTS AND METHODS: This study included 104 Japanese men with metastatic prostate cancer. The association of SHBG gene polymorphism (rs6259, D356N) with clinicopathologic parameters including serum testosterone levels during ADT, as well as prognosis, including progression-free survival and overall survival, was examined. RESULTS: The serum testosterone levels during ADT were comparable between men carrying the homozygous wild-type (GG) and heterozygous/homozygous variant (GA/AA) in the SHBG gene. When adjusted for age, Gleason score, initial prostate-specific antigen, and clinical T-stage, the heterozygous/homozygous variant (GA/AA) in the SHBG gene was associated with a higher risk of progression (hazard ratio, 2.20; 95% confidence interval, 1.10-4.18; P = .027) and any-cause death (hazard ratio, 3.21; 95% confidence interval, 1.31-7.35; P = .012). CONCLUSIONS: This study suggested genetic variation in SHBG (rs6259) might be an independent prognostic biomarker among men treated with primary ADT for metastatic prostate cancer.


Asunto(s)
Antagonistas de Andrógenos/uso terapéutico , Biomarcadores de Tumor/genética , Polimorfismo Genético , Neoplasias de la Próstata/genética , Globulina de Unión a Hormona Sexual/genética , Testosterona/sangre , Anciano , Progresión de la Enfermedad , Estudios de Seguimiento , Humanos , Japón , Masculino , Persona de Mediana Edad , Pronóstico , Neoplasias de la Próstata/sangre , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/secundario , Estudios Retrospectivos
17.
Cell Rep ; 29(9): 2823-2834.e7, 2019 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-31775048

RESUMEN

Intestinal microfold cells (M cells) in Peyer's patches are a special subset of epithelial cells that initiate mucosal immune responses through uptake of luminal antigens. Although the cytokine receptor activator of nuclear factor-κB ligand (RANKL) expressed on mesenchymal cells triggers differentiation into M cells, other environmental cues remain unknown. Here, we show that the metastasis-promoting protein S100A4 is required for development of mature M cells. S100A4-producing cells are a heterogenous cell population including lysozyme-expressing dendritic cells and group 3 innate lymphoid cells. We found that in the absence of DOCK8, a Cdc42 activator critical for interstitial leukocyte migration, S100A4-producing cells are reduced in the subepithelial dome, resulting in a maturation defect of M cells. While S100A4 promotes differentiation into mature M cells in organoid culture, genetic inactivation of S100a4 prevents the development of mature M cells in mice. Thus, S100A4 is a key environmental cue that regulates M cell differentiation in collaboration with RANKL.


Asunto(s)
Linfocitos/metabolismo , Ganglios Linfáticos Agregados/metabolismo , Proteína de Unión al Calcio S100A4/metabolismo , Diferenciación Celular , Humanos
18.
Front Immunol ; 9: 243, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29503648

RESUMEN

A hallmark of humoral immune responses is the production of antibodies. This process involves a complex cascade of molecular and cellular interactions, including recognition of specific antigen by the B cell receptor (BCR), which triggers activation of B cells and differentiation into plasma cells (PCs). Although activation of the small GTPase Rac has been implicated in BCR-mediated antigen recognition, its precise role in humoral immunity and the upstream regulator remain elusive. DOCK2 is a Rac-specific guanine nucleotide exchange factor predominantly expressed in hematopoietic cells. We found that BCR-mediated Rac activation was almost completely lost in DOCK2-deficient B cells, resulting in defects in B cell spreading over the target cell-membrane and sustained growth of BCR microclusters at the interface. When wild-type B cells were stimulated in vitro with anti-IgM F(ab')2 antibody in the presence of IL-4 and IL-5, they differentiated efficiently into PCs. However, BCR-mediated PC differentiation was severely impaired in the case of DOCK2-deficient B cells. Similar results were obtained in vivo when DOCK2-deficient B cells expressing a defined BCR specificity were adoptively transferred into mice and challenged with the cognate antigen. In addition, by generating the conditional knockout mice, we found that DOCK2 expression in B-cell lineage is required to mount antigen-specific IgG antibody. These results highlight important role of the DOCK2-Rac axis in PC differentiation and IgG antibody responses.


