Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Sci Rep ; 14(1): 20575, 2024 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-39232046

RESUMO

Glioblastoma multiforme (GBM) is the most aggressive type of cancer in the brain and has an inferior prognosis because of the lack of suitable medicine, largely due to its tremendous invasion. GBM has selfish metabolic pathways to promote migration, invasion, and proliferation compared to normal cells. Among various metabolic pathways, NAD (nicotinamide adenine dinucleotide) is essential in generating ATP and is used as a resource for cancer cells. LbNOX (Lactobacillus brevis NADH oxidase) is an enzyme that can directly manipulate the NAD+/NADH ratio. In this study, we found that an increased NAD+/NADH ratio by LbNOX or mitoLbNOX reduced intracellular glutamate and calcium responses and reduced invasion capacity in GBM. However, the invasion was not affected in GBM by rotenone, an ETC (Electron Transport Chain) complex I inhibitor, or nicotinamide riboside, a NAD+ precursor, suggesting that the crucial factor is the NAD+/NADH ratio rather than the absolute quantity of ATP or NAD+ for the invasion of GBM. To develop a more accurate and effective GBM treatment, our findings highlight the importance of developing a new medicine that targets the regulation of the NAD+/NADH ratio, given the current lack of effective treatment options for this brain cancer.


Assuntos
Glioblastoma , Metaboloma , NAD , Glioblastoma/metabolismo , Glioblastoma/patologia , NAD/metabolismo , Humanos , Linhagem Celular Tumoral , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Complexos Multienzimáticos/metabolismo , Levilactobacillus brevis/metabolismo , Invasividade Neoplásica , Cálcio/metabolismo , Ácido Glutâmico/metabolismo , Movimento Celular , Trifosfato de Adenosina/metabolismo , NADH NADPH Oxirredutases
2.
Neuro Oncol ; 2023 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-38085571

RESUMO

BACKGROUND: Reactive astrogliosis is a hallmark of various brain pathologies, including neurodegenerative diseases and glioblastomas. However, the specific intermediate metabolites contributing to reactive astrogliosis remain unknown. This study investigated how glioblastomas induce reactive astrogliosis in the neighboring microenvironment and explores 11C-acetate PET as an imaging technique for detecting reactive astrogliosis. METHODS: Through in vitro, mouse models, and human tissue experiments, we examined the association between elevated 11C-acetate uptake and reactive astrogliosis in gliomas. We explored acetate from glioblastoma cells, which triggers reactive astrogliosis in neighboring astrocytes by upregulating MAO-B and MCT1 expression. We evaluated the presence of cancer stem cells in the reactive astrogliosis region of glioblastomas and assessed the correlation between the volume of 11C-acetate uptake beyond MRI and prognosis. RESULTS: Elevated 11C-acetate uptake is associated with reactive astrogliosis and astrocytic MCT1 in the periphery of glioblastomas in human tissues and mouse models. Glioblastoma cells exhibit increased acetate production as a result of glucose metabolism, with subsequent secretion of acetate. Acetate derived from glioblastoma cells induces reactive astrogliosis in neighboring astrocytes by increasing the expression of MAO-B and MCT1. We found cancer stem cells within the reactive astrogliosis at the tumor periphery. Consequently, a larger volume of 11C-acetate uptake beyond contrast-enhanced MRI was associated with worse prognosis. CONCLUSION: Our results highlight the role of acetate derived from glioblastoma cells in inducing reactive astrogliosis and underscore the potential value of 11C-acetate PET as an imaging technique for detecting reactive astrogliosis, offering important implications for the diagnosis and treatment of glioblastomas.

3.
Front Cell Neurosci ; 15: 663092, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34149360

RESUMO

Glioblastoma (GBM) is the most common and malignant form of primary brain tumor with a median survival time of 14-16 months in GBM patients. Surgical treatment with chemotherapy and radiotherapy may help increase survival by removing GBM from the brain. However, complete surgical resection to eliminate GBM is almost impossible due to its high invasiveness. When GBM cells migrate to the brain, they interact with various cells, including astrocytes, neurons, endothelial cells, and the extracellular matrix (ECM). They can also make their cell body shrink to infiltrate into narrow spaces in the brain; thereby, they can invade regions of the brain and escape from surgery. Brain tumor cells create an appropriate microenvironment for migration and invasion by modifying and degrading the ECM. During those processes, the Ca2+ signaling pathway and other signaling cascades mediated by various ion channels contribute mainly to gene expression, motility, and invasion of GBM cells. Furthermore, GBM cells release glutamate, affecting migration via activation of ionotropic glutamate receptors in an autocrine manner. This review focuses on the cellular mechanisms of glioblastoma invasion and motility related to ECM, Ca2+ signaling, and glutamate. Finally, we discuss possible therapeutic interventions to inhibit invasion by GBM cells.

4.
Exp Neurobiol ; 30(2): 120-143, 2021 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-33972466

RESUMO

Central neurocytoma (CN) has been known as a benign neuronal tumor. In rare cases, CN undergoes malignant transformation to glioblastomas (GBM). Here we examined its cellular origin by characterizing differentiation potential and gene expression of CN-spheroids. First, we demonstrate that both CN tissue and cultured primary cells recapitulate the hierarchal cellular composition of subventricular zone (SVZ), which is comprised of neural stem cells (NSCs), transit amplifying progenitors (TAPs), and neuroblasts. We then derived spheroids from CN which displayed EGFR+/ MASH+ TAP and BLBP+ radial glial cell (RGC) characteristic, and mitotic neurogenesis and gliogenesis by single spheroids were observed with cycling multipotential cells. CN-spheroids expressed increased levels of pluripotency and tumor stem cell genes such as KLF4 and TPD5L1, when compared to their differentiated cells and human NSCs. Importantly, Gene Set Enrichment Analysis showed that gene sets of GBM-Spheroids, EGFR Signaling, and Packaging of Telomere Ends are enriched in CN-spheroids in comparison with their differentiated cells. We speculate that CN tumor stem cells have TAP and RGC characteristics, and upregulation of EGFR signaling as well as downregulation of eph-ephrin signaling have critical roles in tumorigenesis of CN. And their ephemeral nature of TAPs destined to neuroblasts, might reflect benign nature of CN.

5.
Front Cell Neurosci ; 12: 319, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30319359

RESUMO

Recently, µ-opioid receptor (MOR), one of the well-known Gi-protein coupled receptors (Gi-GPCR), was reported to be highly expressed in the hippocampal astrocytes. However, the role of astrocytic MOR has not been investigated. Here we report that activation of astrocytic MOR by [D-Ala2,N-MePhe4,Gly-ol]-enkephalin (DAMGO), a selective MOR agonist, causes a fast glutamate release using sniffer patch technique. We also found that the DAMGO-induced glutamate release was not observed in the astrocytes from MOR-deficient mice and MOR-short hairpin RNA (shRNA)-expressed astrocytes. In addition, the glutamate release was significantly reduced by gene silencing of the TREK-1-containing two-pore potassium (K2P) channel, which mediates passive conductance in astrocytes. Our findings were consistent with the previous study demonstrating that activation of Gi-GPCR such as cannabinoid receptor CB1 and adenosine receptor A1 causes a glutamate release through TREK-1-containing K2P channel from hippocampal astrocytes. We also demonstrated that MOR and TREK-1 are significantly co-localized in the hippocampal astrocytes. Furthermore, we found that both MOR and TREK-1-containing K2P channels are localized in the same subcellular compartments, soma and processes, of astrocytes. Our study raises a novel possibility that astrocytic MOR may participate in several physiological and pathological actions of opioids, including analgesia and addiction, through astrocytically released glutamate and its signaling pathway.

6.
Oncotarget ; 9(32): 22631-22641, 2018 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-29854303

RESUMO

INTRODUCTION: Clear-cell renal cell carcinoma (ccRCC) is the sixth most common malignancy in men in North America. Since ccRCC is a malignancy dependent on neovascularization, current first line systemic therapies like sunitinib, target the formation of new vessels allowing nutrient deprivation and cell death. However, recent studies have shown that patients develop resistance after approximately 1 year of treatment and show disease progression while on therapy. Therefore, we propose to identify the protein(s) responsible for increased migration with the aim of developing a new therapy that will target the identified protein and potentially slow down the progression of the disease. MATERIAL AND METHODS: Human renal cancer cell lines (Caki-1, Caki-2, ACHN) were treated with increasing doses of sunitinib to develop a sunitinib-conditioned renal cell carcinoma cell line. mRNA microarray and qPCR were performed to compare the differences in gene expression between Caki-1 sunitinib-conditioned and non-conditioned cells. NTN1 was assessed in our in vivo sunitinib-conditioned mouse model using immunostaining. xCELLigence and scratch assays were used to evaluate migration and MTS was used to evaluate cell viability. RESULTS: Human renal cell carcinoma sunitinib-conditioned cell lines showed upregulation of netrin-1 in microarray and q-PCR. Increased migration was demonstrated in Caki-1 sunitinib-conditioned cells when compared to the non-treated ones as well as, increased endothelial cell migration. Silencing of netrin-1 in sunitinib-conditioned Caki-1 cells did not demonstrate a significant reduction in cell migration. CONCLUSION: Netrin-1 is highly upregulated in renal cell carcinoma treated with sunitinib, but has no influence on cell viability or cell migration in metastatic RCC.

7.
Oncotarget ; 7(32): 51423-51434, 2016 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-27283491

RESUMO

Renal cell carcinomas (RCC) smaller than 7-cm are heterogeneous and exhibit metastatic potential in approximately 15% of cases. Although large-scale characterization of mutations in clear cell RCC (ccRCC), the most common RCC subtype, has been established, the genetic alterations related to ≤7-cm ccRCCs undergoing synchronous metastasis are poorly understood. To discover biomarkers that can be used to estimate the risk of synchronous metastasis in these ccRCC patients, we performed whole exome sequencing on the formalin-fixed paraffin-embedded (FFPE) samples of 10 ccRCC patients with ≤7-cm tumors and synchronous metastasis and expanded our study using The Cancer Genome Atlas (TCGA) ccRCC dataset (n = 201). Recurrent mutations were selected according to functional prediction and statistical significance. Mutations in three candidate genes, RELN (1 out of 10), FOXC2 (1 out of 10), and CLIP4 (2 out of 10) were found in expanded analysis using a TCGA cohort. Furthermore, siRNA-mediated target gene knockdown (FOXC2 and CLIP4) and overexpression (RELN) assays showed that FOXC2 and CLIP4 significantly increased cell migration and viability in ccRCCs. Our study demonstrated that FOXC2 and CLIP4 activity correlates to the presence of ≤7-cm ccRCCs with synchronous metastasis and may be potential molecular predictors of synchronous metastasis of ≤7-cm ccRCCs.


Assuntos
Carcinoma de Células Renais/genética , Carcinoma de Células Renais/patologia , Proteínas de Transporte/fisiologia , Fatores de Transcrição Forkhead/fisiologia , Neoplasias Renais/genética , Neoplasias Renais/patologia , Carga Tumoral/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/fisiologia , Proteínas de Transporte/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Feminino , Fatores de Transcrição Forkhead/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Proteínas de Membrana , Pessoa de Meia-Idade , Metástase Neoplásica , Prognóstico , Proteína Reelina
8.
Cancer Res Treat ; 48(1): 409-14, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25715769

RESUMO

von Hippel-Lindau (VHL) disease is an autosomal dominant inherited tumor syndrome associated with mutations of the VHL tumor suppressor gene located on chromosome 3p25. The loss of functional VHL protein contributes to tumorigenesis. This condition is characterized by development of benign and malignant tumors in the central nervous system (CNS) and the internal organs, including kidney, adrenal gland, and pancreas. We herein describe the case of a 74-year-old man carrying the VHL gene mutation who was affected by simultaneous colorectal adenocarcinoma, renal clear cell carcinoma, and hemangioblastomas of CNS.


Assuntos
Adenocarcinoma/complicações , Carcinoma de Células Renais/complicações , Neoplasias Cerebelares/complicações , Neoplasias Colorretais/complicações , Hemangioblastoma/complicações , Neoplasias Renais/complicações , Doença de von Hippel-Lindau/complicações , Idoso , Humanos , Masculino , Mutação , Proteína Supressora de Tumor Von Hippel-Lindau/genética , Doença de von Hippel-Lindau/genética
9.
Neoplasia ; 17(11): 805-16, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26678908

RESUMO

Vascular endothelial growth factor (VEGF)-targeted antiangiogenic therapy significantly inhibits the growth of clear cell renal cell carcinoma (RCC). Eventually, therapy resistance develops in even the most responsive cases, but the mechanisms of resistance remain unclear. Herein, we developed two tumor models derived from an RCC cell line by conditioning the parental cells to two different stresses caused by VEGF-targeted therapy (sunitinib exposure and hypoxia) to investigate the mechanism of resistance to such therapy in RCC. Sunitinib-conditioned Caki-1 cells in vitro did not show resistance to sunitinib compared with parental cells, but when tested in vivo, these cells appeared to be highly resistant to sunitinib treatment. Hypoxia-conditioned Caki-1 cells are more resistant to hypoxia and have increased vascularity due to the upregulation of VEGF production; however, they did not develop sunitinib resistance either in vitro or in vivo. Human endothelial cells were more proliferative and showed increased tube formation in conditioned media from sunitinib-conditioned Caki-1 cells compared with parental cells. Gene expression profiling using RNA microarrays revealed that several genes related to tissue development and remodeling, including the development and migration of endothelial cells, were upregulated in sunitinib-conditioned Caki-1 cells compared with parental and hypoxia-conditioned cells. These findings suggest that evasive resistance to VEGF-targeted therapy is acquired by activation of VEGF-independent angiogenesis pathways induced through interactions with VEGF-targeted drugs, but not by hypoxia. These results emphasize that increased inhibition of tumor angiogenesis is required to delay the development of resistance to antiangiogenic therapy and maintain the therapeutic response in RCC.


Assuntos
Inibidores da Angiogênese/metabolismo , Carcinoma de Células Renais/metabolismo , Sistemas de Liberação de Medicamentos , Indóis/metabolismo , Neoplasias Renais/metabolismo , Pirróis/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Inibidores da Angiogênese/administração & dosagem , Animais , Carcinoma de Células Renais/tratamento farmacológico , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Relação Dose-Resposta a Droga , Sistemas de Liberação de Medicamentos/métodos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/fisiologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Feminino , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Indóis/administração & dosagem , Neoplasias Renais/tratamento farmacológico , Camundongos , Camundongos Nus , Pirróis/administração & dosagem , Sunitinibe , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
10.
Oncotarget ; 6(33): 34818-30, 2015 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-26472187

RESUMO

Tumor microenvironments are characterized by decreased oxygen and nutrition due to the rapid and progressive nature of tumors and also stresses induced by several anti-tumor therapies. These intense cell stressors trigger a protective cell survival mechanism heralded by the unfolded protein response (UPR). The UPR is induced by an accumulation of unfolded proteins in the endoplasmic reticulum (ER) following cell starvation. Although the ER stress response is implicated in cytoprotection, its precise role during anti-angiogenic therapy remains unclear. One of the major proteins involved in ER stress is glucose-regulated protein 78 (GRP78), which binds to unfolded proteins and dissociates from membrane-bound ER stress sensors. To determine the role of ER stress responses during anti-angiogenic therapy and the potential role of GRP78 in combined therapy in renal cell carcinoma (RCC), we used GRP78 overexpressing or knockdown RCC cells under hypoxic or hypoglycemic conditions in vitro and in animal models treated with sunitinib. Here, we report that GRP78 plays a crucial role in protecting RCC cells from hypoxic and hypoglycemic stress induced by anti-angiogenic therapy. Knockdown of GRP78 using siRNA inhibited cancer cell survival and induced apoptosis in RCC cells in vitro and also resulted in ER stress-induced apoptosis and hypoxic/hypoglycemic stress-induced apoptosis by inactivating the PERK/eIF-2α pathway. Finally, GRP78 knockdown showed potent suppression of tumor growth and enhanced the antitumor effect of sunitinib in RCC xenografts. Our findings suggest that GRP78 may serve as a novel therapeutic target in combination with anti-angiogenic therapy for the management of RCC.


Assuntos
Carcinoma de Células Renais/patologia , Estresse do Retículo Endoplasmático/fisiologia , Proteínas de Choque Térmico/metabolismo , Neoplasias Renais/patologia , eIF-2 Quinase/metabolismo , Inibidores da Angiogênese/farmacologia , Animais , Western Blotting , Hipóxia Celular/fisiologia , Linhagem Celular Tumoral , Chaperona BiP do Retículo Endoplasmático , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Feminino , Imunofluorescência , Técnicas de Silenciamento de Genes , Humanos , Imuno-Histoquímica , Indóis/farmacologia , Camundongos , Camundongos Nus , Microscopia Confocal , Pirróis/farmacologia , RNA Interferente Pequeno , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Estresse Fisiológico , Sunitinibe , Transfecção , Ensaios Antitumorais Modelo de Xenoenxerto
11.
BMC Cancer ; 15: 812, 2015 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-26506914

RESUMO

BACKGROUND: Platinum-based systemic chemotherapy is the treatment of choice for patients with advanced urothelial carcinoma (UC). Although no chemotherapeutic regimen is established as a second-line therapy, recent studies reported that methotrexate, vinblastine, Adriamycin and cisplatin (MVAC) elicited a significant response in patients who failed gemcitabine and platinum (GP) chemotherapy. We investigated the clinical factors useful for predicting a favourable response to MVAC in UC patients who failed GP. METHODS: Forty-five patients with advanced UC who received second-line MVAC chemotherapy after failure with first-line GP chemotherapy were enrolled in this study. Univariate and multivariate analyses based on Cox's regression were performed to identify independent prognostic factors for progression-free survival (PFS) after second-line MVAC chemotherapy. RESULTS: The median follow-up period after the first MVAC administration was 10.0 months. The median PFS and overall survival (OS) were 6.5 months (95% confidence interval [CI]: 5.1-7.9) and 14.5 months (95% CI, 7.4-21.4), respectively. The overall response rate was 57.8%. The response to first-line GP chemotherapy (hazard ratio [HR], 2.500; p = 0.012) and patient age (HR, 1.047; p = 0.033) were predictors of PFS after MVAC chemotherapy. CONCLUSIONS: The response to first-line GP chemotherapy and age were independent predictors of PFS in patients who received second-line MVAC chemotherapy. This report is the first to describe independent predictors of PFS after MVAC chemotherapy.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Desoxicitidina/análogos & derivados , Platina/administração & dosagem , Neoplasias Urológicas/diagnóstico , Neoplasias Urológicas/tratamento farmacológico , Urotélio/patologia , Idoso , Cisplatino/administração & dosagem , Desoxicitidina/administração & dosagem , Intervalo Livre de Doença , Doxorrubicina/administração & dosagem , Feminino , Seguimentos , Humanos , Masculino , Metotrexato/administração & dosagem , Pessoa de Meia-Idade , Valor Preditivo dos Testes , Falha de Tratamento , Vimblastina/administração & dosagem , Gencitabina
12.
Urology ; 86(3): 459-64, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26142711

RESUMO

OBJECTIVE: To identify risk factors for metal stent failure in patients who received polymeric double J (PDJ) ureteral stents for malignant ureteral obstructions (MUOs) and review our clinical experiences using a ureteral metallic stent. PATIENTS AND METHODS: Patients who underwent metallic stent placement to replace a double J ureteral stent for nonurological MUO between January 2011 and February 2014 were included. The collected data included gender, age, laterality, cause of obstruction, PDJ ureteral stenting duration, immediate success of the metal stent, and additional procedures to relieve obstruction after metal stenting (eg, additional metal stenting or percutaneous nephrostomy (PCN) indwelling catheter placement). Cox regression tests were used for the statistical analyses. RESULTS: In this analysis 40 ureteral units were included. There was no initial technical failure. However, 9 (22.5%) units required additional procedures due to de novo ureteral obstruction, including additional indwelling metal stents (7.5%), additional PDJ stenting (10%), or indwelling percutaneous nephrostomy (5%). Univariate and multivariate analyses revealed that the duration of previous PDJ ureteral stenting was an independent prognostic factor for predicting ureteral metal stent failure (hazard ratio = 1.063, 95% confidence interval = 1.004-1.125; P = .037). CONCLUSION: Long-term indwelling of a PDJ ureteral stent increases the risk of additional management for de novo ureteral stricture after ureteral metal stent replacement for nonurological MUO. Our data suggest that careful patient selection and counseling for those at high risk are needed when metal stent replacement is considered for patients with long-term PDJ ureteral stents for MUO.


Assuntos
Falha de Equipamento , Stents Metálicos Autoexpansíveis , Obstrução Ureteral/etiologia , Obstrução Ureteral/cirurgia , Idoso , Neoplasias do Sistema Digestório/patologia , Desenho de Equipamento , Feminino , Neoplasias dos Genitais Femininos/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Reoperação , Estudos Retrospectivos , Fatores de Risco
13.
Urol Oncol ; 33(9): 383.e17-22, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26004165

RESUMO

PURPOSE: Several prostate-specific antigen (PSA) kinetics parameters such as nadir PSA level and time to nadir PSA level are used commonly as predictive prognostic factors for patients with metastatic prostate cancer (mPCa) who have undergone androgen deprivation therapy. However, based on the limitations of these factors, earlier and more clinically available prognostic factors are needed. Therefore, we examined the PSA half-life (PSAHL) estimated at the first follow-up visit as a prognostic factor of newly diagnosed mPCa. METHODS: We performed a retrospective review of 309 patients with newly diagnosed mPCa who had undergone androgen deprivation therapy. After categorizing the included patients to short and long PSAHL groups, based on the median PSAHL value, Cox regression analyses were performed to identify independent prognostic factors of newly diagnosed mPCa. The Kaplan-Meier method was used for detecting differences in survival between both the groups. RESULTS: The median follow-up period was 44 months, and the prostate cancer-specific mortality (PCSM)-free survival length was 65 months in all included patients. Long PSAHL group (hazard ratio [HR] = 2.383, P<0.001), nadir PSA level (HR = 1.004, P<0.001), time to nadir PSA level (HR = 0.856; P<0.001), and Gleason score 8 to 10 relative to 6 to 7 (HR = 2.025; P = 0.008) were found to be independent predictors of the PCSM. By the Kaplan-Meier method, the median PCSM-free survival of the short PSAHL group was 73.7 (95% CI: 54.8-92.6) and of the long PSAHL group was 52.5 months (95% CI: 33.4-71.6). This difference between both the groups was found to be statistically significant (P = 0.014, log rank test). CONCLUSIONS: PSAHL estimated at the first follow-up visit is an independent prognostic factor for newly diagnosed mPCa. If the prospective validation test is performed on a large scale, it may demonstrate that PSAHL is an early surrogate prognostic factor of newly diagnosed mPCa.


Assuntos
Antígeno Prostático Específico/sangue , Neoplasias da Próstata/sangue , Idoso , Biomarcadores Tumorais/sangue , Intervalo Livre de Doença , Meia-Vida , Humanos , Estimativa de Kaplan-Meier , Masculino , Modelos de Riscos Proporcionais , Neoplasias da Próstata/mortalidade , Estudos Retrospectivos
14.
Yonsei Med J ; 56(2): 368-74, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25683983

RESUMO

PURPOSE: To investigate the relationship between rising patterns of prostate-specific antigen (PSA) before chemotherapy and PSA flare during the early phase of chemotherapy in patients with castration-resistant prostate cancer (CRPC). MATERIALS AND METHODS: This study included 55 patients with CRPC who received chemotherapy and in whom pre-treatment or post-treatment PSA levels could be serially obtained. The baseline parameters included age, performance, Gleason score, PSA level, and disease extent. PSA doubling time was calculated using the different intervals: the conventional interval from the second hormone manipulation following the nadir until anti-androgen withdrawal (PSADT1), the interval from the initial rise after anti-androgen withdrawal to the start of chemotherapy (PSADT2), and the interval from the nadir until the start of chemotherapy (PSADT3). The PSA growth patterns were analyzed using the ratio of PSADT2 to PSADT1. RESULTS: There were two growth patterns of PSA doubling time: 22 patients (40.0%) had a steady pattern with a more prolonged PSADT2 than PSADT1, while 33 (60.0%) had an accelerating pattern with a shorter PSADT2 than PSADT1. During three cycles of chemotherapy, PSA flare occurred in 11 patients (20.0%); of these patients, 3 were among 33 (9.1%) patients with an accelerating PSA growth pattern and 8 were among 22 patients (36.4%) with a steady PSA growth pattern (p=0.019). Multivariate analysis showed that only PSA growth pattern was an independent predictor of PSA flare (p=0.034). CONCLUSION: An exponential rise in PSA during anti-androgen withdrawal is a significant predictor for PSA flare during chemotherapy in CRPC patients.


Assuntos
Antineoplásicos/uso terapêutico , Antígeno Prostático Específico/sangue , Neoplasias de Próstata Resistentes à Castração/sangue , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Taxoides/uso terapêutico , Idoso , Idoso de 80 Anos ou mais , Antagonistas de Androgênios , Biomarcadores Tumorais/sangue , Docetaxel , Seguimentos , Humanos , Avaliação de Estado de Karnofsky , Masculino , Pessoa de Meia-Idade , Gradação de Tumores , Valor Preditivo dos Testes , Neoplasias de Próstata Resistentes à Castração/patologia
15.
Mol Cancer Ther ; 14(4): 1024-34, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25657336

RESUMO

Renal cell carcinoma (RCC) is the most common malignancy in the kidney. Antiangiogenic targeted therapies inhibit the progression of RCC, but have limited impacts on invasion or metastasis of tumor cells. Integrin-linked kinase (ILK) is a serine/threonine kinase implicated in the regulation of cell growth/survival, cell-cycle progression, epithelial-mesenchymal transition (EMT), invasion/migration, and angiogenesis. However, the role of ILK in RCC has not been evaluated. We investigated the role of ILK on cancer progression and metastasis and the therapeutic potential of ILK inhibition in RCC. Our investigation reveals that ILK is expressed at a low level in normal cells and low-stage RCC cells and is highly expressed in advanced and metastatic cells. Caki-1, a metastatic RCC cell line, showed higher expression of molecular EMT markers, including Snail and Zeb1, but decreased activity of GSK3ß. Knockdown of ILK using small interference (si)-ILK minimally inhibited tumor proliferation and cell-cycle progression was not significantly affected. However, ILK knockdown suppressed the formation of stress fibers and focal adhesions and impeded phenotypic EMT markers, including cell migration and invasion, in Caki-1 and UMRC-3 cells. Finally, in vivo knockdown of ILK suppressed the progression, invasion, and metastasis of primary RCC in nude mice by downregulation of EMT markers (Snail, Zeb1, vimentin, and E-cadherin). Our results show that ILK may be essential for invasion and metastasis in RCC and regulates vimentin and E-cadherin expression by regulating the EMT-related transcription factors Snail and Zeb1. These results suggest that ILK may be a potential target in RCC.


Assuntos
Carcinoma de Células Renais/genética , Carcinoma de Células Renais/patologia , Transição Epitelial-Mesenquimal/genética , Proteínas Serina-Treonina Quinases/genética , Animais , Biomarcadores , Carcinoma de Células Renais/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Sobrevivência Celular/genética , Modelos Animais de Doenças , Regulação para Baixo , Expressão Gênica , Técnicas de Silenciamento de Genes , Xenoenxertos , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Camundongos , Estadiamento de Neoplasias , Fenótipo , Proteínas Serina-Treonina Quinases/metabolismo , Interferência de RNA , RNA Interferente Pequeno/genética , Fatores de Transcrição da Família Snail , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Homeobox 1 de Ligação a E-box em Dedo de Zinco
16.
BMC Cancer ; 15: 19, 2015 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-25613585

RESUMO

BACKGROUND: Epigenetic modifications play a critical role in the regulation of all DNA-based processes, such as transcription, repair, and replication. Inappropriate histone modifications can result in dysregulation of cell growth, leading to neoplastic transformation and cell death. Renal tumors have been shown to have a higher global methylation percentage and reduced histone acetylation. Preclinical models have revealed that histone gene modifiers and epigenetic alterations play important roles in renal cell carcinoma (RCC) tumorigenesis. Recently, a novel HDAC inhibitor, N-hydroxy-7-(2-naphthylthio) heptanomide (HNHA), has been introduced as an example of a new class of anti-cancer agents. The anti-cancer activity of HNHA and the underlying mechanisms of action remain to be clarified. METHODS: The MTS assay using a panel of RCC cells was used to evaluate the anti-proliferative effects of HNHA. The established HDAC inhibitors, SAHA and TSA, were used for comparison. Western blotting analysis was performed to investigate the acetylation of histone H3 and the expression of apoptotic markers in vitro and in vivo. Subcellular fractionation was performed to evaluate expression of Bax and cytochrome c in the cytosol and mitochondria, and also translocation of cytochrome c from the cytoplasm to the nucleus. A confocal microscopic evaluation was performed to confirm inhibition of cell proliferation, induction of apoptosis, and the nuclear translocation of cytochrome c in RCC cells. RESULTS: In this study, we investigated the apoptosis-inducing activity of HNHA in cultured kidney cancer cells. Apoptosis in the HNHA-treated group was induced significantly, with marked caspase activation and Bcl-2 suppression in RCC cells in vitro and in vivo. HNHA treatment caused cytochrome c release from mitochondria, which was mediated by increased Bax expression and caspase activation. HNHA also induced nuclear translocation of cytochrome c, suggesting that HNHA can induce caspase-independent nuclear apoptosis in RCC cells. An in vivo study showed that HNHA had greater anti-tumor and pro-apoptotic effects on RCC xenografts than the established HDAC inhibitors. CONCLUSIONS: HNHA has more potent anti-tumor activity than established HDAC inhibitors. Its activities are mediated by caspase-dependent and cytochrome-c-mediated apoptosis in RCC cells. These results suggest that HNHA may offer a new therapeutic approach to RCC.


Assuntos
Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Carcinoma de Células Renais/tratamento farmacológico , Citocromos c/metabolismo , Inibidores de Histona Desacetilases/uso terapêutico , Ácidos Hidroxâmicos/uso terapêutico , Neoplasias Renais/tratamento farmacológico , Naftalenos/uso terapêutico , Acetilação , Animais , Western Blotting/métodos , Carcinoma de Células Renais/enzimologia , Carcinoma de Células Renais/patologia , Caspases/metabolismo , Fracionamento Celular/métodos , Histonas/metabolismo , Humanos , Proteínas I-kappa B/metabolismo , Marcação In Situ das Extremidades Cortadas , Neoplasias Renais/enzimologia , Neoplasias Renais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Mitocôndrias/enzimologia , Transplante de Neoplasias , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Células Tumorais Cultivadas , Proteína X Associada a bcl-2/metabolismo
17.
J Urol ; 193(4): 1239-44, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25444987

RESUMO

PURPOSE: We determined the prognostic impact of a synchronous second primary malignancy on overall survival in patients with metastatic prostate cancer. Identifying features that stratify the risk of overall survival is critical for judiciously applying definitive therapy. MATERIALS AND METHODS: We retrospectively analyzed the records of 582 consecutive patients with prostate cancer diagnosed with metastasis between May 7, 1998 and August 27, 2011. Patient age, body mass index, ECOG performance status, Charlson comorbidity index, prostate specific antigen, T and N stages, Gleason and ASA® scores, progression to castration resistant prostate cancer, prior local treatments and synchronous second primary malignancies at metastasis were assessed. A synchronous second primary malignancy was defined as a cytologically or histologically proven solid malignancy. Cox proportional hazards regression analysis was done to estimate overall survival by second primary type and evaluate predictive variables. RESULTS: A total of 164 patients (28.1%) had a synchronous second primary malignancy, of which colorectal (9.1%), stomach (7.3%) and lung (7.1%) cancers were the most prevalent types. During a median followup of 34.1 months patients without a synchronous second primary malignancy had a significantly higher overall survival rate than those with lung or stomach cancer. However, men without a second malignancy had outcomes comparable to those in men with colorectal cancer. Clinical stage T4 or greater, ASA score 1 or greater and lung or stomach cancer were independent predictors of overall mortality. CONCLUSIONS: A substantial proportion of patients with metastatic prostate cancer present with a synchronous second primary malignancy. Definitive therapy targeting prostate cancer may confer a limited survival benefit in patients with synchronous lung or stomach cancer.


Assuntos
Segunda Neoplasia Primária/mortalidade , Neoplasias da Próstata/mortalidade , Neoplasias da Próstata/patologia , Humanos , Masculino , Metástase Neoplásica , Prognóstico , Estudos Retrospectivos , Risco , Taxa de Sobrevida
18.
J Cancer Res Clin Oncol ; 140(10): 1769-76, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24858569

RESUMO

PURPOSE: A transient rise in prostate-specific antigen (PSA) after the initiation of chemotherapy, called as PSA flare, has been frequently reported in patients with castration-resistant prostate cancer (CRPC) but there has been no way to differentiate PSA rises in CRPC. We investigated whether bone-related serum markers differentiate PSA flare from progression in CRPC patients with bone metastasis. METHODS: We reviewed CRPC patients with bone metastasis who received systemic chemotherapy from 2002 to 2008. Pretreatment baseline and follow-up data including age, performance score, PSA, Gleason score, alkaline phosphatase (ALP), calcium level, and hemoglobin were evaluated. Pretreatment parameters and follow-up serum parameters after the first cycle of chemotherapy were included in statistical analyses. RESULTS: PSA increased in 38 patients (45.8 %) at the first evaluation after chemotherapy. Among the PSA rises, PSA increased continuously or did not decrease to the stabilization level by the third evaluation in 22 (26.5 %) patients, while PSA decreased to the stabilization or response level by the third evaluation in 16 (19.3 %). PSA flare occurred in 17 (20.5 %). The univariate analyses showed that no baseline parameters were associated with PSA flare, but the initial ALP decrease, changed ALP ratio, and median calcium level were significantly associated with PSA flare (p = 0.001, p = 0.008 and p = 0.012, respectively). Multivariate logistic regression analysis showed that a change in the ALP level is an independent predictive factor for PSA flare (p = 0.017). CONCLUSIONS: ALP is a useful biomarker to differentiate PSA flare from early PSA progression during docetaxel chemotherapy in CRPC patients with bone metastasis.


Assuntos
Fosfatase Alcalina/sangue , Antineoplásicos Fitogênicos/uso terapêutico , Biomarcadores Tumorais/sangue , Neoplasias Ósseas/secundário , Antígeno Prostático Específico/sangue , Neoplasias de Próstata Resistentes à Castração/sangue , Neoplasias de Próstata Resistentes à Castração/patologia , Taxoides/uso terapêutico , Idoso , Idoso de 80 Anos ou mais , Análise de Variância , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Ósseas/sangue , Cálcio/sangue , Progressão da Doença , Intervalo Livre de Doença , Docetaxel , Seguimentos , Hemoglobinas/metabolismo , Humanos , Modelos Logísticos , Masculino , Pessoa de Meia-Idade , Gradação de Tumores , Valor Preditivo dos Testes , Modelos de Riscos Proporcionais , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Neoplasias de Próstata Resistentes à Castração/enzimologia , Neoplasias de Próstata Resistentes à Castração/imunologia , Estudos Retrospectivos
19.
Cancer ; 116(10): 2332-42, 2010 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-20225226

RESUMO

BACKGROUND: Tumor vascularity is a potential predictor of treatment outcomes in metastatic renal cell carcinoma (mRCC), and contrast enhancement of tumors in computed tomography (CT) is correlated significantly with microvessel density. In this study, the authors investigated whether tumor enhancement in contrast-enhanced CT (CECT) is useful for predicting outcomes in patients with mRCC who are receiving antiangiogenic therapy. METHODS: Attenuation values were reviewed retrospectively on CECT images of all metastatic lesions in 66 patients from February 2007 to November 2008. All patients received a tyrosine kinase inhibitor (either sunitinib or sorafenib). Tumor response was evaluated on CECT studies every 12 weeks. The authors analyzed the association between contrast enhancement and treatment outcomes, including objective response, tumor size reduction rate, time to response, and time to progression. RESULTS: In 46 patients, 198 metastatic lesions were assessed. Tumor size was reduced in 140 lesions (70.7%) and was increased in 58 lesions (29.3%). The mean reduction in size was 23.8%. The overall mean time to response and the time to progression were 8.6 months and 16.4 months, respectively. In multivariate analyses, tumor enhancement and enhancement pattern were associated with objective responses (P = .003 and P = .028, respectively). In addition, tumor enhancement was associated with tumor size reduction (P = .004). In Cox proportional hazards models, only tumor enhancement was associated significantly with the time to size reduction and progression-free survival (P = .03 and P = .015, respectively). CONCLUSIONS: Tumor enhancement on CECT images was associated with treatment outcomes and was identified as a potential predictor of treatment outcomes after antiangiogenic therapy in patients with mRCC.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Neoplasias Renais/diagnóstico por imagem , Neoplasias Renais/tratamento farmacológico , Metástase Neoplásica/diagnóstico por imagem , Tomografia Computadorizada por Raios X/métodos , Adulto , Idoso , Carcinoma de Células Renais/diagnóstico por imagem , Meios de Contraste , Progressão da Doença , Sistemas de Liberação de Medicamentos , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Inibidores de Proteínas Quinases/uso terapêutico , Resultado do Tratamento
20.
Cancer Res ; 70(3): 1173-83, 2010 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-20103623

RESUMO

Calcium signaling is important in many signaling processes in cancer cell proliferation and motility including in deadly glioblastomas of the brain that aggressively invade neighboring tissue. We hypothesized that disturbing Ca(2+) signaling pathways might decrease the invasive behavior of giloblastoma, extending survival. Evaluating a panel of small-molecule modulators of Ca(2+) signaling, we identified caffeine as an inhibitor of glioblastoma cell motility. Caffeine, which is known to activate ryanodine receptors, paradoxically inhibits Ca(2+) increase by inositol 1,4,5-trisphospate receptor subtype 3 (IP(3)R3), the expression of which is increased in glioblastoma cells. Consequently, by inhibiting IP(3)R3-mediated Ca(2+) release, caffeine inhibited migration of glioblastoma cells in various in vitro assays. Consistent with these effects, caffeine greatly increased mean survival in a mouse xenograft model of glioblastoma. These findings suggest IP(3)R3 as a novel therapeutic target and identify caffeine as a possible adjunct therapy to slow invasive growth of glioblastoma.


Assuntos
Cafeína/farmacologia , Cálcio/metabolismo , Glioblastoma/tratamento farmacológico , Receptores de Inositol 1,4,5-Trifosfato/antagonistas & inibidores , Animais , Linhagem Celular , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Estimulantes do Sistema Nervoso Central/farmacologia , Relação Dose-Resposta a Droga , Regulação Neoplásica da Expressão Gênica , Glioblastoma/metabolismo , Glioblastoma/patologia , Humanos , Receptores de Inositol 1,4,5-Trifosfato/genética , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Invasividade Neoplásica , Análise de Sequência com Séries de Oligonucleotídeos , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Análise de Sobrevida , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA