Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Magn Reson Med ; 91(2): 819-827, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37815014

RESUMO

PURPOSE: To develop a portable MR perfusion phantom for quality-controlled assessment and reproducibility of arterial spin labeled (ASL) perfusion measurement. METHODS: A 3D-printed perfusion phantom was developed that mimics the branching of arterial vessels, capillaries, and a chamber containing cellulose sponge representing tissue characteristics. A peristaltic pump circulated distilled water through the phantom, and was first evaluated at 300, 400, and 500 mL/min. Longitudinal reproducibility of perfusion was performed using 2D pseudo-continuous ASL at 20 post-label delays (PLDs, ranging between 0.2 and 7.8 s at 0.4-s intervals) over a period of 16 weeks, with three repetitions each week. Multi-PLD data were fitted into a general kinetic model for perfusion quantification (f) and arterial transit time (ATT). Intraclass correlation coefficient was used to assess intersession reproducibility. RESULTS: MR perfusion signals acquired in the 3D-printed perfusion phantom agreed well with the experimental conditions, with progressively increasing signal intensities and decreasing ATT for pump flow rates from 300 to 500 mL/min. The perfusion signal at 400 mL/min and the general kinetic model-derived f and ATT maps were similar across all PLDs for both intrasession and intersession reproducibility. Across all 48 experimental time points, the average f was 75.55 ± 3.83 × 10-3 mL/mL/s, the corresponding ATT was 2.10 ± 0.20 s, and the T1 was 1.84 ± 0.102 s. Intraclass correlation coefficient was 0.92 (95% confidence interval 0.83-0.97) for f, 0.96 (0.91-0.99) for ATT, and 0.94 (0.88-0.98) for T1 , demonstrating excellent reproducibility. CONCLUSION: A simple, portable 3D-printed perfusion phantom with excellent reproducibility of 2D pseudo-continuous ASL measurements was demonstrated that can serve for quality-controlled and reliable measurements of ASL perfusion.


Assuntos
Circulação Cerebrovascular , Imageamento por Ressonância Magnética , Marcadores de Spin , Reprodutibilidade dos Testes , Perfusão , Impressão Tridimensional
2.
Magn Reson Imaging ; 104: 80-87, 2023 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-37769882

RESUMO

PURPOSE: To evaluate different approaches for the effective assessment of pulmonary perfusion with a pseudo-continuous arterial spin labeled (pCASL) MRI. MATERIALS AND METHODS: Four different approaches were evaluated: 1) Cardiac-triggered inferior vena cava (IVC) labeling; 2) IVC labeling with cardiac-triggered acquisition; 3) Right pulmonary artery (RPA) labeling with cardiac-triggered acquisition; and 4) Cardiac-triggered RPA labeling with background suppression (BGS). Each approach was evaluated in 5 healthy volunteers (n = 20) using coefficient of variation (COV) across averages. Approach 4 was also compared against a flow alternating inversion recovery (FAIR). RESULTS: The IVC labeling (Approach 1) achieved perfusion-weighted images of both lungs, although this approach was more sensitive to variations in heart rate. Cardiac-triggered acquisitions using IVC (Approach 2) and RPA (Approach 3) labeling improved signal consistencies, but were incompatible with BGS. The cardiac-triggered RPA labeling with BGS (Approach 4) achieved a COV of 0.34 ± 0.03 (p < 0.05 compared to IVC labeling approaches) and resulted in perfusion value of 434 ± 64 mL/100 g/min, which was comparable to 451 ± 181 mL/100 g/min measured by FAIR (p = 0.82). DISCUSSION: Pulmonary perfusion imaging using pCASL-MRI is highly sensitive to cardiac phase, and requires approaches to minimize flow-induced signal variations. Cardiac-triggered RPA labeling with BGS achieves reduced COV and enables robust pulmonary perfusion imaging.

3.
Eur Radiol ; 33(12): 9223-9232, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37466705

RESUMO

OBJECTIVES: To evaluate longitudinal placental perfusion using pseudo-continuous arterial spin-labeled (pCASL) MRI in normal pregnancies and in pregnancies affected by chronic hypertension (cHTN), who are at the greatest risk for placental-mediated disease conditions. METHODS: Eighteen normal and 23 pregnant subjects with cHTN requiring antihypertensive therapy were scanned at 3 T using free-breathing pCASL-MRI at 16-20 and 24-28 weeks of gestational age. RESULTS: Mean placental perfusion was 103.1 ± 48.0 and 71.4 ± 18.3 mL/100 g/min at 16-20 and 24-28 weeks respectively in normal pregnancies and 79.4 ± 27.4 and 74.9 ± 26.6 mL/100 g/min in cHTN pregnancies. There was a significant decrease in perfusion between the first and second scans in normal pregnancies (p = 0.004), which was not observed in cHTN pregnancies (p = 0.36). The mean perfusion was not statistically different between normal and cHTN pregnancies at both scans, but the absolute change in perfusion per week was statistically different between these groups (p = 0.044). Furthermore, placental perfusion was significantly lower at both time points (p = 0.027 and 0.044 respectively) in the four pregnant subjects with cHTN who went on to have infants that were small for gestational age (52.7 ± 20.4 and 50.4 ± 20.9 mL/100 g/min) versus those who did not (85 ± 25.6 and 80.0 ± 25.1 mL/100 g/min). CONCLUSION: pCASL-MRI enables longitudinal assessment of placental perfusion in pregnant subjects. Placental perfusion in the second trimester declined in normal pregnancies whereas it remained unchanged in cHTN pregnancies, consistent with alterations due to vascular disease pathology. Perfusion was significantly lower in those with small for gestational age infants, indicating that pCASL-MRI-measured perfusion may be an effective imaging biomarker for placental insufficiency. CLINICAL RELEVANCE STATEMENT: pCASL-MRI enables longitudinal assessment of placental perfusion without administering exogenous contrast agent and can identify placental insufficiency in pregnant subjects with chronic hypertension that can lead to earlier interventions. KEY POINTS: • Arterial spin-labeled (ASL) magnetic resonance imaging (MRI) enables longitudinal assessment of placental perfusion without administering exogenous contrast agent. • ASL-MRI-measured placental perfusion decreased significantly between 16-20 week and 24-28 week gestational age in normal pregnancies, while it remained relatively constant in hypertensive pregnancies, attributed to vascular disease pathology. • ASL-MRI-measured placental perfusion was significantly lower in subjects with hypertension who had a small for gestational age infant at 16-20-week gestation, indicating perfusion as an effective biomarker of placental insufficiency.


Assuntos
Hipertensão , Insuficiência Placentária , Gravidez , Feminino , Humanos , Lactente , Placenta/diagnóstico por imagem , Marcadores de Spin , Meios de Contraste , Imageamento por Ressonância Magnética/métodos , Perfusão , Biomarcadores
4.
Front Med (Lausanne) ; 10: 1199146, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37441689

RESUMO

Chimeric antigen receptor (CAR) T-cell therapies have evolved as breakthrough treatment options for the management of hematological malignancies and are also being developed as therapeutics for solid tumors. However, despite the impressive patient responses from CD19-directed CAR T-cell therapies, ~ 40%-60% of these patients' cancers eventually relapse, with variable prognosis. Such relapses may occur due to a combination of molecular resistance mechanisms, including antigen loss or mutations, T-cell exhaustion, and progression of the immunosuppressive tumor microenvironment. This class of therapeutics is also associated with certain unique toxicities, such as cytokine release syndrome, immune effector cell-associated neurotoxicity syndrome, and other "on-target, off-tumor" toxicities, as well as anaphylactic effects. Furthermore, manufacturing limitations and challenges associated with solid tumor infiltration have delayed extensive applications. The molecular imaging modalities of immunological positron emission tomography and single-photon emission computed tomography (immuno-PET/-SPECT) offer a target-specific and highly sensitive, quantitative, non-invasive platform for longitudinal detection of dynamic variations in target antigen expression in the body. Leveraging these imaging strategies as guidance tools for use with CAR T-cell therapies may enable the timely identification of resistance mechanisms and/or toxic events when they occur, permitting effective therapeutic interventions. In addition, the utilization of these approaches in tracking the CAR T-cell pharmacokinetics during product development and optimization may help to assess their efficacy and accordingly to predict treatment outcomes. In this review, we focus on current challenges and potential opportunities in the application of immuno-PET/-SPECT imaging strategies to address the challenges encountered with CAR T-cell therapies.

5.
Clin Cancer Res ; 27(17): 4794-4806, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34210685

RESUMO

PURPOSE: Intratumoral heterogeneity (ITH) challenges the molecular characterization of clear cell renal cell carcinoma (ccRCC) and is a confounding factor for therapy selection. Most approaches to evaluate ITH are limited by two-dimensional ex vivo tissue analyses. Dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) can noninvasively assess the spatial landscape of entire tumors in their natural milieu. To assess the potential of DCE-MRI, we developed a vertically integrated radiogenomics colocalization approach for multi-region tissue acquisition and analyses. We investigated the potential of spatial imaging features to predict molecular subtypes using histopathologic and transcriptome correlatives. EXPERIMENTAL DESIGN: We report the results of a prospective study of 49 patients with ccRCC who underwent DCE-MRI prior to nephrectomy. Surgical specimens were sectioned to match the MRI acquisition plane. RNA sequencing data from multi-region tumor sampling (80 samples) were correlated with percent enhancement on DCE-MRI in spatially colocalized regions of the tumor. Independently, we evaluated clinical applicability of our findings in 19 patients with metastatic RCC (39 metastases) treated with first-line antiangiogenic drugs or checkpoint inhibitors. RESULTS: DCE-MRI identified tumor features associated with angiogenesis and inflammation, which differed within and across tumors, and likely contribute to the efficacy of antiangiogenic drugs and immunotherapies. Our vertically integrated analyses show that angiogenesis and inflammation frequently coexist and spatially anti-correlate in the same tumor. Furthermore, MRI contrast enhancement identifies phenotypes with better response to antiangiogenic therapy among patients with metastatic RCC. CONCLUSIONS: These findings have important implications for decision models based on biopsy samples and highlight the potential of more comprehensive imaging-based approaches.


Assuntos
Carcinoma de Células Renais/diagnóstico por imagem , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/patologia , Neoplasias Renais/diagnóstico por imagem , Neoplasias Renais/genética , Neoplasias Renais/patologia , Imageamento por Ressonância Magnética/métodos , Genômica por Radiação , Microambiente Tumoral , Adulto , Idoso , Idoso de 80 Anos ou mais , Inibidores da Angiogênese/uso terapêutico , Carcinoma de Células Renais/tratamento farmacológico , Feminino , Humanos , Neoplasias Renais/tratamento farmacológico , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos
6.
Magn Reson Med ; 86(3): 1463-1471, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33929055

RESUMO

PURPOSE: To develop a true single shot turbo spin echo (SShTSE) acquisition with Dixon for robust T2 -weighted abdominal imaging with uniform fat and water separation at 3T. METHODS: The in-phase (IP) and out-of-phase (OP) echoes for Dixon processing were acquired in the same repetition time of a SShTSE using partial echoes. A phase-preserved bi-directional homodyne reconstruction was developed to compensate the partial echo and the partial phase encoding of SShTSE. With IRB approval, the SShTSE-Dixon was compared against standard SShTSE, without and with fat suppression using spectral adiabatic inversion recovery (SPAIR) in 5 healthy volunteers and 5 patients. The SNR and contrast ratio (CR) of spleen to liver were compared among different acquisitions. RESULTS: The bi-directional homodyne reconstruction successfully minimized ringing artifacts because of partial acquisitions. SShTSE-Dixon achieved uniform fat suppression compared to SShTSE-SPAIR with fat suppression failures of 1/10 versus 10/10 in the axial plane and 0/5 versus 5/5 in the coronal plane, respectively. The SNRs of the liver (12.2 ± 4.9 vs. 11.7 ± 5.2; P = .76) and spleen (25.9 ± 11.6 vs. 23.7 ± 9.7; P = .14) were equivalent between fat-suppressed images (SShTSE-Dixon water-only and SShTSE-SPAIR). The SNRs of liver (14.4 ± 5.7 vs. 13.4 ± 5.0; P = .60) and spleen (26.5 ± 10.1 vs. 25.7 ± 8.5; P = .56) were equivalent between non-fat-suppressed images (SShTSE-Dixon IP and SShTSE). The CRs of spleen to liver were also similar between fat-suppressed images (2.6 ± 0.4 vs. 2.5 ± 0.5; P =.92) and non-fat-suppressed images (2.3 ± 0.6 vs. 2.2 ± 0.4; P =.84). CONCLUSION: SShTSE-Dixon generates robust abdominal T2 -weighted images at 3T with and without uniform fat suppression, along with perfectly co-registered fat-only images in a single acquisition.


Assuntos
Imageamento por Ressonância Magnética , Água , Tecido Adiposo/diagnóstico por imagem , Humanos , Aumento da Imagem , Interpretação de Imagem Assistida por Computador
7.
Clin Genitourin Cancer ; 19(1): 12-21.e1, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32669212

RESUMO

INTRODUCTION: Percutaneous renal mass biopsy results can accurately diagnose clear cell renal cell carcinoma (ccRCC); however, their reliability to determine nuclear grade in larger, heterogeneous tumors is limited. We assessed the ability of radiomics analyses of magnetic resonance imaging (MRI) to predict high-grade (HG) histology in ccRCC. PATIENTS AND METHODS: Seventy patients with a renal mass underwent 3 T MRI before surgery between August 2012 and August 2017. Tumor length, first-order statistics, and Haralick texture features were calculated on T2-weighted and dynamic contrast-enhanced (DCE) MRI after manual tumor segmentation. After a variable clustering algorithm was applied, tumor length, washout, and all cluster features were evaluated univariably by receiver operating characteristic curves. Three logistic regression models were constructed to assess the predictability of HG ccRCC and then cross-validated. RESULTS: At univariate analysis, area under the curve values of length, and DCE texture cluster 1 and cluster 3 for diagnosis of HG ccRCC were 0.7 (95% confidence interval [CI], 0.58-0.82, false discovery rate P = .008), 0.72 (95% CI, 0.59-0.84, false discovery rate P = .004), and 0.75 (95% CI, 0.63-0.87, false discovery rate P = .0009), respectively. At multivariable analysis, area under the curve for model 1 (tumor length only), model 2 (length + DCE clusters 3 and 4), and model 3 (DCE cluster 1 and 3) for diagnosis of HG ccRCC were 0.67 (95% CI, 0.54-0.79), 0.82 (95% CI, 0.71-0.92), and 0.81 (95% CI, 0.70-0.91), respectively. CONCLUSION: Radiomics analysis of MRI images was superior to tumor size for the prediction of HG histology in ccRCC in our cohort.


Assuntos
Carcinoma de Células Renais , Neoplasias Renais , Carcinoma de Células Renais/diagnóstico por imagem , Humanos , Neoplasias Renais/diagnóstico por imagem , Imageamento por Ressonância Magnética , Necrose/diagnóstico por imagem , Reprodutibilidade dos Testes , Estudos Retrospectivos
8.
Contrast Media Mol Imaging ; 2018: 2082154, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30534026

RESUMO

Widespread use of screening mammography has recently increased the detection of breast microcalcifications. These nonpalpable microcalcifications with specific features in breast tissues are clinically considered an early indicator of breast carcinoma. Our goal in this study was to develop a murine breast microcalcification model for optimizing in vivo imaging. Recombinant human BMP-2 was expressed in E. coli, and the purified bioactive protein was used as inducing factor for the production of breast microcalcifications in a murine animal model. Syngeneic breast tumors were obtained by injection of MDA-MB-231 human breast cancer cells with Matrigel into the mammary fat pad of female nude mice. Different doses of bioactive rhBMP-2 were administered either as single or multiple intraperitoneal injections or directly into tumor on a weekly basis. Three weeks after the first injection of rhBMP-2, the microcalcification of breast tumor was detected by microcomputed tomography followed by intravenous injection of radiotracer [18F] Sodium fluoride for positron emission tomography imaging. Our findings indicate that rhBMP-2 induced microcalcifications of breast tumor by both systemic and direct injection of rhBMP-2 into tumors in a dose-dependent manner. Although little is known about the molecular mechanism of microcalcification, here we report a new murine model of human breast tumor induced microcalcification by rhBMP-2 to optimize in vivo imaging methods and to study the role of BMP-2 as a mediator of pathological mineralization and bone-like microcalcification formation in breast tumor. This BMP-2-induced microcalcification model may allow us to discriminate the type of microcalcification in tumors and to perform quantitative analysis on the calcification as a new detection strategy for early identification of pathological mineralization of breast tissues in women.


Assuntos
Proteína Morfogenética Óssea 2/administração & dosagem , Neoplasias da Mama/diagnóstico por imagem , Calcinose/induzido quimicamente , Transplante Heterólogo/métodos , Animais , Proteína Morfogenética Óssea 2/farmacologia , Calcinose/diagnóstico , Linhagem Celular , Feminino , Radioisótopos de Flúor , Humanos , Camundongos , Camundongos Nus , Tomografia por Emissão de Pósitrons/métodos , Fluoreto de Sódio/administração & dosagem , Microtomografia por Raio-X
9.
Cancer Res ; 78(3): 781-797, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29229598

RESUMO

Checkpoint kinase inhibitors (CHKi) exhibit striking single-agent activity in certain tumors, but the mechanisms accounting for hypersensitivity are poorly understood. We screened a panel of 49 established human head and neck squamous cell carcinoma (HNSCC) cell lines and report that nearly 20% are hypersensitive to CHKi monotherapy. Hypersensitive cells underwent early S-phase arrest at drug doses sufficient to inhibit greater than 90% of CHK1 activity. Reduced rate of DNA replication fork progression and chromosomal shattering were also observed, suggesting replication stress as a root causative factor in CHKi hypersensitivity. To explore genomic underpinnings of CHKi hypersensitivity, comparative genomic analysis was performed between hypersensitive cells and cells categorized as least sensitive because they showed drug IC50 value greater than the cell panel median and lacked early S-phase arrest. Novel association between CDKN2A/p16 copy number loss, CDK2 activation, replication stress, and hypersensitivity of HNSCC cells to CHKi monotherapy was found. Restoring p16 in cell lines harboring CDKN2A/p16 genomic deletions alleviated CDK2 activation and replication stress, attenuating CHKi hypersensitivity. Taken together, our results suggest a biomarker-driven strategy for selecting HNSCC patients who may benefit the most from CHKi therapy.Significance: These results suggest a biomarker-driven strategy for selecting HNSCC patients who may benefit the most from therapy with CHK inhibitors. Cancer Res; 78(3); 781-97. ©2017 AACR.


Assuntos
Carcinoma de Células Escamosas/patologia , Quinase 1 do Ponto de Checagem/antagonistas & inibidores , Quinase 2 Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p18/genética , Inibidores Enzimáticos/farmacologia , Neoplasias de Cabeça e Pescoço/patologia , Fase S , Antineoplásicos/farmacologia , Apoptose , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Proliferação de Células , Quinase 2 Dependente de Ciclina/genética , Inibidor p16 de Quinase Dependente de Ciclina , Replicação do DNA , Ativação Enzimática , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Neoplasias de Cabeça e Pescoço/genética , Neoplasias de Cabeça e Pescoço/metabolismo , Humanos , Deleção de Sequência , Células Tumorais Cultivadas
10.
JCI Insight ; 2(15)2017 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-28768909

RESUMO

BACKGROUND: Dysregulated lipid and glucose metabolism in clear cell renal cell carcinoma (ccRCC) has been implicated in disease progression, and whole tumor tissue-based assessment of these changes is challenged by the tumor heterogeneity. We studied a noninvasive quantitative MRI method that predicts metabolic alterations in the whole tumor. METHODS: We applied Dixon-based MRI for in vivo quantification of lipid accumulation (fat fraction [FF]) in targeted regions of interest of 45 primary ccRCCs and correlated these MRI measures to mass spectrometry-based lipidomics and metabolomics of anatomically colocalized tissue samples isolated from the same tumor after surgery. RESULTS: In vivo tumor FF showed statistically significant (P < 0.0001) positive correlation with histologic fat content (Spearman correlation coefficient, ρ = 0.79), spectrometric triglycerides (ρ = 0.56) and cholesterol (ρ = 0.47); it showed negative correlation with free fatty acids (ρ = -0.44) and phospholipids (ρ = -0.65). We observed both inter- and intratumoral heterogeneity in lipid accumulation within the same tumor grade, whereas most aggressive tumors (International Society of Urological Pathology [ISUP] grade 4) exhibited reduced lipid accumulation. Cellular metabolites in tumors were altered compared with adjacent renal parenchyma. CONCLUSION: Our results support the use of noninvasive quantitative Dixon-based MRI as a biomarker of reprogrammed lipid metabolism in ccRCC, which may serve as a predictor of tumor aggressiveness before surgical intervention. FUNDING: NIH R01CA154475 (YZ, MF, PK, IP), NIH P50CA196516 (IP, JB, RJD, JAC, PK), Welch Foundation I-1832 (JY), and NIH P01HL020948 (JGM).

11.
J Biol Chem ; 292(1): 112-120, 2017 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-27872191

RESUMO

Glutathione S-transferase pi 1 (GSTP1) is frequently overexpressed in cancerous tumors and is a putative target of the plant compound piperlongumine (PL), which contains two reactive olefins and inhibits proliferation in cancer cells but not normal cells. PL exposure of cancer cells results in increased reactive oxygen species and decreased GSH. These data in tandem with other information led to the conclusion that PL inhibits GSTP1, which forms covalent bonds between GSH and various electrophilic compounds, through covalent adduct formation at the C7-C8 olefin of PL, whereas the C2-C3 olefin of PL was postulated to react with GSH. However, direct evidence for this mechanism has been lacking. To investigate, we solved the X-ray crystal structure of GSTP1 bound to PL and GSH at 1.1 Å resolution to rationalize previously reported structure activity relationship studies. Surprisingly, the structure showed that a hydrolysis product of PL (hPL) was conjugated to glutathione at the C7-C8 olefin, and this complex was bound to the active site of GSTP1; no covalent bond formation between hPL and GSTP1 was observed. Mass spectrometry (MS) analysis of the reactions between PL and GSTP1 confirmed that PL does not label GSTP1. Moreover, MS data also indicated that nucleophilic attack on PL at the C2-C3 olefin led to PL hydrolysis. Although hPL inhibits GSTP1 enzymatic activity in vitro, treatment of cells susceptible to PL with hPL did not have significant anti-proliferative effects, suggesting that hPL is not membrane-permeable. Altogether, our data suggest a model wherein PL is a prodrug whose intracellular hydrolysis initiates the formation of the hPL-GSH conjugate, which blocks the active site of and inhibits GSTP1 and thereby cancer cell proliferation.


Assuntos
Proliferação de Células/efeitos dos fármacos , Dioxolanos/farmacologia , Glutationa S-Transferase pi/química , Glutationa S-Transferase pi/metabolismo , Glutationa/metabolismo , Neoplasias Pancreáticas/patologia , Cristalografia por Raios X , Humanos , Espectrometria de Massas , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/enzimologia , Ligação Proteica , Conformação Proteica , Células Tumorais Cultivadas
12.
Radiat Res ; 185(5): 527-38, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-27135971

RESUMO

Several classes of inhibitors of the mammalian target of rapamycin (mTOR) have been developed based on its central role in sensing growth factor and nutrient levels to regulate cellular metabolism. However, its ATP-binding site closely resembles other phosphatidylinositol 3-kinase-related kinase (PIKK) family members, resulting in reactivity with these targets that may also be therapeutically useful. The ATP-competitive mTOR inhibitor, Torin2, shows biochemical activity against the DNA repair-associated proteins ATM, ATR and DNA-PK, which raises the possibility that Torin2 and related compounds might radiosensitize cancerous tumors. In this study Torin2 was also found to enhance ionizing radiation-induced cell killing in conditions where ATM was dispensable, confirming the requirement for multiple PIKK targets. Moreover, Torin2 did not influence the initial appearance of γ-H2AX foci after irradiation but significantly delayed the disappearance of radiation-induced γ-H2AX foci, indicating a DNA repair defect. Torin2 increased the number of radiation-induced S-phase specific chromosome aberrations and reduced the frequency of radiation-induced CtIP and Rad51 foci formation, suggesting that Torin2 works by blocking homologous recombination (HR)-mediated DNA repair resulting in an S-phase specific DNA repair defect. Accordingly, Torin2 reduced HR-mediated repair of I-Sce1-induced DNA damage and contributed to replication fork stalling. We conclude that radiosensitization of tumor cells by Torin2 is associated with disrupting ATR- and ATM-dependent DNA damage responses. Our findings support the concept of developing combination cancer therapies that incorporate ionizing radiation therapy and Torin2 or compounds with similar properties.


Assuntos
Dano ao DNA , Reparo do DNA/efeitos dos fármacos , Reparo do DNA/efeitos da radiação , Naftiridinas/farmacologia , Radiossensibilizantes/farmacologia , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Aberrações Cromossômicas/efeitos dos fármacos , Aberrações Cromossômicas/efeitos da radiação , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Quebras de DNA de Cadeia Dupla/efeitos da radiação , Replicação do DNA/efeitos dos fármacos , Replicação do DNA/efeitos da radiação , Fase G2/efeitos dos fármacos , Fase G2/efeitos da radiação , Histonas/metabolismo , Recombinação Homóloga/efeitos dos fármacos , Recombinação Homóloga/efeitos da radiação , Humanos , Cinética , Fase S/efeitos dos fármacos , Fase S/efeitos da radiação
13.
Biochem Biophys Res Commun ; 463(4): 473-8, 2015 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-25998385

RESUMO

Nonhomologous end joining (NHEJ) is a major pathway for repair of DNA double-strand breaks. We have previously shown that a complex of SFPQ (PSF) and NONO (p54(nrb)) cooperates with Ku protein at an early step of NHEJ, forming a committed preligation complex and stimulating end-joining activity by 10-fold or more. SFPQ and NONO show no resemblance to other repair factors, and their mechanism of action is uncertain. Here, we use an optimized microwell-based assay to characterize the in vitro DNA binding behavior of the native SFPQ·NONO complex purified from human (HeLa) cells. SFPQ·NONO and Ku protein bind independently to DNA, with little evidence of cooperativity and only slight mutual interference at high concentration. Whereas Ku protein requires free DNA ends for binding, SFPQ·NONO does not. Both Ku and SFPQ·NONO have pairing activity, as measured by the ability of DNA-bound protein to capture a second DNA fragment in a microwell-based assay. Additionally, SFPQ·NONO stimulates DNA-dependent protein kinase autophosphorylation, consistent with the ability to promote formation of a synaptic complex formation without occluding the DNA termini proper. These findings suggest that SFPQ·NONO promotes end joining by binding to internal DNA sequences and cooperating with other repair proteins to stabilize a synaptic pre-ligation complex.


Assuntos
Reparo do DNA , DNA/metabolismo , Proteínas Associadas à Matriz Nuclear/metabolismo , Fatores de Transcrição de Octâmero/metabolismo , Proteínas de Ligação a RNA/metabolismo , Reparo do DNA por Junção de Extremidades , DNA Helicases/metabolismo , Proteínas de Ligação a DNA , Células HeLa , Humanos , Autoantígeno Ku
14.
Methods Mol Biol ; 1288: 317-36, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25827888

RESUMO

Loss of function or mutation of the ataxia-telangiectasia mutated gene product (ATM) results in inherited genetic disorders characterized by neurodegeneration, immunodeficiency, and cancer. Ataxia-telangiectasia mutated (ATM) gene product belongs to the PI3K-like protein kinase (PIKKs) family and is functionally implicated in mitogenic signal transduction, chromosome condensation, meiotic recombination, cell-cycle control, and telomere maintenance. The ATM protein kinase is primarily activated in response to DNA double strand breaks (DSBs), the most deleterious form of DNA damage produced by ionizing radiation (IR) or radiomimetic drugs. It is detected at DNA damage sites, where ATM autophosphorylation causes dissociation of the inactive homodimeric form to the activated monomeric form. Interestingly, heat shock can activate ATM independent of the presence of DNA strand breaks. ATM is an integral part of the sensory machinery that detects DSBs during meiosis, mitosis, or DNA breaks mediated by free radicals. These DNA lesions can trigger higher order chromatin reorganization fuelled by posttranslational modifications of histones and histone binding proteins. Our group, and others, have shown that ATM activation is tightly regulated by chromatin modifications. This review summarizes the multiple approaches used to discern the role of ATM and other associated proteins in chromatin modification in response to DNA damage.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Cromatina/genética , Cromatina/metabolismo , Dano ao DNA , Western Blotting/métodos , Imunoprecipitação da Cromatina/métodos , Análise Citogenética/métodos
15.
Cancer Res ; 75(5): 858-69, 2015 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-25589350

RESUMO

Proliferating mammalian stem and cancer cells express telomerase [telomerase reverse transcriptase (TERT)] in an effort to extend chromosomal G-overhangs and maintain telomere ends. Telomerase-expressing cells also have higher levels of the single-stranded DNA-binding protein SSB1, which has a critical role in DNA double-strand break (DSB) repair. Here, we report that SSB1 binds specifically to G-strand telomeric DNA in vitro and associates with telomeres in vivo. SSB1 interacts with the TERT catalytic subunit and regulates its interaction with telomeres. Deletion of SSB1 reduces TERT interaction with telomeres and leads to G-overhang loss. Although SSB1 is recruited to DSB sites, we found no corresponding change in TERT levels at these sites, implying that SSB1-TERT interaction relies upon a specific chromatin structure or context. Our findings offer an explanation for how telomerase is recruited to telomeres to facilitate G-strand DNA extension, a critical step in maintaining telomere ends and cell viability in all cancer cells. Cancer Res; 75(5); 858-69. ©2015 AACR.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Telomerase/metabolismo , Telômero/metabolismo , Animais , Dano ao DNA , DNA de Cadeia Simples/metabolismo , Células HCT116 , Células HEK293 , Humanos , Camundongos , Camundongos Knockout , Proteínas Mitocondriais/metabolismo , Ligação Proteica , Fase S/fisiologia , Proteína 1 de Ligação a Repetições Teloméricas/metabolismo
16.
Nat Chem Biol ; 10(12): 1006-12, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25326665

RESUMO

Her3 (also known as ErbB3) belongs to the epidermal growth factor receptor tyrosine kinases and is well credentialed as an anti-cancer target but is thought to be 'undruggable' using ATP-competitive small molecules because it lacks appreciable kinase activity. Here we report what is to our knowledge the first selective Her3 ligand, TX1-85-1, that forms a covalent bond with Cys721 located in the ATP-binding site of Her3. We demonstrate that covalent modification of Her3 inhibits Her3 signaling but not proliferation in some Her3-dependent cancer cell lines. Subsequent derivatization with a hydrophobic adamantane moiety demonstrates that the resultant bivalent ligand (TX2-121-1) enhances inhibition of Her3-dependent signaling. Treatment of cells with TX2-121-1 results in partial degradation of Her3 and serendipitously interferes with productive heterodimerization between Her3 with either Her2 or c-Met. These results suggest that small molecules will be capable of perturbing the biological function of Her3 and ∼60 other pseudokinases found in human cells.


Assuntos
Acrilamidas/farmacologia , Adenina/análogos & derivados , Antineoplásicos/farmacologia , Regulação Neoplásica da Expressão Gênica , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-met/química , Receptor ErbB-2/química , Receptor ErbB-3/antagonistas & inibidores , Acrilamidas/síntese química , Adamantano/química , Adenina/síntese química , Adenina/farmacologia , Trifosfato de Adenosina/química , Trifosfato de Adenosina/metabolismo , Antineoplásicos/síntese química , Domínio Catalítico , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Cisteína/química , Humanos , Interações Hidrofóbicas e Hidrofílicas , Simulação de Acoplamento Molecular , Terapia de Alvo Molecular , Inibidores de Proteínas Quinases/síntese química , Multimerização Proteica , Proteólise , Proteínas Proto-Oncogênicas c-met/genética , Proteínas Proto-Oncogênicas c-met/metabolismo , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Receptor ErbB-3/química , Receptor ErbB-3/genética , Transdução de Sinais
17.
Cell Rep ; 8(1): 177-89, 2014 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-24953651

RESUMO

Cell-cycle phase is a critical determinant of the choice between DNA damage repair by nonhomologous end-joining (NHEJ) or homologous recombination (HR). Here, we report that double-strand breaks (DSBs) induce ATM-dependent MOF (a histone H4 acetyl-transferase) phosphorylation (p-T392-MOF) and that phosphorylated MOF colocalizes with γ-H2AX, ATM, and 53BP1 foci. Mutation of the phosphorylation site (MOF-T392A) impedes DNA repair in S and G2 phase but not G1 phase cells. Expression of MOF-T392A also blocks the reduction in DSB-associated 53BP1 seen in wild-type S/G2 phase cells, resulting in enhanced 53BP1 and reduced BRCA1 association. Decreased BRCA1 levels at DSB sites correlates with defective repairosome formation, reduced HR repair, and decreased cell survival following irradiation. These data support a model whereby ATM-mediated MOF-T392 phosphorylation modulates 53BP1 function to facilitate the subsequent recruitment of HR repair proteins, uncovering a regulatory role for MOF in DSB repair pathway choice during S/G2 phase.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Histona Acetiltransferases/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Reparo de DNA por Recombinação , Animais , Proteínas Mutadas de Ataxia Telangiectasia/genética , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Linhagem Celular Tumoral , Quebras de DNA de Cadeia Dupla , Pontos de Checagem da Fase G1 do Ciclo Celular , Pontos de Checagem da Fase G2 do Ciclo Celular , Células HEK293 , Histona Acetiltransferases/genética , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos , Mutação , Fosforilação , Proteína 1 de Ligação à Proteína Supressora de Tumor p53
18.
J Invest Dermatol ; 134(4): 1083-1090, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24335898

RESUMO

Long-wave UVA is the major component of terrestrial UV radiation and is also the predominant constituent of indoor sunlamps, both of which have been shown to increase cutaneous melanoma risk. Using a two-chamber model, we show that UVA-exposed target cells induce intercellular oxidative signaling to non-irradiated bystander cells. This UVA-mediated bystander stress is observed between all three cutaneous cell types (i.e., keratinocytes, melanocytes, and fibroblasts). Significantly, melanocytes appear to be more resistant to direct UVA effects compared with keratinocytes and fibroblasts, although melanocytes are also more susceptible to bystander oxidative signaling. The extensive intercellular flux of oxidative species has not been previously appreciated and could possibly contribute to the observed cancer risk associated with prolonged UVA exposure.


Assuntos
Efeito Espectador , Melanócitos/citologia , Melanócitos/efeitos da radiação , Estresse Oxidativo , Transdução de Sinais , Animais , Sobrevivência Celular , Células Cultivadas , Técnicas de Cocultura , Ensaio Cometa , Fibroblastos/efeitos da radiação , Humanos , Queratinócitos/efeitos da radiação , Melanoma/prevenção & controle , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Neoplasias Induzidas por Radiação/prevenção & controle , Oxigênio/química , Espécies Reativas de Oxigênio , Neoplasias Cutâneas/prevenção & controle , Raios Ultravioleta , Melanoma Maligno Cutâneo
19.
Cancer Res ; 73(1): 50-61, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23135917

RESUMO

Tumors are composed of cancer cells but also a larger number of diverse stromal cells in the tumor microenvironment. Stromal cells provide essential supports to tumor pathophysiology but the distinct characteristics of their signaling networks are not usually considered in developing drugs to target tumors. This oversight potentially confounds proof-of-concept studies and increases drug development risks. Here, we show in established murine and human models of breast cancer how differential regulation of Akt by the small GTPase RhoB in cancer cells or stromal endothelial cells determines their dormancy versus outgrowth when angiogenesis becomes critical. In cancer cells in vitro or in vivo, RhoB functions as a tumor suppressor that restricts EGF receptor (EGFR) cell surface occupancy as well as Akt signaling. However, after activation of the angiogenic switch, RhoB functions as a tumor promoter by sustaining endothelial Akt signaling, growth, and survival of stromal endothelial cells that mediate tumor neoangiogenesis. Altogether, the positive impact of RhoB on angiogenesis and progression supercedes its negative impact in cancer cells themselves. Our findings elucidate the dominant positive role of RhoB in cancer. More generally, they illustrate how differential gene function effects on signaling pathways in the tumor stromal component can complicate the challenge of developing therapeutics to target cancer pathophysiology.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Transformação Celular Neoplásica/metabolismo , Células Endoteliais/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Células Estromais/metabolismo , Proteína rhoB de Ligação ao GTP/metabolismo , Animais , Carcinoma Ductal de Mama/metabolismo , Carcinoma Ductal de Mama/patologia , Feminino , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica , Humanos , Immunoblotting , Imuno-Histoquímica , Imunoprecipitação , Hibridização In Situ , Camundongos , Camundongos Transgênicos , Neovascularização Patológica/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Microambiente Tumoral/fisiologia
20.
Semin Cutan Med Surg ; 29(3): 190-5, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21051013

RESUMO

In the last 2 decades, advances in genomic technologies and molecular biology have accelerated the identification of multiple genetic loci that confer risk for cutaneous melanoma. The risk alleles range from rarely occurring, high-risk variants with a strong familial predisposition to low-risk to moderate-risk variants with modest melanoma association. Although the high-risk alleles are limited to the CDKN2A and CDK4 loci, the authors of recent genome-wide association studies have uncovered a set of variants in pigmentation loci that contribute to low risk. A biological validation of these new findings would provide greater understanding of the disease. In this review we describe some of the important risk loci and their association to risk of developing cutaneous melanoma and also address the current clinical challenges in CDKN2A genetic testing.


Assuntos
Predisposição Genética para Doença , Melanoma/genética , Neoplasias Cutâneas/genética , Genes p16 , Humanos , Fatores de Risco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...