Asunto(s)
Diferenciación Celular/inmunología , Proteínas Activadoras de GTPasa/fisiología , Inmunidad Humoral , Inmunoglobulina G/metabolismo , Células Plasmáticas/fisiología , Traslado Adoptivo , Animales , Membrana Celular/inmunología , Femenino , Factores de Intercambio de Guanina Nucleótido , Inmunoglobulina G/inmunología , Sinapsis Inmunológicas/inmunología , Sinapsis Inmunológicas/metabolismo , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Células Madre Embrionarias de Ratones/trasplante , Receptores de Antígenos de Linfocitos B/inmunología , Receptores de Antígenos de Linfocitos B/metabolismo , Quimera por Trasplante , Proteínas de Unión al GTP rac/inmunología , Proteínas de Unión al GTP rac/metabolismo
19.
Sci Signal ; 11(541)2018 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-30065028

RESUMEN

Although immune responses are essential to protect the body from infection, they can also harm tissues. Certain tissues and organs, including the eye, constitute specialized microenvironments that locally inhibit immune reactivity. Dedicator of cytokinesis protein 2 (DOCK2) is a Rac-specific guanine nucleotide exchange factor (GEF) that is predominantly found in hematopoietic cells. DOCK2 plays a key role in immune surveillance because it is essential for the activation and migration of leukocytes. DOCK2 mutations cause severe immunodeficiency in humans. We found that DOCK2-mediated Rac activation and leukocyte migration were effectively inhibited by cholesterol sulfate (CS), but not by cholesterol or other sulfated steroids. CS bound to the catalytic domain of DOCK2 and suppressed its GEF activity. Mass spectrometric quantification revealed that CS was most abundantly produced in the Harderian gland, which provides the lipids that form the oily layer of the tear film. Sulfation of cholesterol is mediated by the sulfotransferases SULT2B1b and, to a lesser extent, SULT2B1a, which are produced from the same gene through alternative splicing. By genetically inactivating Sult2b1, we showed that the lack of CS in mice augmented ultraviolet- and antigen-induced ocular surface inflammation, which was suppressed by administration of eye drops containing CS. Thus, CS is a naturally occurring DOCK2 inhibitor and contributes to the generation of the immunosuppressive microenvironment in the eye.


Asunto(s)
Ésteres del Colesterol/metabolismo , Ojo/inmunología , Proteínas Activadoras de GTPasa/antagonistas & inhibidores , Evasión Inmune , Queratitis/prevención & control , Trastornos por Fotosensibilidad/prevención & control , Animales , Modelos Animales de Enfermedad , Ojo/efectos de los fármacos , Ojo/metabolismo , Proteínas Activadoras de GTPasa/genética , Proteínas Activadoras de GTPasa/metabolismo , Factores de Intercambio de Guanina Nucleótido , Queratitis/etiología , Queratitis/inmunología , Queratitis/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Trastornos por Fotosensibilidad/etiología , Trastornos por Fotosensibilidad/inmunología , Trastornos por Fotosensibilidad/metabolismo , Inhibidores de Serina Proteinasa/farmacología , Sulfotransferasas/fisiología
20.
Nat Commun ; 8: 13946, 2017 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-28067314

RESUMEN

Mutations of DOCK8 in humans cause a combined immunodeficiency characterized by atopic dermatitis with high serum IgE levels. However, the molecular link between DOCK8 deficiency and atopic skin inflammation is unknown. Here we show that CD4+ T cells from DOCK8-deficient mice produce large amounts of IL-31, a major pruritogen associated with atopic dermatitis. IL-31 induction critically depends on the transcription factor EPAS1, and its conditional deletion in CD4+ T cells abrogates skin disease development in DOCK8-deficient mice. Although EPAS1 is known to form a complex with aryl hydrocarbon receptor nuclear translocator (ARNT) and control hypoxic responses, EPAS1-mediated Il31 promoter activation is independent of ARNT, but in collaboration with SP1. On the other hand, we find that DOCK8 is an adaptor and negative regulator of nuclear translocation of EPAS1. Thus, EPAS1 links DOCK8 deficiency to atopic skin inflammation via IL-31 induction in CD4+ T cells.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Linfocitos T CD4-Positivos/inmunología , Dermatitis Atópica/genética , Factores de Intercambio de Guanina Nucleótido/genética , Interleucinas/genética , Transporte Activo de Núcleo Celular , Animales , Translocador Nuclear del Receptor de Aril Hidrocarburo/genética , Translocador Nuclear del Receptor de Aril Hidrocarburo/inmunología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/deficiencia , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/inmunología , Linfocitos T CD4-Positivos/patología , Núcleo Celular/inmunología , Núcleo Celular/metabolismo , Citosol/inmunología , Citosol/metabolismo , Dermatitis Atópica/inmunología , Dermatitis Atópica/patología , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Factores de Intercambio de Guanina Nucleótido/deficiencia , Factores de Intercambio de Guanina Nucleótido/inmunología , Heterocigoto , Inmunoglobulina E/genética , Inmunoglobulina E/inmunología , Interleucinas/inmunología , Masculino , Ratones , Ratones Noqueados , Regiones Promotoras Genéticas , Transporte de Proteínas , Transducción de Señal , Factor de Transcripción Sp1/genética , Factor de Transcripción Sp1/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA