Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
1.
Sci Rep ; 11(1): 1720, 2021 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-33462262

RESUMO

Exposure to Ionizing radiation (IR) poses a severe threat to human health. Therefore, there is an urgent need to develop potent and safe radioprotective agents for radio-nuclear emergencies. Phosphatidylinositol-3-kinase (PI3K) mediates its cytoprotective signaling against IR by phosphorylating membrane phospholipids to phosphatidylinositol 3,4,5 triphosphate, PIP3, that serve as a docking site for AKT. Phosphatase and Tensin Homolog on chromosome 10 (PTEN) antagonizes PI3K activity by dephosphorylating PIP3, thus suppressing PI3K/AKT signaling that could prevent IR induced cytotoxicity. The current study was undertaken to investigate the radioprotective potential of PTEN inhibitor (PTENi), bpV(HOpic). The cell cytotoxicity, proliferation index, and clonogenic survival assays were performed for assessing the radioprotective potential of bpV(HOpic). A safe dose of bpV(HOpic) was shown to be radioprotective in three radiosensitive tissue origin cells. Further, bpV(HOpic) significantly reduced the IR-induced apoptosis and associated pro-death signaling. A faster and better DNA repair kinetics was also observed in bpV(HOpic) pretreated cells exposed to IR. Additionally, bpV(HOpic) decreased the IR-induced oxidative stress and significantly enhanced the antioxidant defense mechanism in cells. The radioprotective effect of bpV(HOpic) was found to be AKT dependant and primarily regulated by the enhanced glycolysis and associated signaling. Furthermore, this in-vitro observation was verified in-vivo, where administration of bpV(HOpic) in C57BL/6 mice resulted in AKT activation and conferred survival advantage against IR-induced mortality. These results imply that bpV(HOpic) ameliorates IR-induced oxidative stress and cell death by inducing AKT signaling mediated antioxidant defense system and DNA repair pathways, thus strengthening its potential to be used as a radiation countermeasure.


Assuntos
Proliferação de Células/efeitos dos fármacos , PTEN Fosfo-Hidrolase/antagonistas & inibidores , Radiação Ionizante , Compostos de Vanádio/farmacologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Linhagem Celular , Proliferação de Células/efeitos da radiação , Dano ao DNA/efeitos dos fármacos , Dano ao DNA/efeitos da radiação , Glicólise/efeitos dos fármacos , Glicólise/efeitos da radiação , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/efeitos da radiação , PTEN Fosfo-Hidrolase/metabolismo , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/efeitos da radiação , Compostos de Vanádio/administração & dosagem , Irradiação Corporal Total
2.
Ann Surg ; 272(2): 311-318, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32675544

RESUMO

OBJECTIVE: We aimed to determine whether tumor metabolism could be prognostic of cure in L-EAC patients who receive definitive chemoradiation. SUMMARY BACKGROUND DATA: Patients with inoperable localized esophageal adenocarcinoma (L-EAC) often receive definitive chemoradiation; however, biomarkers and/or imaging variables to prognosticate cure are missing. METHODS: Two hundred sixty-six patients with L-EAC who had chemoradiation but not surgery were analyzed from the prospectively maintained EAC databases in the Department of Gastrointestinal Medical Oncology at The University of Texas MD Anderson Cancer Center (Texas, USA) between March 2002 and April 2015. Maximum standardized uptake value (SUVmax) and total lesion glycolysis (TLG) from the positron emission tomography data were evaluated. RESULTS: Of 266 patients, 253 (95%) were men; the median age was 67 years (range 20-91 yrs) and 153 had poorly differentiated L-EAC. The median SUVmax was 10.3 (range 0-87) and the median TLG was 85.7 (range 0-3227). Both SUVmax and TLG were higher among those with: tumors >5 cm in length, high clinical stage, and high tumor and node categories by TNM staging (all P < 0.0001). Of 234 patients evaluable for cure, 60 (25.6%) achieved cure. In the multivariable logistic regression model, low TLG (but not low SUVmax) was associated with cure (continuous TLG value: odds ratio 0.70, 95% confidence interval (CI) 0.54-0.92). TLG was quantified into 4 quartile categorical variables; first quartile (Q1; <32), second quartile (Q2; 32.0-85.6), third quartile (Q3; 85.6-228.4), and fourth quartile (Q4; >228.4); the cure rate was only 10.3% in Q4 and 5.1% in Q3 but increased to 28.8% in Q2, and 58.6% in Q1. The cross-validation resulted in an average accuracy of prediction score of 0.81 (95% CI, 0.75-0.86). CONCLUSIONS: In this cross-validated model, 59% of patients in the 1st quartile were cured following definitive chemoradiation. Baseline TLG could be pursued as one of the tools for esophageal preservation.


Assuntos
Adenocarcinoma/patologia , Adenocarcinoma/terapia , Quimiorradioterapia/métodos , Neoplasias Esofágicas/patologia , Neoplasias Esofágicas/terapia , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada/métodos , Adenocarcinoma/diagnóstico por imagem , Adenocarcinoma/mortalidade , Adulto , Idoso , Idoso de 80 Anos ou mais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Institutos de Câncer , Estudos de Coortes , Bases de Dados Factuais , Intervalo Livre de Doença , Neoplasias Esofágicas/diagnóstico por imagem , Neoplasias Esofágicas/mortalidade , Feminino , Seguimentos , Glicólise/efeitos dos fármacos , Glicólise/efeitos da radiação , Humanos , Estimativa de Kaplan-Meier , Modelos Logísticos , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica/patologia , Estadiamento de Neoplasias , Estudos Retrospectivos , Medição de Risco , Estatísticas não Paramétricas , Análise de Sobrevida , Texas , Fatores de Tempo , Resultado do Tratamento , Carga Tumoral/efeitos dos fármacos , Carga Tumoral/efeitos da radiação
3.
Cancer Lett ; 478: 107-121, 2020 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-32113989

RESUMO

Patients with mutated TP53 have been identified as having comparatively poor outcomes compared to those retaining wild-type p53 in many cancers, including squamous cell carcinomas of the head and neck (SCCHN). We have examined the role of p53 in regulation of metabolism in SCCHN cells and find that loss of p53 function determines the Warburg effect in these cells. Moreover, this metabolic adaptation to loss of p53 function creates an Achilles' heel for tumour cells that can be exploited for potential therapeutic benefit. Specifically, cells lacking normal wild-type p53 function, whether through mutation or RNAi-mediated downregulation, display a lack of metabolic flexibility, becoming more dependent on glycolysis and losing the ability to increase energy production from oxidative phosphorylation. Thus, cells that have compromised p53 function can be sensitised to ionizing radiation by pre-treatment with a glycolytic inhibitor. These results demonstrate the deterministic role of p53 in regulating energy metabolism and provide proof of principle evidence for an opportunity for patient stratification based on p53 status that can be exploited therapeutically using current standard of care treatment with ionising radiation.


Assuntos
Metabolismo Energético , Neoplasias de Cabeça e Pescoço/genética , Mutação , Proteína Supressora de Tumor p53/genética , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Metabolismo Energético/efeitos dos fármacos , Metabolismo Energético/efeitos da radiação , Glicólise/efeitos dos fármacos , Glicólise/efeitos da radiação , Neoplasias de Cabeça e Pescoço/metabolismo , Neoplasias de Cabeça e Pescoço/terapia , Humanos , Fenótipo , Radioterapia , Espécies Reativas de Oxigênio/metabolismo
4.
Neuro Oncol ; 22(1): 139-151, 2020 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-31398252

RESUMO

BACKGROUND: Despite increased understanding of the genetic events underlying pediatric high-grade gliomas (pHGGs), therapeutic progress is static, with poor understanding of nongenomic drivers. We therefore investigated the role of alterations in mitochondrial function and developed an effective combination therapy against pHGGs. METHODS: Mitochondrial DNA (mtDNA) copy number was measured in a cohort of 60 pHGGs. The implication of mtDNA alteration in pHGG tumorigenesis was studied and followed by an efficacy investigation using patient-derived cultures and orthotopic xenografts. RESULTS: Average mtDNA content was significantly lower in tumors versus normal brains. Decreasing mtDNA copy number in normal human astrocytes led to a markedly increased tumorigenicity in vivo. Depletion of mtDNA in pHGG cells promoted cell migration and invasion and therapeutic resistance. Shifting glucose metabolism from glycolysis to mitochondrial oxidation with the adenosine monophosphate-activated protein kinase activator AICAR (5-aminoimidazole-4-carboxamide ribonucleotide) or the pyruvate dehydrogenase kinase inhibitor dichloroacetate (DCA) significantly inhibited pHGG viability. Using DCA to shift glucose metabolism to mitochondrial oxidation and then metformin to simultaneously target mitochondrial function disrupted energy homeostasis of tumor cells, increasing DNA damage and apoptosis. The triple combination with radiation therapy, DCA and metformin led to a more potent therapeutic effect in vitro and in vivo. CONCLUSIONS: Our results suggest metabolic alterations as an onco-requisite factor of pHGG tumorigenesis. Targeting reduced mtDNA quantity represents a promising therapeutic strategy for pHGG.


Assuntos
Aminoimidazol Carboxamida/análogos & derivados , Neoplasias Encefálicas/metabolismo , DNA Mitocondrial/metabolismo , Ácido Dicloroacético/farmacologia , Metabolismo Energético/fisiologia , Glioma/metabolismo , Ribonucleotídeos/farmacologia , Aminoimidazol Carboxamida/farmacologia , Animais , Neoplasias Encefálicas/genética , Criança , DNA Mitocondrial/efeitos dos fármacos , DNA Mitocondrial/efeitos da radiação , Metabolismo Energético/efeitos dos fármacos , Metabolismo Energético/efeitos da radiação , Dosagem de Genes , Glioma/genética , Glicólise/efeitos dos fármacos , Glicólise/efeitos da radiação , Humanos , Camundongos , Ensaios Antitumorais Modelo de Xenoenxerto
5.
J Biochem ; 167(3): 303-314, 2020 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-31670806

RESUMO

Interleukin-6 (IL-6)-induced glycolysis and therapeutic resistance is reported in some cell systems; however, the mechanism of IL-6-induced glycolysis in radio-resistance is unexplored. Therefore, to investigate, we treated Raw264.7 cells with IL-6 (1 h prior to irradiation) and examined the glycolytic flux. Increased expression of mRNA and protein levels of key glycolytic enzymes was observed after IL-6 treatment, which conferred glycolysis dependent resistance from radiation-induced cell death. We further established that IL-6-induced glycolysis is activated by Akt signalling and knocking down Akt or inhibition of pan Akt phosphorylation significantly abrogated the IL-6-induced radio-resistance. Moreover, reduction of IL-6-induced pAkt level suppressed the expression of Hexokinase-2 and its translocation to the mitochondria, thereby inhibiting the glycolysis-induced resistance to radiation. IL-6-induced glycolysis also minimized the radiation-induced mitochondrial damage. These results suggest that IL-6-induced glycolysis observed in cells may be responsible for IL-6-mediated therapeutic radio-resistance in cancer cells, partly by activation of Akt signalling.


Assuntos
Glicólise/efeitos dos fármacos , Interleucina-6/farmacologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Tolerância a Radiação/efeitos dos fármacos , Animais , Morte Celular/efeitos dos fármacos , Morte Celular/genética , Morte Celular/efeitos da radiação , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Proliferação de Células/efeitos da radiação , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Sobrevivência Celular/efeitos da radiação , Técnicas de Silenciamento de Genes , Glicólise/genética , Glicólise/efeitos da radiação , Hexoquinase/genética , Hexoquinase/metabolismo , Camundongos , Mitocôndrias/patologia , Mitocôndrias/efeitos da radiação , Fosforilação , Proteínas Proto-Oncogênicas c-akt/genética , Células RAW 264.7 , RNA Interferente Pequeno , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Transdução de Sinais/efeitos da radiação
6.
Ann Nucl Med ; 34(2): 128-135, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31768818

RESUMO

OBJECTIVES: In patients with advanced ovarian, fallopian and primary peritoneal carcinoma, complete interval debulking surgery (IDS) is often performed after neoadjuvant chemotherapy (NAC) to achieve long progression-free survival (PFS) and overall survival (OS). We aimed to investigate the utility of 2-deoxy-2-[F-18]fluoro-D-glucose (FDG) PET/CT in patients with these malignancies who underwent complete IDS. METHODS: Between 2009 and 2017, twenty-two patients underwent FDG PET/CT scans before and after NAC. The highest SUVmax/peak (standardized uptake value), metabolic tumor volume (MTV) and total lesion glycolysis (TLG) for whole lesions were defined as target SUVmax/peak, tMTV and tTLG, respectively. We also calculated these reduction rates during NAC. These parameters were compared between the groups with platinum-free interval (PFI) > 12 months (n = 10) and those with PFI ≤ 12 months (n = 12). The PFS and OS were evaluated using these quantitative parameters, and in terms of the presence of visually detectable residual lesions after NAC. RESULTS: The target SUVmax/peak before NAC, the reduction rates in the target SUVmax, tMTV and tTLG were significantly higher in the group with PFI > 12 months than the shorter PFI group (p < 0.05). Especially in PFS, the higher reduction rates in the target SUVmax/peak, tMTV, and tTLG had an excellent prognostic stratification (p < 0.05) and the FDG visually negative group after NAC had a significantly better prognosis than the other group (p < 0.01). CONCLUSIONS: The reduction rate of FDG PET-based quantitative values and visual analysis after NAC demonstrated prognostic potential, especially in PFS.


Assuntos
Fluordesoxiglucose F18/química , Terapia Neoadjuvante/métodos , Neoplasias Ovarianas/radioterapia , Neoplasias Peritoneais/radioterapia , Tomografia por Emissão de Pósitrons/métodos , Idoso , Feminino , Glicólise/efeitos da radiação , Humanos , Pessoa de Meia-Idade , Imagem Multimodal/métodos , Neoplasias Ovarianas/cirurgia , Neoplasias Peritoneais/cirurgia , Prognóstico , Compostos Radiofarmacêuticos/química , Estudos Retrospectivos , Fatores de Tempo , Resultado do Tratamento , Carga Tumoral/efeitos da radiação
7.
Lung Cancer ; 126: 32-40, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30527190

RESUMO

PURPOSE: Metabolic inhibition might sensitize tumors to irradiation. Here, we examined the effect of lonidamine (several metabolic effects, inhibiting hexokinase amongst others) and/or 968 (glutaminase inhibitor) on tumor cell metabolism, cell growth, cytotoxicity and radiosensitivity in NSCLC cell lines in vitro in relation to histology. MATERIALS AND METHODS: Adeno- (H23, HCC827, H1975) and squamous cell carcinoma (H520, H292, SW900) NSCLC cells were treated with lonidamine and/or 968 for 72 h under physiological levels of glucose (1.5 mM). Cells were irradiated with 0, 4 or 8 Gy. Cell growth of H2B-mCherry transduced cells and cytotoxicity (CellTox™ Green Cytotoxicity Assay) were measured using live cell imaging (IncuCyte). Inhibitory effects on metabolic profiles was determined using the Seahorse XF96 extracellular Flux analyzer. RESULTS: NSCLC cell lines responded differently to glycolysis (lonidamine) and/or glutaminase (968) inhibition, largely corresponding with changes in glycolytic and mitochondrial metabolism upon treatment. Response patterns were not related to histology. 968 was cytotoxic in cell lines with high glutaminase C expression (H1975 and H520), whereas combination treatment was cytotoxic in KRAS mutated cell lines SW900 and H23. H292 and HCC827 were resistant to combination treatment. Treatment with 968 and especially lonidamine resulted in radiosensitization of H292 and HCC827 in terms of decreased relative cell growth and increased cytotoxicity. CONCLUSION: NSCLC is a heterogeneous disease, which is reflected in the response of different cell lines to the treatment (combinations) reported here. Only a part of NSCLC patients may benefit from the combination of radiation therapy and metabolic inhibition, making stratification necessary.


Assuntos
Inibidores Enzimáticos/farmacologia , Glucose/metabolismo , Glutamina/metabolismo , Indazóis/farmacologia , Tolerância a Radiação/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Relação Dose-Resposta à Radiação , Glutaminase/antagonistas & inibidores , Glutaminase/metabolismo , Glicólise/efeitos dos fármacos , Glicólise/efeitos da radiação , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Raios X
8.
Comp Med ; 68(4): 269-279, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29875029

RESUMO

Liver cancer is the second leading cause of cancer death worldwide. Metabolic pathways within the liver and liver cancers are highly regulated by the central circadian clock in the suprachiasmatic nuclei (SCN). Daily light and dark cycles regulate the SCN-driven pineal production of the circadian anticancer hormone melatonin and temporally coordinate circadian rhythms of metabolism and physiology in mammals. In previous studies, we demonstrated that melatonin suppresses linoleic acid metabolism and the Warburg effect (aerobic glycolysis)in human breast cancer xenografts and that blue-enriched light (465-485 nm) from light-emitting diode lighting at daytime (bLAD) amplifies nighttime circadian melatonin levels in rats by 7-fold over cool white fluorescent (CWF) lighting. Here we tested the hypothesis that daytime exposure of tissue-isolated Morris hepatoma 7288CTC-bearing male rats to bLAD amplifies the nighttime melatonin signal to enhance the inhibition of tumor growth. Compared with rats housed under a 12:12-h light:dark cycle in CWF light, rats in bLAD light evinced a 7-fold higher peak plasma melatonin level at the mid-dark phase; in addition, high melatonin levels were prolonged until 4 h into the light phase. After implantation of tissue-isolated hepatoma 7288CTC xenografts, tumor growth rates were markedly delayed, and tumor cAMP levels, LA metabolism, the Warburg effect, and growth signaling activities were decreased in rats in bLAD compared with CWF daytime lighting. These data show that the increased nighttime circadian melatonin levels due to bLAD exposure decreases hepatoma metabolic, signaling, and proliferative activities beyond what occurs after normal melatonin signaling under CWF light.


Assuntos
Carcinoma Hepatocelular/patologia , Ritmo Circadiano/efeitos da radiação , Progressão da Doença , Neoplasias Hepáticas Experimentais/patologia , Melatonina/sangue , Fotoperíodo , Animais , Carcinoma Hepatocelular/metabolismo , Glicólise/efeitos da radiação , Xenoenxertos , Luz , Neoplasias Hepáticas Experimentais/metabolismo , Masculino , Ratos , Transdução de Sinais
9.
Nucl Med Commun ; 39(9): 845-852, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29901488

RESUMO

PURPOSE: This study aims to evaluate the prognostic role of the decrease in total lesion glycolysis (TLG) assessed by fluorine-18-fluorodeoxyglucose (F-FDG) PET-computed tomography (CT) 1 month after yttrium-90-radioembolization (Y-RE) in patients affected by hepatocellular carcinoma (HCC) with portal vein tumour thrombosis (PVTT). PATIENTS AND METHODS: Twenty-one patients with histologically proven poorly differentiated HCC and evidence of PVTT at the enhanced multislice CT underwent Y-RE. All patients underwent an F-FDG PET-CT scan at baseline and 1 month after the loco-regional therapy. The variation in TLG (ΔTLG) between the two studies was calculated. Patients were divided in two groups (group 1: 1 month ΔTLG >50%, group 2: ΔTLG <50%). Statistical analysis was carried out to assess the prognostic role of TLG in overall survival (OS). RESULTS: On the 21 patients enrolled, all presented a decrease in TLG after the administration of Y-microspheres. The OS was 11.5±1.2 months. Nine out of 21 (42.9%) patients showed ΔTLG more than 50% with a mean OS of 16.8±1.3 months, whereas the remaining 12 (57.1%) patients had ΔTLG less than 50% with a mean OS of 7.5±0.5 months. Statistical analysis showed ΔTLG to be a significant (P<0.001) predictor of survival. None of the other examined variables including age, Child-Pugh classification, previously performed therapies, the presence of extrahepatic metastases, α-fetoprotein and bilirubin levels had a significant prognostic impact on patients' outcome. CONCLUSION: Decrease in TLG measured by F-FDG PET-CT is correlated with a trend towards a longer median survival in patients affected by HCC and PVTT who have undergone Y-RE.


Assuntos
Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/radioterapia , Embolização Terapêutica , Glicólise/efeitos da radiação , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/radioterapia , Trombose/complicações , Idoso , Carcinoma Hepatocelular/complicações , Carcinoma Hepatocelular/diagnóstico por imagem , Feminino , Humanos , Neoplasias Hepáticas/complicações , Neoplasias Hepáticas/diagnóstico por imagem , Masculino , Pessoa de Meia-Idade , Veia Porta , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Estudos Retrospectivos , Análise de Sobrevida , Radioisótopos de Ítrio/uso terapêutico
10.
Nucl Med Commun ; 39(6): 553-563, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29668513

RESUMO

PURPOSE: The aim of this study was to evaluate the prognostic importance metabolic tumor volume (MTV), total lesion glycolysis (TLG), and standardized uptake value (SUV) in patients with esophageal cancer treated with definitive chemoradiotherapy. PATIENTS AND METHODS: Seventy-two esophageal cancer patients treated with definitive chemoradiotherapy [57 (79%) patients] or definitive radiotherapy [15 (21%) patients] were retrospectively analyzed. The regions equal to or greater than SUV of 2.5 were selected to delineate MTV and TLG was calculated by multiplying the mean SUV by the MTV of the primary lesions. The overall survival (OS) and disease-free survival (DFS) were evaluated for all patients and also patients with squamous cell carcinoma. RESULTS: The median survival time was 13.4 months (range: 1.8-119.3 months) for all patients. Maximum SUV, mean SUV, MTV, and TLG values were significantly higher in patients with extensive T-stage (T3-T4) compared with patients with T1-T2 disease. Patients with regional lymph node metastasis had significantly higher MTV and TLG values compared with patients with no lymph node metastasis. On multivariate analysis, MTV, TLG, presence of lymph node metastasis, and lack of concurrent chemotherapy were negative significant prognostic factors for OS and DFS for the entire cohort and for patients with squamous cell carcinoma esophageal cancer. CONCLUSION: Metabolic volumes (MTV and TLG), regional lymph node metastasis, and concurrent chemotherapy are major prognostic factors for DFS and OS in patients with esophageal carcinoma. In addition, MTV and TLG are important in predicting nodal metastasis, and together with metabolic volumes, SUV are associated significantly with local tumor invasion.


Assuntos
Quimiorradioterapia , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patologia , Glicólise , Tomografia por Emissão de Pósitrons , Carga Tumoral , Adulto , Idoso , Idoso de 80 Anos ou mais , Transporte Biológico , Neoplasias Esofágicas/diagnóstico , Neoplasias Esofágicas/terapia , Feminino , Glicólise/efeitos dos fármacos , Glicólise/efeitos da radiação , Humanos , Masculino , Pessoa de Meia-Idade , Prognóstico , Estudos Retrospectivos , Carga Tumoral/efeitos dos fármacos , Carga Tumoral/efeitos da radiação
11.
Exp Dermatol ; 27(9): 941-949, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29658146

RESUMO

Ultraviolet (UV) radiation has a plethora of effects on human tissues. In the UV spectrum, wavelengths above 320 nm fall into the UVA range, and for these, it has been shown that they induce reactive oxygen species (ROS), DNA mutations and are capable to induce melanoma in mice. In addition to this, it was recently shown that UVA irradiation and UVA-induced ROS also increase glucose metabolism of melanoma cells. UVA irradiation causes a persistent increase in glucose consumption, accompanied by increased glycolysis, increased lactic acid production and activation of the pentose phosphate pathway. Furthermore, it was shown that the enhanced secretion of lactic acid is important for invasion of melanoma in vitro. The current knowledge of this link between UVA, metabolism and melanoma, possible mechanisms of UVA-induced glucose metabolism and their starting points are discussed in this review with focus on ROS- and UVA-induced cellular stress signalling, DNA damage signalling and DNA repair systems. When looking at the benefits of UVA-induced glucose metabolism, it becomes apparent that there are more advantages of these metabolic changes than one would expect. Besides the role of lactic acid as initiator of protease expression and invasion, its role for immune escape of melanoma cells and the pentose phosphate pathway-derived nicotinamide adenine dinucleotide phosphate (NADPH) as part of a ROS detoxification strategy are discussed.


Assuntos
Glucose/metabolismo , Melanoma/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Neoplasias Cutâneas/metabolismo , Pele/efeitos da radiação , Raios Ultravioleta/efeitos adversos , Animais , Sobrevivência Celular/efeitos da radiação , Dano ao DNA , Glicólise/efeitos da radiação , Humanos , Ácido Láctico/metabolismo , Melanoma/imunologia , Melanoma/patologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NADP/metabolismo , Invasividade Neoplásica , Via de Pentose Fosfato , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ácido Pirúvico/metabolismo , Pele/metabolismo , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/patologia , Evasão Tumoral
12.
Anticancer Res ; 38(3): 1291-1301, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29491052

RESUMO

BACKGROUND/AIM: Laser photochemotherapy is a new approach in cancer treatment using low-level laser therapy (LLLT) to enhance the effect of chemotherapy. MATERIALS AND METHODS: In order to evaluate the effect of LLLT on tumor cells, HeLa cells were treated with cisplatin or zoledronic acid (ZA) followed by LLLT. Cell viability was evaluated with 2,3-bis-(2-methoxy-4-nitro-5-sulfophenyl)-2H-tetrazolium-5-carboxanilide assay. Oxidative phosphorylation and glycolysis were measured using extracellular flux analysis. Immunocytochemistry of heat-shock protein 70 (HSP70) and western blot analysis were performed. RESULTS: LLLT alone increased viability and was associated with lower oxidative phosphorylation but higher glycolysis rates. Cisplatin and ZA alone lowered cell viability, glycolysis and oxidative phosphorylation. This effect was significantly enhanced in conjunction with LLLT and was accompanied by reduced oxidative phosphorylation and collapse of glycolysis. CONCLUSION: Our observations indicate that LLLT may raise the cytotoxicity of cisplatin and ZA by modulating cellular metabolism, pointing to a possible application in cancer treatment.


Assuntos
Cisplatino/farmacologia , Difosfonatos/farmacologia , Imidazóis/farmacologia , Lasers , Western Blotting , Conservadores da Densidade Óssea/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Glicólise/efeitos dos fármacos , Glicólise/efeitos da radiação , Proteínas de Choque Térmico HSP70/metabolismo , Células HeLa , Humanos , Terapia com Luz de Baixa Intensidade/métodos , Fosforilação Oxidativa/efeitos dos fármacos , Fosforilação Oxidativa/efeitos da radiação , Fotoquimioterapia/métodos , Radiossensibilizantes/farmacologia , Ácido Zoledrônico
13.
Chemosphere ; 200: 302-312, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29494911

RESUMO

Proton is a major particle of space radiation environment and a prospective radiotherapy beam. However, its risk needs to be fully evaluated for the understanding and to establish the better protective strategy for astronaut and patient. Zebrafish is an ideal model for the toxicity studies on medicines and environmental genetic toxicants. In the current study, embryos of zebrafish at 24 h post-fertilization (hpf) were exposed to proton beam. Some toxic parameters of embryo-larval development were investigated. Microarray combining with qRT-PCR were used to detect the gene expression situation. Generally, fractions of a variety of abnormal phenotypes of embryos and larvae increased in a dose-dependent manner after irradiation. The copy number of mitochondria, the basal respiration rate and the maximum respiration rate of embryos significantly decreased after irradiation. Microarray data demonstrated that MAPK signaling pathway, cell communication, glycolysis and TGF-ß signaling pathway were significantly affected in the irradiated group. The expressions of matrix metallopeptidase 9 (mmp9) and TIMP metallopeptidase inhibitor 2b (timp2b) genes, and enzymatic activity of MMP9 were significantly upregulated in irradiated group. Overall, these results suggest that acute radiation of proton severely affects the development of organism and results in aberration occurrence in the early stage of zebrafish development, which may relates to mitochondrial and glycolytic dysfunction.


Assuntos
Embrião não Mamífero/citologia , Glicólise/efeitos da radiação , Larva/citologia , Mitocôndrias/efeitos da radiação , Prótons/efeitos adversos , Peixe-Zebra/embriologia , Animais , Embrião não Mamífero/efeitos da radiação , Larva/efeitos da radiação , Peixe-Zebra/metabolismo
14.
Oncol Res ; 26(4): 547-556, 2018 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-28800787

RESUMO

MicroRNA dysregulation contributes to malignant progression, dissemination, and profound treatment resistance in multiple cancers. miR-449a is recognized as a tumor suppresser. However, the roles of miR-449a in lung cancer initiation and progression are largely unknown. Our study aims to investigate the roles and underlying mechanism of miR-449a in modulating sensitivity to ionizing radiation (IR) in non-small cell lung cancer (NSCLC). Lung cancer cells were transfected with miR-449a mimics or negative control and exposed to IR; the levels of target protein, glycolysis, cell viability, apoptosis, and DNA damage were examined. miR-449a was suppressed in lung cancer tissues and cancer cells (A549 and H1299). IR exposure significantly increased the expression of miR-449a in A549 cells at doses ranging from 4 to 8 Gy at 24 h, whereas no significant change in miR-449a was found in H1299 cells after IR. When A549 cells were exposed to IR at a dose of 8 Gy, the miR-449a level only had an acute increase within 12 h. miR-449a restoration dramatically suppressed IR-induced cell apoptosis and DNA damage in both A549 and H1299 cells. Bioinformatics analysis indicated that lactate dehydrogenase A (LDHA) was a potential target of miR-449a. miR-449a mimic transfection substantially suppressed the LDHA expression and production of lactate catalyzed by LDHA as well as glucose uptake. We confirmed that miR-449a could bind to the 3'-UTR of LDHA mRNA using luciferase reporter assay. LDHA siRNA-transfected cells showed enhanced cell apoptosis and DNA damage in response to IR exposure. miR-449a upregulation enhanced IR sensitivity of lung cancer cells by suppressing LDHA and the subsequent glycolysis.


Assuntos
Apoptose/genética , Carcinoma Pulmonar de Células não Pequenas/genética , Glicólise/genética , L-Lactato Desidrogenase/genética , Neoplasias Pulmonares/genética , MicroRNAs/genética , Radiação Ionizante , Regiões 3' não Traduzidas/genética , Células A549 , Apoptose/efeitos da radiação , Carcinoma Pulmonar de Células não Pequenas/patologia , Dano ao DNA , Glicólise/efeitos da radiação , Humanos , Isoenzimas/genética , Lactato Desidrogenase 5 , Neoplasias Pulmonares/patologia
15.
Clin Cancer Res ; 23(19): 5881-5891, 2017 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-28720669

RESUMO

Purpose:MUC1, an oncogene overexpressed in multiple solid tumors, including pancreatic cancer, reduces overall survival and imparts resistance to radiation and chemotherapies. We previously identified that MUC1 facilitates growth-promoting metabolic alterations in pancreatic cancer cells. The present study investigates the role of MUC1-mediated metabolism in radiation resistance of pancreatic cancer by utilizing cell lines and in vivo models.Experimental Design: We used MUC1-knockdown and -overexpressed cell line models for evaluating the role of MUC1-mediated metabolism in radiation resistance through in vitro cytotoxicity, clonogenicity, DNA damage response, and metabolomic evaluations. We also investigated whether inhibition of glycolysis could revert MUC1-mediated metabolic alterations and radiation resistance by using in vitro and in vivo models.Results: MUC1 expression diminished radiation-induced cytotoxicity and DNA damage in pancreatic cancer cells by enhancing glycolysis, pentose phosphate pathway, and nucleotide biosynthesis. Such metabolic reprogramming resulted in high nucleotide pools and radiation resistance in in vitro models. Pretreatment with the glycolysis inhibitor 3-bromopyruvate abrogated MUC1-mediated radiation resistance both in vitro and in vivo, by reducing glucose flux into nucleotide biosynthetic pathways and enhancing DNA damage, which could again be reversed by pretreatment with nucleoside pools.Conclusions: MUC1-mediated nucleotide metabolism plays a key role in facilitating radiation resistance in pancreatic cancer and targeted effectively through glycolytic inhibition. Clin Cancer Res; 23(19); 5881-91. ©2017 AACR.


Assuntos
Dano ao DNA/efeitos da radiação , Mucina-1/genética , Neoplasias Pancreáticas/radioterapia , Tolerância a Radiação/genética , Animais , Linhagem Celular Tumoral , Proliferação de Células/genética , Proliferação de Células/efeitos da radiação , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Técnicas de Silenciamento de Genes , Glucose/metabolismo , Glicólise/efeitos da radiação , Humanos , Camundongos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Transdução de Sinais/efeitos da radiação , Ensaios Antitumorais Modelo de Xenoenxerto
16.
PLoS One ; 12(4): e0176243, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28448534

RESUMO

A wide search for ischemic preconditioning (IPC) mechanisms of cardioprotection identified the light elicited circadian rhythm protein Period 2 (Per2) to be cardioprotective. Studies on cardiac metabolism found a key role for light elicited Per2 in mediating metabolic dependence on carbohydrate metabolism. To profile Per2 mediated pathways following IPC of the mouse heart, we performed a genome array and identified 352 abundantly expressed and well-characterized Per2 dependent micro RNAs. One prominent result of our in silico analysis for cardiac Per2 dependent micro RNAs revealed a selective role for miR-21 in the regulation of hypoxia and metabolic pathways. Based on this Per2 dependency, we subsequently found a diurnal expression pattern for miR-21 with higher miR-21 expression levels at Zeitgeber time (ZT) 15 compared to ZT3. Gain or loss of function studies for miR-21 using miRNA mimics or miRNA inhibitors and a Seahorse Bioanalyzer uncovered a critical role of miR-21 for cellular glycolysis, glycolytic capacity, and glycolytic reserve. Exposing mice to intense light, a strategy to induce Per2, led to a robust induction of cardiac miR-21 tissue levels and decreased infarct sizes, which was abolished in miR-21-/- mice. Similarly, first translational studies in humans using intense blue light exposure for 5 days in healthy volunteers resulted in increased plasma miR-21 levels which was associated with increased phosphofructokinase activity, the rate-limiting enzyme in glycolysis. Together, we identified miR-21 as cardioprotective downstream target of Per2 and suggest intense light therapy as a potential strategy to enhance miR-21 activity and subsequent carbohydrate metabolism in humans.


Assuntos
Glicólise/efeitos da radiação , Coração/efeitos da radiação , Luz , MicroRNAs/genética , Miocárdio/metabolismo , Proteínas Circadianas Period/metabolismo , Regulação para Cima/efeitos da radiação , Adulto , Animais , Células Endoteliais/metabolismo , Células Endoteliais/efeitos da radiação , Feminino , Humanos , Precondicionamento Isquêmico , Pulmão/metabolismo , Pulmão/efeitos da radiação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/genética , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Miocárdio/patologia , Proteínas Circadianas Period/deficiência , Fosfofrutoquinases/metabolismo , Adulto Jovem
17.
Int J Oncol ; 50(5): 1531-1540, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28339028

RESUMO

HIF-1α overexpression is associated with radio-resistance of various cancers. A radioresistant human melanoma cell model MDA-MB-435R (435R) was established by us previously. Compared with the parental cells MDA-MB­435 (435S), an elevated level of HIF-1α expression in 435R cells was demonstrated in our recent experiments. Therefore, in the current study, we sought to determine whether selective HIF-1α inhibitors could radiosensitize the 435R cells to X-ray, and to identify the potential mechanisms. Our data demonstrated that inhibition of HIF-1α with 2-methoxyestradiol (2-MeOE2) significantly enhanced radiosensitivity of 435R cells. 2-MeOE2 increased DNA damage and ratio of apoptosis cells induced by irradiation. Whereas, cell proliferation and the expression of pyruvate dehydrogenase kinase 1 (PDK1) were decreased after 2-MeOE2 treatment. The change of expression of GLUT1, LDHA and the cellular ATP level and extracellular lactate production indicates that 2-MeOE2 suppressed glycolytic state of 435R cells. In addition, the radioresistance, glycolytic state and cell proliferation of 435R cells were also decreased after inhibiting pyruvate dehydrogenase kinase 1 (PDK1) with dichloroacetate (DCA). DCA could also increase DNA damage and ratio of apoptotic cells induced by irradiation. These results also suggest that inhibition of HIF-1α with 2-MeOE2 sensitizes radioresistant melanoma cells 435R to X-ray irradiation through targeting the glycolysis that is regulated by PDK1. Selective inhibitors of HIF-1α and glycolysis are potential drugs to enhance radio-sensitivity of melanoma cells.


Assuntos
Estradiol/análogos & derivados , Subunidade alfa do Fator 1 Induzível por Hipóxia/biossíntese , Melanoma/tratamento farmacológico , Melanoma/radioterapia , Proteínas Serina-Treonina Quinases/biossíntese , 2-Metoxiestradiol , Apoptose/efeitos dos fármacos , Hipóxia Celular/efeitos dos fármacos , Hipóxia Celular/efeitos da radiação , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Dano ao DNA/efeitos da radiação , Estradiol/administração & dosagem , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glicólise/efeitos dos fármacos , Glicólise/efeitos da radiação , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Melanoma/patologia , Proteínas Serina-Treonina Quinases/genética , Piruvato Desidrogenase Quinase de Transferência de Acetil , Tolerância a Radiação/efeitos dos fármacos , Raios X
18.
Radiat Res ; 187(4): 441-453, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28170315

RESUMO

The unique cellular and molecular consequences of cytoplasmic damage caused by ionizing radiation were studied using a precision microbeam irradiator. Our results indicated that targeted cytoplasmic irradiation induced metabolic shift from an oxidative to glycolytic phenotype in human small airway epithelial cells (SAE). At 24 h postirradiation, there was an increase in the mRNA expression level of key glycolytic enzymes as well as lactate secretion in SAE cells. Using RNA-sequencing analysis to compare genes that were responsive to cytoplasmic versus nuclear irradiation, we found a glycolysis related gene, Pim-1, was significantly upregulated only in cytoplasmic irradiated SAE cells. Inhibition of Pim-1 activity using the selective pharmaceutic inhibitor Smi-4a significantly reduced the level of lactate production and glucose uptake after cytoplasmic irradiation. In addition, Pim-1 also inhibited AMPK activity, which is a well-characterized negative regulator of glycolysis. Distinct from the glycolysis induced by cytoplasmic irradiation, targeted nuclear irradiation also induced a transient and minimal increase in glycolysis that correlated with increased expression of Hif-1α. In an effort to explore the underline mechanism, we found that inhibition of mitochondria fission using the cell-permeable inhibitor mdivi-1 suppressed the induction of Pim-1, thus confirming Pim-1 upregulation as a downstream effect of mitochondrial dysfunction. Our data show and, for the first time, that cytoplasmic irradiation mediate expression level of Pim-1, which lead to glycolytic shift in SAE cells. Additionally, since glycolysis is frequently linked to cancer cell metabolism, our findings further suggest a role of cytoplasmic damage in promoting neoplastic changes.


Assuntos
Citoplasma/efeitos da radiação , Células Epiteliais/metabolismo , Células Epiteliais/efeitos da radiação , Proteínas Proto-Oncogênicas c-pim-1/metabolismo , Mucosa Respiratória/metabolismo , Mucosa Respiratória/efeitos da radiação , Proteínas Quinases Ativadas por AMP/antagonistas & inibidores , Proteínas Quinases Ativadas por AMP/metabolismo , Sequência de Bases/efeitos da radiação , Linhagem Celular , Citoplasma/enzimologia , Citoplasma/metabolismo , Ativação Enzimática/efeitos da radiação , Células Epiteliais/enzimologia , Glicólise/efeitos da radiação , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , RNA/genética , RNA/efeitos da radiação , Mucosa Respiratória/citologia , Mucosa Respiratória/enzimologia , Regulação para Cima/efeitos da radiação
19.
Biochim Biophys Acta Proteins Proteom ; 1865(4): 404-413, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28087425

RESUMO

Lonicera japonica Thunb., also known as Jin Yin Hua and Japanese honeysuckle, is used as a herbal medicine in Asian countries. Its flowers have been used in folk medicine in the clinic and in making food or healthy beverages for over 1500years in China. To investigate the molecular processes involved in L. japonica development from buds to flowers exposed to UV radiation, a comparative proteomics analysis was performed. Fifty-four proteins were identified as differentially expressed, including 42 that had increased expression and 12 that had decreased expression. The levels of the proteins related to glycolysis, TCA/organic acid transformation, major carbohydrate metabolism, oxidative pentose phosphate, stress, secondary metabolism, hormone, and mitochondrial electron transport were increased during flower opening process after exposure to UV radiation. Six metabolites in L. japonica buds and flowers were identified and relatively quantified using LC-MS/MS. The antioxidant activity was performed using a 1,1-diphenyl-2-picrylhydrazyl assay, which revealed that L. japonica buds had more activity than the UV irradiated flowers. This suggests that UV-B radiation induces production of endogenous ethylene in L. japonica buds, thus facilitating blossoming of the buds and activating the antioxidant system. Additionally, the higher metabolite contents and antioxidant properties of L. japonica buds indicate that the L. japonica bud stage may be a more optimal time to harvest than the flower stage when using for medicinal properties.


Assuntos
Flores/metabolismo , Lonicera/metabolismo , Metaboloma/efeitos da radiação , Proteoma/biossíntese , Raios Ultravioleta , Ciclo do Ácido Cítrico/efeitos da radiação , Glicólise/efeitos da radiação
20.
Sci Rep ; 6: 30986, 2016 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-27507219

RESUMO

The mechanism of Amifostine (WR-2721) mediated radioprotection is poorly understood. The effects of amifostine on human basal metabolism, mouse liver metabolism and on normal and tumor hepatic cells were studied. Indirect calorimetric canopy tests showed significant reductions in oxygen consumption and of carbon dioxide emission in cancer patients receiving amifostine. Glucose levels significantly decreased and lactate levels increased in patient venous blood. Although amifostine in vitro did not inhibit the activity of the prolyl-hydroxylase PHD2, experiments with mouse liver showed that on a short timescale WR-1065 induced expression of the Hypoxia Inducible Factor HIF1α, lactate dehydrogenase LDH5, glucose transporter GLUT2, phosphorylated pyruvate dehydrogenase pPDH and PDH-kinase. This effect was confirmed on normal mouse NCTC hepatocytes, but not on hepatoma cells. A sharp reduction of acetyl-CoA and ATP levels in NCTC cells indicated reduced mitochondrial usage of pyruvate. Transient changes of mitochondrial membrane potential and reactive oxygen species ROS production were evident. Amifostine selectively protects NCTC cells against radiation, whilst HepG2 neoplastic cells are sensitized. The radiation protection was correlates with HIF levels. These findings shed new light on the mechanism of amifostine cytoprotection and encourage clinical research with this agent for the treatment of primary and metastatic liver cancer.


Assuntos
Amifostina/farmacologia , Neoplasias da Mama/radioterapia , Protetores contra Radiação/farmacologia , Trifosfato de Adenosina/metabolismo , Animais , Metabolismo Basal/efeitos dos fármacos , Glicemia/metabolismo , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Feminino , Transportador de Glucose Tipo 2/metabolismo , Glicólise/efeitos dos fármacos , Glicólise/efeitos da radiação , Hepatócitos/efeitos dos fármacos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Potencial da Membrana Mitocondrial/efeitos da radiação , Camundongos Endogâmicos BALB C , Oxigênio/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Piruvato Desidrogenase Quinase de Transferência de Acetil
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA