Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 214
Filtrar
2.
Proc Natl Acad Sci U S A ; 117(36): 22378-22389, 2020 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-32839325

RESUMO

Hyperpolarized [1-13C]pyruvate magnetic resonance spectroscopic imaging (MRSI) is a noninvasive metabolic-imaging modality that probes carbon flux in tissues and infers the state of metabolic reprograming in tumors. Prevailing models attribute elevated hyperpolarized [1-13C]pyruvate-to-[1-13C]lactate conversion rates in aggressive tumors to enhanced glycolytic flux and lactate dehydrogenase A (LDHA) activity (Warburg effect). By contrast, we find by cross-sectional analysis using genetic and pharmacological tools in mechanistic studies applied to well-defined genetically engineered cell lines and tumors that initial hyperpolarized [1-13C]pyruvate-to-[1-13C]lactate conversion rates as well as global conversion were highly dependent on and critically rate-limited by the transmembrane influx of [1-13C]pyruvate mediated predominately by monocarboxylate transporter-1 (MCT1). Specifically, in a cell-encapsulated alginate bead model, induced short hairpin (shRNA) knockdown or overexpression of MCT1 quantitatively inhibited or enhanced, respectively, unidirectional pyruvate influxes and [1-13C]pyruvate-to-[1-13C]lactate conversion rates, independent of glycolysis or LDHA activity. Similarly, in tumor models in vivo, hyperpolarized [1-13C]pyruvate-to-[1-13C]lactate conversion was highly dependent on and critically rate-limited by the induced transmembrane influx of [1-13C]pyruvate mediated by MCT1. Thus, hyperpolarized [1-13C]pyruvate MRSI measures primarily MCT1-mediated [1-13C]pyruvate transmembrane influx in vivo, not glycolytic flux or LDHA activity, driving a reinterpretation of this maturing new technology during clinical translation. Indeed, Kaplan-Meier survival analysis for patients with pancreatic, renal, lung, and cervical cancers showed that high-level expression of MCT1 correlated with poor overall survival, and only in selected tumors, coincident with LDHA expression. Thus, hyperpolarized [1-13C]pyruvate MRSI provides a noninvasive functional assessment primarily of MCT1 as a clinical biomarker in relevant patient populations.


Assuntos
Isótopos de Carbono/metabolismo , Membrana Celular/metabolismo , Ácido Láctico/metabolismo , Transportadores de Ácidos Monocarboxílicos/metabolismo , Ácido Pirúvico/metabolismo , Simportadores/metabolismo , Animais , Isótopos de Carbono/análise , Isótopos de Carbono/química , Linhagem Celular Tumoral , Membrana Celular/química , Feminino , Humanos , Ácido Láctico/análise , Ácido Láctico/química , Imageamento por Ressonância Magnética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Ácido Pirúvico/análise , Ácido Pirúvico/química
3.
Proc Natl Acad Sci U S A ; 117(22): 12121-12130, 2020 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-32424096

RESUMO

HRAS, NRAS, and KRAS4A/KRAS4B comprise the RAS family of small GTPases that regulate signaling pathways controlling cell proliferation, differentiation, and survival. RAS pathway abnormalities cause developmental disorders and cancers. We found that KRAS4B colocalizes on the cell membrane with other RAS isoforms and a subset of prenylated small GTPase family members using a live-cell quantitative split luciferase complementation assay. RAS protein coclustering is mainly mediated by membrane association-facilitated interactions (MAFIs). Using the RAS-RBD (CRAF RAS binding domain) interaction as a model system, we showed that MAFI alone is not sufficient to induce RBD-mediated RAS inhibition. Surprisingly, we discovered that high-affinity membrane-targeted RAS binding proteins inhibit RAS activity and deplete RAS proteins through an autophagosome-lysosome-mediated degradation pathway. Our results provide a mechanism for regulating RAS activity and protein levels, a more detailed understanding of which should lead to therapeutic strategies for inhibiting and depleting oncogenic RAS proteins.


Assuntos
Autofagossomos/metabolismo , Membrana Celular/metabolismo , Lisossomos/metabolismo , Proteínas ras/metabolismo , Humanos , Modelos Moleculares , Domínios e Motivos de Interação entre Proteínas , Isoformas de Proteínas , Transdução de Sinais
4.
J Biol Chem ; 297(1): 100775, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34022218

RESUMO

Cellular pyruvate is an essential metabolite at the crossroads of glycolysis and oxidative phosphorylation, capable of supporting fermentative glycolysis by reduction to lactate mediated by lactate dehydrogenase (LDH) among other functions. Several inherited diseases of mitochondrial metabolism impact extracellular (plasma) pyruvate concentrations, and [1-13C]pyruvate infusion is used in isotope-labeled metabolic tracing studies, including hyperpolarized magnetic resonance spectroscopic imaging. However, how these extracellular pyruvate sources impact intracellular metabolism is not clear. Herein, we examined the effects of excess exogenous pyruvate on intracellular LDH activity, extracellular acidification rates (ECARs) as a measure of lactate production, and hyperpolarized [1-13C]pyruvate-to-[1-13C]lactate conversion rates across a panel of tumor and normal cells. Combined LDH activity and LDHB/LDHA expression analysis intimated various heterotetrameric isoforms comprising LDHA and LDHB in tumor cells, not only canonical LDHA. Millimolar concentrations of exogenous pyruvate induced substrate inhibition of LDH activity in both enzymatic assays ex vivo and in live cells, abrogated glycolytic ECAR, and inhibited hyperpolarized [1-13C]pyruvate-to-[1-13C]lactate conversion rates in cellulo. Of importance, the extent of exogenous pyruvate-induced inhibition of LDH and glycolytic ECAR in live cells was highly dependent on pyruvate influx, functionally mediated by monocarboxylate transporter-1 localized to the plasma membrane. These data provided evidence that highly concentrated bolus injections of pyruvate in vivo may transiently inhibit LDH activity in a tissue type- and monocarboxylate transporter-1-dependent manner. Maintaining plasma pyruvate at submillimolar concentrations could potentially minimize transient metabolic perturbations, improve pyruvate therapy, and enhance quantification of metabolic studies, including hyperpolarized [1-13C]pyruvate magnetic resonance spectroscopic imaging and stable isotope tracer experiments.


Assuntos
L-Lactato Desidrogenase/antagonistas & inibidores , Transportadores de Ácidos Monocarboxílicos/metabolismo , Ácido Pirúvico/farmacologia , Simportadores/metabolismo , Ácidos/metabolismo , Soluções Tampão , Isótopos de Carbono , Extratos Celulares , Linhagem Celular Tumoral , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Espaço Extracelular/química , Glicólise/efeitos dos fármacos , Humanos , Concentração Inibidora 50 , Cinética , L-Lactato Desidrogenase/metabolismo , Ácido Láctico/biossíntese , Especificidade por Substrato/efeitos dos fármacos
5.
Nature ; 523(7559): 177-82, 2015 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-26106858

RESUMO

Exosomes are lipid-bilayer-enclosed extracellular vesicles that contain proteins and nucleic acids. They are secreted by all cells and circulate in the blood. Specific detection and isolation of cancer-cell-derived exosomes in the circulation is currently lacking. Using mass spectrometry analyses, we identify a cell surface proteoglycan, glypican-1 (GPC1), specifically enriched on cancer-cell-derived exosomes. GPC1(+) circulating exosomes (crExos) were monitored and isolated using flow cytometry from the serum of patients and mice with cancer. GPC1(+) crExos were detected in the serum of patients with pancreatic cancer with absolute specificity and sensitivity, distinguishing healthy subjects and patients with a benign pancreatic disease from patients with early- and late-stage pancreatic cancer. Levels of GPC1(+) crExos correlate with tumour burden and the survival of pre- and post-surgical patients. GPC1(+) crExos from patients and from mice with spontaneous pancreatic tumours carry specific KRAS mutations, and reliably detect pancreatic intraepithelial lesions in mice despite negative signals by magnetic resonance imaging. GPC1(+) crExos may serve as a potential non-invasive diagnostic and screening tool to detect early stages of pancreatic cancer to facilitate possible curative surgical therapy.


Assuntos
Exossomos/metabolismo , Glipicanas , Neoplasias Pancreáticas/diagnóstico , Animais , Biomarcadores/sangue , Linhagem Celular Tumoral , Exossomos/genética , Feminino , Glipicanas/sangue , Glipicanas/metabolismo , Células HCT116 , Humanos , Células MCF-7 , Masculino , Camundongos , Células NIH 3T3 , Neoplasias Pancreáticas/sangue , Neoplasias Pancreáticas/patologia , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas p21(ras) , Proteínas ras/metabolismo
6.
Cancer Immunol Immunother ; 69(8): 1519-1534, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32300858

RESUMO

Enhanced tumor glycolytic activity is a mechanism by which tumors induce an immunosuppressive environment to resist adoptive T cell therapy; therefore, methods of assessing intratumoral glycolytic activity are of considerable clinical interest. In this study, we characterized the relationships among tumor 18F-fluorodeoxyglucose (FDG) retention, tumor metabolic and immune phenotypes, and survival in patients with resected non-small cell lung cancer (NSCLC). We retrospectively analyzed tumor preoperative positron emission tomography (PET) 18F-FDG uptake in 59 resected NSCLCs and investigated correlations between PET parameters (SUVMax, SUVTotal, SUVMean, TLG), tumor expression of glycolysis- and immune-related genes, and tumor-associated immune cell densities that were quantified by immunohistochemistry. Tumor glycolysis-associated immune gene signatures were analyzed for associations with survival outcomes. We found that each 18F-FDG PET parameter was positively correlated with tumor expression of glycolysis-related genes. Elevated 18F-FDG SUVMax was more discriminatory of glycolysis-associated changes in tumor immune phenotypes than other 18F-FDG PET parameters. Increased SUVMax was associated with multiple immune factors characteristic of an immunosuppressive and poorly immune infiltrated tumor microenvironment, including elevated PD-L1 expression, reduced CD57+ cell density, and increased T cell exhaustion gene signature. Elevated SUVMax identified immune-related transcriptomic signatures that were associated with enhanced tumor glycolytic gene expression and poor clinical outcomes. Our results suggest that 18F-FDG SUVMax has potential value as a noninvasive, clinical indicator of tumor immunometabolic phenotypes in patients with resectable NSCLC and warrants investigation as a potential predictor of therapeutic response to immune-based treatment strategies.


Assuntos
Biomarcadores Tumorais/análise , Carcinoma Pulmonar de Células não Pequenas/imunologia , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Fluordesoxiglucose F18/metabolismo , Linfócitos do Interstício Tumoral/imunologia , Tomografia por Emissão de Pósitrons/métodos , Microambiente Tumoral/imunologia , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Carcinoma Pulmonar de Células não Pequenas/diagnóstico por imagem , Carcinoma Pulmonar de Células não Pequenas/cirurgia , Glicólise , Humanos , Neoplasias Pulmonares/diagnóstico por imagem , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/cirurgia , Prognóstico , Compostos Radiofarmacêuticos/metabolismo , Estudos Retrospectivos , Taxa de Sobrevida , Transcriptoma
7.
Nanomedicine ; 25: 102169, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32059873

RESUMO

Generation of durable tumor-specific immune response without isolation and expansion of dendritic cells or T cells ex vivo remains a challenge. In this study, we investigated the impact of nanoparticle-mediated photothermolysis in combination with checkpoint inhibition on the induction of systemic antitumor immunity. Photothermolysis based on near-infrared light-absorbing copper sulfide nanoparticles and 15-ns laser pulses combined with the immune checkpoint inhibitor anti-PD-1 antibody (αPD-1) increased tumor infiltration by antigen-presenting cells and CD8-positive T lymphocytes in the B16-OVA mouse model. Moreover, combined photothermolysis, polymeric conjugate of the Toll-like receptor 9 agonist CpG, and αPD-1 significantly prolonged mouse survival after re-inoculation of tumor cells at a distant site compared to individual treatments alone in the poorly immunogenic syngeneic ID8-ip1-Luc ovarian tumor model. Thus, photothermolysis is a promising interventional technique that synergizes with Toll-like receptor 9 agonists and immune checkpoint inhibitors to enhance the abscopal effect in tumors.


Assuntos
Melanoma Experimental/tratamento farmacológico , Terapia Fototérmica , Receptor de Morte Celular Programada 1/genética , Receptor Toll-Like 9/genética , Animais , Terapia Combinada , Modelos Animais de Doenças , Humanos , Inibidores de Checkpoint Imunológico/farmacologia , Imunidade Inata/efeitos dos fármacos , Imunoterapia/métodos , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Camundongos , Nanopartículas/química , Nanopartículas/uso terapêutico , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor de Morte Celular Programada 1/imunologia , Receptor Toll-Like 9/agonistas
8.
Int J Mol Sci ; 21(10)2020 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-32466260

RESUMO

While pancreatic cancer (PC) survival rates have recently shown modest improvement, the disease remains largely incurable. Early detection of pancreatic cancer may result in improved outcomes and therefore, methods for early detection of cancer, even premalignant lesions, may provide more favorable outcomes. Pancreatic intraepithelial neoplasias (PanINs) have been identified as premalignant precursor lesions to pancreatic cancer. However, conventional imaging methods used for screening high-risk populations do not have the sensitivity to detect PanINs. Here, we have employed hyperpolarized metabolic imaging in vivo and nuclear magnetic resonance (1H-NMR) metabolomics ex vivo to identify and understand metabolic changes, towards enabling detection of early PanINs and progression to advanced PanINs lesions that precede pancreatic cancer formation. Progression of disease from tissue containing predominantly low-grade PanINs to tissue with high-grade PanINs showed a decreasing alanine/lactate ratio from high-resolution NMR metabolomics ex vivo. Hyperpolarized magnetic resonance spectroscopy (HP-MRS) allows over 10,000-fold sensitivity enhancement relative to conventional magnetic resonance. Real-time HP-MRS was employed to measure non-invasively changes of alanine and lactate metabolites with disease progression and in control mice in vivo, following injection of hyperpolarized [1-13C] pyruvate. The alanine-to-lactate signal intensity ratio was found to decrease as the disease progressed from low-grade PanINs to high-grade PanINs. The biochemical changes of alanine transaminase (ALT) and lactate dehydrogenase (LDH) enzyme activity were assessed. These results demonstrate that there are significant alterations of ALT and LDH activities during the transformation from early to advanced PanINs lesions. Furthermore, we demonstrate that real-time conversion kinetic rate constants (kPA and kPL) can be used as metabolic imaging biomarkers of pancreatic premalignant lesions. Findings from this emerging HP-MRS technique can be translated to the clinic for detection of pancreatic premalignant lesion in high-risk populations.


Assuntos
Carcinoma in Situ/diagnóstico por imagem , Espectroscopia de Ressonância Magnética/métodos , Neoplasias Pancreáticas/diagnóstico por imagem , Alanina Transaminase/sangue , Animais , Isótopos de Carbono , Carcinoma in Situ/sangue , Carcinoma in Situ/genética , L-Lactato Desidrogenase/metabolismo , Espectroscopia de Ressonância Magnética/normas , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Pancreáticas/sangue , Neoplasias Pancreáticas/genética , Sensibilidade e Especificidade
9.
J Proteome Res ; 18(7): 2826-2834, 2019 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-31120258

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is a deadly cancer that progresses without any symptom, and oftentimes, it is detected at an advanced stage. The lack of prior symptoms and effective treatments have created a knowledge gap in the management of this lethal disease. This issue can be addressed by developing novel noninvasive imaging-based biomarkers in PDAC. We explored in vivo hyperpolarized (HP) 13C MRS of pyruvate to lactate conversion and ex vivo 1H NMR spectroscopy in a panel of well-annotated patient-derived PDAC xenograft (PDXs) model and investigated the correlation between aberrant glycolytic metabolism and aggressiveness of the tumor. Real-time metabolic imaging data demonstrate the immediate intracellular conversion of HP 13C pyruvate to lactate after intravenous injection interrogating upregulated lactate dehydrogenase (LDH) activity in aggressive PDXs. Total ex vivo lactate measurement by 1H NMR spectroscopy showed a direct correlation with in vivo dynamic pyruvate-to-lactate conversion and demonstrated the potential of dynamic metabolic flux as a biomarker of total lactate concentration and aggressiveness of the tumor. Furthermore, the metabolite concentrations were very distinct among all four tumor types analyzed in this study. Overexpression of LDH-A and hypoxia-inducible factor (HIF-1α) plays a significant role in the conversion kinetics of HP pyruvate-to-lactate in tumors. Collectively, these data identified aberrant metabolic characteristics of pancreatic cancer PDXs and could potentially delineate metabolic targets for therapeutic intervention. Metabolic imaging with HP pyruvate and NMR metabolomics may enable identification and classification of aggressive subtypes of patient-derived xenografts. Translation of this real-time metabolic technique to the clinic may have the potential to improve the management of patients at high risk of developing pancreatic diseases.


Assuntos
Biomarcadores Tumorais/metabolismo , Imageamento por Ressonância Magnética/métodos , Neoplasias Pancreáticas/diagnóstico , Animais , Carcinoma Ductal Pancreático , Glicólise , Xenoenxertos , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , L-Lactato Desidrogenase/metabolismo , Ácido Láctico/metabolismo , Espectroscopia de Ressonância Magnética/métodos , Neoplasias Pancreáticas/metabolismo , Ácido Pirúvico/metabolismo
10.
Genes Dev ; 25(13): 1426-38, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21724834

RESUMO

Cell cycle regulation in hematopoietic stem cells (HSCs) is tightly controlled during homeostasis and in response to extrinsic stress. p53, a well-known tumor suppressor and transducer of diverse stress signals, has been implicated in maintaining HSC quiescence and self-renewal. However, the mechanisms that control its activity in HSCs, and how p53 activity contributes to HSC cell cycle control, are poorly understood. Here, we use a genetically engineered mouse to show that p53 C-terminal modification is critical for controlling HSC abundance during homeostasis and HSC and progenitor proliferation after irradiation. Preventing p53 C-terminal modification renders mice exquisitely radiosensitive due to defects in HSC/progenitor proliferation, a critical determinant for restoring hematopoiesis after irradiation. We show that fine-tuning the expression levels of the cyclin-dependent kinase inhibitor p21, a p53 target gene, contributes significantly to p53-mediated effects on the hematopoietic system. These results have implications for understanding cell competition in response to stresses involved in stem cell transplantation, recovery from adverse hematologic effects of DNA-damaging cancer therapies, and development of radioprotection strategies.


Assuntos
Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/efeitos da radiação , Homeostase/genética , Tolerância a Radiação/genética , Proteína Supressora de Tumor p53/metabolismo , Animais , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p21/genética , Feminino , Raios gama , Dosagem de Genes , Regulação da Expressão Gênica , Técnicas de Introdução de Genes , Longevidade/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Proteínas Proto-Oncogênicas c-mdm2/genética , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteína Supressora de Tumor p53/genética
11.
Molecules ; 24(3)2019 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-30759785

RESUMO

Colorectal cancer is the third most commonly occurring cancer in men and the second most commonly occurring cancer in women worldwide. We have recently reported that curcuminoid complexes labelled with gallium-68 have demonstrated preferential uptake in HT29 colorectal cancer and K562 lymphoma cell lines compared to normal human lymphocytes. In the present study, we report a new gallium-68-labelled curcumin derivative (68Ga-DOTA-C21) and its initial validation as marker for early detection of colorectal cancer. The precursor and non-radioactive complexes were synthesized and deeply characterized by analytical methods then the curcuminoid was radiolabelled with gallium-68. The in vitro stability, cell uptake, internalization and efflux properties of the probe were studied in HT29 cells, and the in vivo targeting ability and biodistribution were investigated in mice bearing HT29 subcutaneous tumour model. 68Ga-DOTA-C21 exhibits decent stability (57 ± 3% after 120 min of incubation) in physiological media and a curcumin-mediated cellular accumulation in colorectal cancer cell line (121 ± 4 KBq of radiotracer per mg of protein within 60 min of incubation). In HT29 tumour-bearing mice, the tumour uptake of 68Ga-DOTA-C21 is 3.57 ± 0.3% of the injected dose per gram of tissue after 90 min post injection with a tumour to muscle ratio of 2.2 ± 0.2. High amount of activity (12.73 ± 1.9% ID/g) is recorded in blood and significant uptake of the radiotracer occurs in the intestine (13.56 ± 3.3% ID/g), lungs (8.42 ± 0.8% ID/g), liver (5.81 ± 0.5% ID/g) and heart (4.70 ± 0.4% ID/g). Further studies are needed to understand the mechanism of accumulation and clearance; however, 68Ga-DOTA-C21 provides a productive base-structure to develop further radiotracers for imaging of colorectal cancer.


Assuntos
Neoplasias Colorretais/radioterapia , Curcumina/química , Curcumina/farmacologia , Radioisótopos de Gálio/química , Radioisótopos de Gálio/farmacologia , Compostos Heterocíclicos com 1 Anel/química , Animais , Linhagem Celular Tumoral , Neoplasias Colorretais/metabolismo , Curcumina/metabolismo , Feminino , Radioisótopos de Gálio/metabolismo , Células HT29 , Compostos Heterocíclicos com 1 Anel/metabolismo , Humanos , Camundongos Nus , Compostos Radiofarmacêuticos/química , Compostos Radiofarmacêuticos/metabolismo , Compostos Radiofarmacêuticos/farmacologia , Distribuição Tecidual
12.
J Biol Chem ; 292(37): 15266-15276, 2017 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-28765281

RESUMO

Persistent DNA damage induces profound alterations in gene expression that, in turn, influence tissue homeostasis, tumorigenesis, and cancer treatment outcome. However, the underlying mechanism for gene expression reprogramming induced by persistent DNA damage remains poorly understood. Here, using a highly effective bioluminescence-based reporter system and other tools, we report that persistent DNA damage inhibits nonsense-mediated RNA decay (NMD), an RNA surveillance and gene-regulatory pathway, in noncycling cells. NMD suppression by persistent DNA damage required the activity of the p38α MAPK. Activating transcription factor 3 (ATF3), an NMD target and a key stress-inducible transcription factor, was stabilized in a p38α- and NMD-dependent manner following persistent DNA damage. Our results reveal a novel p38α-dependent pathway that regulates NMD activity in response to persistent DNA damage, which, in turn, controls ATF3 expression in affected cells.


Assuntos
Fator 3 Ativador da Transcrição/metabolismo , Dano ao DNA , Regulação da Expressão Gênica , Proteína Quinase 14 Ativada por Mitógeno/metabolismo , Degradação do RNAm Mediada por Códon sem Sentido , RNA Mensageiro/metabolismo , Fator 3 Ativador da Transcrição/química , Fator 3 Ativador da Transcrição/genética , Biomarcadores/metabolismo , Bleomicina/toxicidade , Células Cultivadas , Senescência Celular , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/efeitos da radiação , Raios gama/efeitos adversos , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos da radiação , Genes Reporter/efeitos dos fármacos , Genes Reporter/efeitos da radiação , Células HEK293 , Humanos , Medições Luminescentes , Proteína Quinase 14 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 14 Ativada por Mitógeno/genética , Mutagênicos/toxicidade , Degradação do RNAm Mediada por Códon sem Sentido/efeitos dos fármacos , Degradação do RNAm Mediada por Códon sem Sentido/efeitos da radiação , Estresse Oxidativo , Estabilidade Proteica/efeitos dos fármacos , Estabilidade Proteica/efeitos da radiação , Interferência de RNA , Estabilidade de RNA/efeitos dos fármacos , Estabilidade de RNA/efeitos da radiação , RNA Mensageiro/química
13.
Bioconjug Chem ; 29(9): 3180-3195, 2018 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-30168713

RESUMO

Quantitative imaging of apoptosis in vivo could enable real-time monitoring of acute cell death pathologies such as traumatic brain injury, as well as the efficacy and safety of cancer therapy. Here, we describe the development and validation of F-18-labeled caspase-3 substrates for PET/CT imaging of apoptosis. Preliminary studies identified the O-benzylthreonine-containing substrate 2MP-TbD-AFC as a highly caspase 3-selective and cell-permeable fluorescent reporter. This lead compound was converted into the radiotracer [18F]-TBD, which was obtained at 10% decay-corrected yields with molar activities up to 149 GBq/µmol on an automated radiosynthesis platform. [18F]-TBD accumulated in ovarian cancer cells in a caspase- and cisplatin-dependent fashion. PET imaging of a Jo2-induced hepatotoxicity model showed a significant increase in [18F]-TBD signal in the livers of Jo2-treated mice compared to controls, driven through a reduction in hepatobiliary clearance. A chemical control tracer that could not be cleaved by caspase 3 showed no change in liver accumulation after induction of hepatocyte apoptosis. Our data demonstrate that [18F]-TBD provides an immediate pharmacodynamic readout of liver apoptosis in mice by dynamic PET/CT and suggest that [18F]-TBD could be used to interrogate apoptosis in other disease states.


Assuntos
Apoptose , Caspase 3/metabolismo , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada/métodos , Animais , Linhagem Celular Tumoral , Feminino , Camundongos , Camundongos Nus , Especificidade por Substrato
14.
Proc Natl Acad Sci U S A ; 112(51): E7148-54, 2015 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-26644583

RESUMO

Short-term fasting protects mice from lethal doses of chemotherapy through undetermined mechanisms. Herein, we demonstrate that fasting preserves small intestinal (SI) architecture by maintaining SI stem cell viability and SI barrier function following exposure to high-dose etoposide. Nearly all SI stem cells were lost in fed mice, whereas fasting promoted sufficient SI stem cell survival to preserve SI integrity after etoposide treatment. Lineage tracing demonstrated that multiple SI stem cell populations, marked by Lgr5, Bmi1, or HopX expression, contributed to fasting-induced survival. DNA repair and DNA damage response genes were elevated in SI stem/progenitor cells of fasted etoposide-treated mice, which importantly correlated with faster resolution of DNA double-strand breaks and less apoptosis. Thus, fasting preserved SI stem cell viability as well as SI architecture and barrier function suggesting that fasting may reduce host toxicity in patients undergoing dose intensive chemotherapy.


Assuntos
Dano ao DNA , Jejum/metabolismo , Intestino Delgado/metabolismo , Intestino Delgado/patologia , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/efeitos adversos , Apoptose/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Etoposídeo/administração & dosagem , Etoposídeo/efeitos adversos , Feminino , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Intestino Delgado/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Células-Tronco/patologia
15.
Bioconjug Chem ; 28(2): 583-589, 2017 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-28150941

RESUMO

Radiolabeling of substrates with 2-[18F]fluoroethylazide exploits the rapid kinetics, chemical selectivity, and mild conditions of the copper-catalyzed azide-alkyne cycloaddition reaction. While this methodology has proven to result in near-quantitative labeling of alkyne-tagged precursors, the relatively small size of the fluoroethylazide group makes separation of the 18F-labeled radiotracer and the unreacted precursor challenging, particularly with precursors >500 Da (e.g., peptides). We have developed an inexpensive azide-functionalized resin to rapidly remove unreacted alkyne precursor following the fluoroethylazide labeling reaction and integrated it into a fully automated radiosynthesis platform. We have carried out 2-[18F]fluoroethylazide labeling of four different alkynes ranging from <300 Da to >1700 Da and found that >98% of the unreacted alkyne was removed in less than 20 min at room temperature to afford the final radiotracers at >99% radiochemical purity with specific activities up to >200 GBq/µmol. We have applied this technique to label a novel cyclic peptide previously evolved to bind the Her2 receptor with high affinity, and demonstrated tumor-specific uptake and low nonspecific background by PET/CT. This resin-based methodology is automated, rapid, mild, and general allowing peptide-based fluorine-18 radiotracers to be obtained with clinically relevant specific activities without chromatographic separation and with only a minimal increase in total synthesis time.


Assuntos
Alcinos/química , Azidas/química , Radioisótopos de Flúor/química , Peptídeos Cíclicos/química , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada/métodos , Química Click/métodos , Cobre/química , Reação de Cicloadição/métodos
16.
Breast Cancer Res ; 18(1): 13, 2016 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-26818199

RESUMO

BACKGROUND: Despite advances in early diagnosis and treatment of cancer patients, metastasis remains the major cause of mortality. TP53 is one of the most frequently mutated genes in human cancer, and these alterations can occur during the early stages of oncogenesis or as later events as tumors progress to more aggressive forms. Previous studies have suggested that p53 plays a role in cellular pathways that govern metastasis. To investigate how p53 deficiency contributes to late-stage tumor growth and metastasis, we developed paired isogenic patient-derived xenograft (PDX) models of triple-negative breast cancer (TNBC) differing only in p53 status for longitudinal analysis. METHODS: Patient-derived isogenic human tumor lines differing only in p53 status were implanted into mouse mammary glands. Tumor growth and metastasis were monitored with bioluminescence imaging, and circulating tumor cells (CTCs) were quantified by flow cytometry. RNA-Seq was performed on p53-deficient and p53 wild-type tumors, and functional validation of a lead candidate gene was performed in vivo. RESULTS: Isogenic p53 wild-type and p53-deficient tumors metastasized out of mammary glands and colonized distant sites with similar frequency. However, p53-deficient tumors metastasized earlier than p53 wild-type tumors and grew faster in both primary and metastatic sites as a result of increased proliferation and decreased apoptosis. In addition, greater numbers of CTCs were detected in the blood of mice engrafted with p53-deficient tumors. However, when normalized to tumor mass, the number of CTCs isolated from mice bearing parental and p53-deficient tumors was not significantly different. Gene expression profiling followed by functional validation identified B cell translocation gene 2 (BTG2), a downstream effector of p53, as a negative regulator of tumor growth both at primary and metastatic sites. BTG2 expression status correlated with survival of TNBC patients. CONCLUSIONS: Using paired isogenic PDX-derived metastatic TNBC cells, loss of p53 promoted tumor growth and consequently increased tumor cell shedding into the blood, thus enhancing metastasis. Loss of BTG2 expression in p53-deficient tumors contributed to this metastatic potential by enhancing tumor growth in primary and metastatic sites. Furthermore, clinical data support conclusions generated from PDX models and indicate that BTG2 expression is a candidate prognostic biomarker for TNBC.


Assuntos
Proliferação de Células/genética , Proteínas Imediatamente Precoces/biossíntese , Neoplasias de Mama Triplo Negativas/genética , Proteína Supressora de Tumor p53/genética , Proteínas Supressoras de Tumor/biossíntese , Animais , Apoptose/genética , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Humanos , Proteínas Imediatamente Precoces/genética , Camundongos , Metástase Neoplásica , Células Neoplásicas Circulantes/patologia , Neoplasias de Mama Triplo Negativas/patologia , Proteínas Supressoras de Tumor/genética , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Mol Ther ; 23(6): 1110-1122, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25807290

RESUMO

Described herein is a first-in-man attempt to both genetically modify T cells with an imagable suicide gene and track these transduced donor T cells in allogeneic stem cell transplantation recipients using noninvasive positron emission tomography/computerized tomography (PET/CT) imaging. A suicide gene encoding a human CD34-Herpes Simplex Virus-1-thymidine kinase (CD34-TK75) fusion enabled enrichment of retrovirally transduced T cells (TdT), control of graft-versus-host disease and imaging of TdT migration and expansion in vivo in mice and man. Analysis confirmed that CD34-TK75-enriched TdT contained no replication competent γ-retrovirus, were sensitive to ganciclovir, and displayed characteristic retroviral insertion sites (by targeted sequencing). Affinity-purified CD34-TK75(+)-selected donor T cells (1.0-13 × 10(5))/kg were infused into eight patients who relapsed after allogeneic stem cell transplantation. Six patients also were administered 9-[4-((18)F)fluoro-3-hydroxymethyl-butyl]guanine ([(18)F]FHBG) to specifically track the genetically modified donor T cells by PET/CT at several time points after infusion. All patients were assessed for graft-versus-host disease, response to ganciclovir, circulating TdT cells (using both quantitative polymerase chain reaction and [(18)F]FHBG PET/CT imaging), TdT cell clonal expansion, and immune response to the TdT. This phase 1 trial demonstrated that genetically modified T cells and [(18)F]FHBG can be safely infused in patients with relapsed hematologic malignancies after allogeneic stem cell transplantation.


Assuntos
Antígenos CD34/imunologia , Tomografia por Emissão de Pósitrons/métodos , Transplante de Células-Tronco/métodos , Linfócitos T/imunologia , Transdução Genética , Transplante Homólogo/métodos , Animais , Antígenos CD34/genética , Antígenos CD34/metabolismo , Linhagem Celular Tumoral , Estudos de Viabilidade , Citometria de Fluxo , Ganciclovir/farmacologia , Doença Enxerto-Hospedeiro/imunologia , Guanina/administração & dosagem , Guanina/análogos & derivados , Herpesvirus Humano 1/genética , Humanos , Leucócitos Mononucleares/metabolismo , Camundongos , Células NIH 3T3 , Projetos Piloto , Linfócitos T/metabolismo , Timidina Quinase/genética , Timidina Quinase/metabolismo , Resultado do Tratamento
18.
J Labelled Comp Radiopharm ; 59(3): 103-8, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26853088

RESUMO

Anaplastic lymphoma kinase (ALK), an oncogenic receptor tyrosine kinase, has emerged as a therapeutic target in solid and hematologic tumors. Although several ALK inhibitors have gained recent approval for therapy, non-invasive indicators of target engagement or for use as predictive biomarkers in vivo are lacking. Therefore, we designed and synthesized a radiolabeled analogue of the ALK inhibitor ceritinib, [(18)F]fluoroethyl-ceritinib ([(18)F]-FEC), for use with positron emission tomography. We used two methods to synthesize [(18)F]-FEC. First, [(18)F]fluoroethyl-tosylate was prepared, coupled with ceritinib, and the product purified to yield [(18)F]-FEC. Alternatively, a precursor compound was synthesized, directly fluorinated with (18)F-fluoride, and purified to yield [(18)F]-FEC. The first method produced [(18)F]-FEC with an average decay-corrected yield of 24% (n = 4), specific activity of 1200 mCi/µmol, and >99% purity; synthesis time was 115 min from the end of bombardment. The second method produced [(18)F]-FEC with an average yield of 7% (n = 4), specific activity of 1500 mCi/µmol, and >99% purity; synthesis time was 65 min from the end of bombardment. The synthesis of a novel (18)F-labeled analogue of ceritinib has been achieved in acceptable yields, at high purity, and with high specific activity. The compound is a potential positron emission tomography imaging agent for the detection of ALK-overexpressing solid tumors such as lung cancer.


Assuntos
Radioisótopos de Flúor/química , Inibidores de Proteínas Quinases/química , Pirimidinas/química , Pirimidinas/síntese química , Compostos Radiofarmacêuticos/síntese química , Sulfonas/química , Sulfonas/síntese química , Tomografia por Emissão de Pósitrons
19.
Proc Natl Acad Sci U S A ; 109(1): 137-42, 2012 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-22190492

RESUMO

Binding of EGF to its receptor induces dimerization of the normally monomeric receptor. Activation of its intracellular tyrosine kinase then occurs through the formation of an asymmetric kinase dimer in which one subunit, termed the "receiver" kinase, is activated by interaction with the other subunit, termed the "activator" kinase [Zhang, et al. (2006) Cell 125: 1137-1149]. Although there is significant experimental support for this model, the relationship between ligand binding and the mechanics of kinase activation are not known. Here we use luciferase fragment complementation in EGF receptor (EGFR)/ErbB2 heterodimers to probe the mechanics of ErbB kinase activation. Our data support a model in which ligand binding causes the cis-kinase (the EGFR) to adopt the receiver position in the asymmetric dimer and to be activated first. If the EGF receptor is kinase active, this results in the phosphorylation of the trans-kinase (ErbB2). However, if the EGF receptor kinase is kinase dead, the ErbB2 kinase is never activated. Thus, activation of the kinases in the EGFR/ErbB2 asymmetric dimer occurs in a specific sequence and depends on the kinase activity of the EGF receptor.


Assuntos
Bioquímica/métodos , Receptores ErbB/metabolismo , Imageamento Tridimensional/métodos , Luciferases/metabolismo , Fragmentos de Peptídeos/metabolismo , Receptor ErbB-2/metabolismo , Animais , Células CHO , Cricetinae , Cricetulus , Ativação Enzimática/efeitos dos fármacos , Fator de Crescimento Epidérmico/farmacologia , Receptores ErbB/química , Teste de Complementação Genética , Modelos Moleculares , Multimerização Proteica/efeitos dos fármacos , Estrutura Terciária de Proteína , Receptor ErbB-2/química , Transdução de Sinais/efeitos dos fármacos
20.
J Biol Chem ; 288(42): 30773-30784, 2013 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-24014028

RESUMO

ErbB3 is a member of the ErbB family of receptor tyrosine kinases. It is unique because it is the only member of the family whose kinase domain is defective. As a result, it is obliged to form heterodimers with other ErbB receptors to signal. In this study, we characterized the interaction of ErbB3 with the EGF receptor and ErbB2 and assessed the effects of Food and Drug Administration-approved therapeutic agents on these interactions. Our findings support the concept that ErbB3 exists in preformed clusters that can be dissociated by NRG-1ß and that it interacts with other ErbB receptors in a distinctly hierarchical fashion. Our study also shows that all pairings of the EGF receptor, ErbB2, and ErbB3 form ligand-independent dimers/oligomers. The small-molecule tyrosine kinase inhibitors erlotinib and lapatinib differentially enhance the dimerization of the various ErbB receptor pairings, with the EGFR/ErbB3 heterodimer being particularly sensitive to the effects of erlotinib. The data suggest that the physiological effects of these drugs may involve not only inhibition of tyrosine kinase activity but also a dynamic restructuring of the entire network of receptors.


Assuntos
Receptores ErbB/metabolismo , Complexos Multienzimáticos/metabolismo , Multimerização Proteica , Receptor ErbB-2/metabolismo , Receptor ErbB-3/metabolismo , Animais , Células CHO , Cricetinae , Cricetulus , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/genética , Cloridrato de Erlotinib , Humanos , Lapatinib , Luciferases/genética , Luciferases/metabolismo , Complexos Multienzimáticos/antagonistas & inibidores , Complexos Multienzimáticos/genética , Neuregulina-1/genética , Neuregulina-1/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Quinazolinas/farmacologia , Receptor ErbB-2/antagonistas & inibidores , Receptor ErbB-2/genética , Receptor ErbB-3/antagonistas & inibidores , Receptor ErbB-3/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA