Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
EJNMMI Phys ; 11(1): 36, 2024 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-38581561

RESUMO

PURPOSE: A 2D image navigator (iNAV) based 3D whole-heart sequence has been used to perform MRI and PET non-rigid respiratory motion correction for hybrid PET/MRI. However, only the PET data acquired during the acquisition of the 3D whole-heart MRI is corrected for respiratory motion. This study introduces and evaluates an MRI-based respiratory motion correction method of the complete PET data. METHODS: Twelve oncology patients scheduled for an additional cardiac 18F-Fluorodeoxyglucose (18F-FDG) PET/MRI and 15 patients with coronary artery disease (CAD) scheduled for cardiac 18F-Choline (18F-FCH) PET/MRI were included. A 2D iNAV recorded the respiratory motion of the myocardium during the 3D whole-heart coronary MR angiography (CMRA) acquisition (~ 10 min). A respiratory belt was used to record the respiratory motion throughout the entire PET/MRI examination (~ 30-90 min). The simultaneously acquired iNAV and respiratory belt signal were used to divide the acquired PET data into 4 bins. The binning was then extended for the complete respiratory belt signal. Data acquired at each bin was reconstructed and combined using iNAV-based motion fields to create a respiratory motion-corrected PET image. Motion-corrected (MC) and non-motion-corrected (NMC) datasets were compared. Gating was also performed to correct cardiac motion. The SUVmax and TBRmax values were calculated for the myocardial wall or a vulnerable coronary plaque for the 18F-FDG and 18F-FCH datasets, respectively. RESULTS: A pair-wise comparison showed that the SUVmax and TBRmax values of the motion corrected (MC) datasets were significantly higher than those for the non-motion-corrected (NMC) datasets (8.2 ± 1.0 vs 7.5 ± 1.0, p < 0.01 and 1.9 ± 0.2 vs 1.2 ± 0.2, p < 0.01, respectively). In addition, the SUVmax and TBRmax of the motion corrected and gated (MC_G) reconstructions were also higher than that of the non-motion-corrected but gated (NMC_G) datasets, although for the TBRmax this difference was not statistically significant (9.6 ± 1.3 vs 9.1 ± 1.2, p = 0.02 and 2.6 ± 0.3 vs 2.4 ± 0.3, p = 0.16, respectively). The respiratory motion-correction did not lead to a change in the signal to noise ratio. CONCLUSION: The proposed respiratory motion correction method for hybrid PET/MRI improved the image quality of cardiovascular PET scans by increased SUVmax and TBRmax values while maintaining the signal-to-noise ratio. Trial registration METC162043 registered 01/03/2017.

2.
J Vis Exp ; (193)2023 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-37036233

RESUMO

Fibrosis occurs in various tissues as a reparative response to injury or damage. If excessive, however, fibrosis can lead to tissue scarring and organ failure, which is associated with high morbidity and mortality. Collagen is a key driver of fibrosis, with type I and type III collagen being the primary types involved in many fibrotic diseases. Unlike conventional protocols used to immobilize other proteins (e.g., elastin, albumin, fibronectin, etc.), comprehensive protocols to reproducibly immobilize different types of collagens in order to produce stable coatings are not readily available. Immobilizing collagen is surprisingly challenging because multiple experimental conditions may affect the efficiency of immobilization, including the type of collagen, the pH, the temperature, and the type of microplate used. Here, a detailed protocol to reproducibly immobilize and quantify type I and III collagens resulting in stable and reproducible gels/films is provided. Furthermore, this work demonstrates how to perform, analyze, and interpret in vitro time-resolved fluorescence binding studies to investigate the interactions between collagens and candidate collagen-binding compounds (e.g., a peptide conjugated to a metal chelate carrying, for example, europium [Eu(III)]). Such an approach can be universally applied to various biomedical applications, including the field of molecular imaging to develop targeted imaging probes, drug development, cell toxicity studies, cell proliferation studies, and immunoassays.


Assuntos
Colágeno , Transdução de Sinais , Humanos , Colágeno/metabolismo , Fibrose , Peptídeos/metabolismo
3.
NMR Biomed ; 36(8): e4932, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-36940044

RESUMO

Nonalcoholic fatty liver disease (NAFLD) is the leading cause of chronic liver disease worldwide. Liver biopsy remains the gold standard for diagnosis and staging of disease. There is a clinical need for noninvasive diagnostic tools for risk stratification, follow-up, and monitoring treatment response that are currently lacking, as well as preclinical models that recapitulate the etiology of the human condition. We have characterized the progression of NAFLD in eNOS-/- mice fed a high fat diet (HFD) using noninvasive Dixon-based magnetic resonance imaging and single voxel STEAM spectroscopy-based protocols to measure liver fat fraction at 3 T. After 8 weeks of diet intervention, eNOS-/- mice exhibited significant accumulation of intra-abdominal and liver fat compared with control mice. Liver fat fraction measured by 1 H-MRS in vivo showed a good correlation with the NAFLD activity score measured by histology. Treatment of HFD-fed NOS3-/- mice with metformin showed significantly reduced liver fat fraction and altered hepatic lipidomic profile compared with untreated mice. Our results show the potential of in vivo liver MRI and 1 H-MRS to noninvasively diagnose and stage the progression of NAFLD and to monitor treatment response in an eNOS-/- murine model that represents the classic NAFLD phenotype associated with metabolic syndrome.


Assuntos
Metformina , Hepatopatia Gordurosa não Alcoólica , Humanos , Animais , Camundongos , Hepatopatia Gordurosa não Alcoólica/diagnóstico por imagem , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Hepatopatia Gordurosa não Alcoólica/metabolismo , Ácidos Graxos/metabolismo , Metformina/farmacologia , Metformina/uso terapêutico , Modelos Animais de Doenças , Fígado/metabolismo , Espectroscopia de Ressonância Magnética/métodos , Camundongos Endogâmicos C57BL
4.
Radiology ; 306(1): 150-159, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36040337

RESUMO

Background Liver MR fingerprinting (MRF) enables simultaneous quantification of T1, T2, T2*, and proton density fat fraction (PDFF) maps in single breath-hold acquisitions. Histopathologic correlation studies are desired for its clinical use. Purpose To compare liver MRF-derived metrics with separate reference quantitative MRI in participants with diffuse liver disease, evaluate scan-rescan repeatability of liver MRF, and validate MRF-derived measurements for histologic grading of liver biopsies. Materials and Methods This prospective study included participants with diffuse liver disease undergoing MRI from July 2021 to January 2022. Participants underwent two-dimensional single-section liver MRF and separate reference quantitative MRI. Linear regression, Bland-Altman plots, and coefficients of variation were used to assess the bias and repeatability of liver MRF measurements. For participants undergoing liver biopsy, the association between mapping and histologic grading was evaluated by using the Spearman correlation coefficient. Results Fifty-six participants (mean age, 59 years ± 15 [SD]; 32 women) were included to compare mapping techniques and 23 participants were evaluated with liver biopsy (mean age, 52.7 years ± 12.7; 14 women). The linearity of MRF with reference measurements in participants with diffuse liver disease (R2 value) for T1, T2, T2*, and PDFF maps was 0.86, 0.88, 0.54, and 0.99, respectively. The overall coefficients of variation for repeatability in the liver were 3.2%, 5.5%, 7.1%, and 4.6% for T1, T2, T2*, and PDFF maps, respectively. MRF-derived metrics showed high diagnostic performance in differentiating moderate or severe changes from mild or no changes (area under the receiver operating characteristic curve for fibrosis, inflammation, steatosis, and siderosis: 0.62 [95% CI: 0.52, 0.62], 0.92 [95% CI: 0.88, 0.92], 0.97 [95% CI: 0.96, 0.97], and 0.74 [95% CI: 0.57, 0.74], respectively). Conclusion Liver MR fingerprinting provided repeatable T1, T2, T2*, and proton density fat fraction maps in high agreement with reference quantitative mapping and may correlate with pathologic grades in participants with diffuse liver disease. © RSNA, 2022 Online supplemental material is available for this article.


Assuntos
Fígado Gorduroso , Prótons , Humanos , Feminino , Pessoa de Meia-Idade , Correlação de Dados , Estudos Prospectivos , Fígado/patologia , Imageamento por Ressonância Magnética/métodos , Fígado Gorduroso/patologia
5.
J Cardiovasc Magn Reson ; 24(1): 44, 2022 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-35864534

RESUMO

Cardiovascular magnetic resonance (CMR) is widely used for diagnostic imaging in the pediatric population. In addition to structural congenital heart disease (CHD), for which published guidelines are available, CMR is also performed for non-structural pediatric heart disease, for which guidelines are not available. This article provides guidelines for the performance and reporting of CMR in the pediatric population for non-structural ("non-congenital") heart disease, including cardiomyopathies, myocarditis, Kawasaki disease and systemic vasculitides, cardiac tumors, pericardial disease, pulmonary hypertension, heart transplant, and aortopathies. Given important differences in disease pathophysiology and clinical manifestations as well as unique technical challenges related to body size, heart rate, and sedation needs, these guidelines focus on optimization of the CMR examination in infants and children compared to adults. Disease states are discussed, including the goals of CMR examination, disease-specific protocols, and limitations and pitfalls, as well as newer techniques that remain under development.


Assuntos
Cardiopatias Congênitas , Imageamento por Ressonância Magnética , Adulto , Criança , Consenso , Cardiopatias Congênitas/diagnóstico por imagem , Cardiopatias Congênitas/cirurgia , Humanos , Lactente , Imageamento por Ressonância Magnética/métodos , Espectroscopia de Ressonância Magnética , Valor Preditivo dos Testes
6.
J Vis Exp ; (178)2021 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-34978293

RESUMO

Cardiovascular diseases are the leading causes of death worldwide. A permeable/leaky and dysfunctional endothelium is considered the earliest marker of vascular damage and thought to drive atherosclerosis. A method to identify these changes in vivo would be desirable in the clinic. Magnetic resonance imaging (MRI)-based tools and other technologies have enabled a profound understanding of the role of the endothelium in cardiovascular diseases and risk in vivo. There is, however, a need for reproducible and simple approaches for extracting quantifiable data reflective of endothelial damage from a single imaging study. A non-invasive, easy-to-implement, and quantitative MRI workflow was developed to acquire and analyze images that allow the quantification of two imaging biomarkers of arterial endothelial damage (leakiness/permeability and dysfunction). Here, the protocol describes the application of this method in the brachiocephalic artery of atherosclerotic ApoE-/- mice using a clinical MRI scanner. First, late gadolinium enhancement (LGE) and Modified Look-Locker Inversion Recovery (MOLLI) T1 mapping protocols to quantify endothelial leakage using an albumin-binding probe are described. Second, anatomic, and quantitative blood flow sequences to measure endothelial dysfunction, in response to acetylcholine are described. Importantly, the method outlined here allows the acquisition of high-spatial-resolution 3D images with large volumetric coverage enabling accurate segmentation of vessel wall structures to improve inter- and intra-observer variability and to increase reliability and reproducibility. Additionally, it provides quantitative data without the need for high-temporal resolution for complex kinetic modeling, making it model-independent and even allowing for imaging of highly mobile vessels (coronary arteries). Therefore, the approach simplifies and expedites data analysis. Finally, this method can be implemented on different scanners, can be extended to image different arterial beds, and is clinically applicable for use in humans. This method could be used to diagnose and treat patients with atherosclerosis by adopting a precision-medicine approach.


Assuntos
Aterosclerose , Gadolínio , Animais , Aterosclerose/diagnóstico por imagem , Meios de Contraste , Endotélio Vascular/diagnóstico por imagem , Humanos , Imageamento por Ressonância Magnética/métodos , Camundongos , Permeabilidade , Reprodutibilidade dos Testes
7.
Sci Rep ; 10(1): 13710, 2020 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-32792507

RESUMO

Cardiac CINE magnetic resonance imaging is the gold-standard for the assessment of cardiac function. Imaging accelerations have shown to enable 3D CINE with left ventricular (LV) coverage in a single breath-hold. However, 3D imaging remains limited to anisotropic resolution and long reconstruction times. Recently deep learning has shown promising results for computationally efficient reconstructions of highly accelerated 2D CINE imaging. In this work, we propose a novel 4D (3D + time) deep learning-based reconstruction network, termed 4D CINENet, for prospectively undersampled 3D Cartesian CINE imaging. CINENet is based on (3 + 1)D complex-valued spatio-temporal convolutions and multi-coil data processing. We trained and evaluated the proposed CINENet on in-house acquired 3D CINE data of 20 healthy subjects and 15 patients with suspected cardiovascular disease. The proposed CINENet network outperforms iterative reconstructions in visual image quality and contrast (+ 67% improvement). We found good agreement in LV function (bias ± 95% confidence) in terms of end-systolic volume (0 ± 3.3 ml), end-diastolic volume (- 0.4 ± 2.0 ml) and ejection fraction (0.1 ± 3.2%) compared to clinical gold-standard 2D CINE, enabling single breath-hold isotropic 3D CINE in less than 10 s scan and ~ 5 s reconstruction time.


Assuntos
Doenças Cardiovasculares/diagnóstico , Aprendizado Profundo , Interpretação de Imagem Assistida por Computador/métodos , Imagem Cinética por Ressonância Magnética/instrumentação , Imagem Cinética por Ressonância Magnética/métodos , Análise Espaço-Temporal , Adulto , Suspensão da Respiração , Estudos de Casos e Controles , Feminino , Humanos , Imageamento Tridimensional , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos
8.
Arterioscler Thromb Vasc Biol ; 40(9): 2159-2170, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32673527

RESUMO

OBJECTIVE: Evidence from preclinical and clinical studies has demonstrated that myocardial infarction promotes atherosclerosis progression. The impact of focal vascular inflammation on the progression and phenotype of remote atherosclerosis remains unknown. Approach and Results: We used a novel ApoE-/- knockout mouse model of sustained arterial inflammation, initiated by mechanical injury in the abdominal aorta. Using serial in vivo molecular MRI and ex vivo histology and flow cytometry, we demonstrate that focal arterial inflammation triggered by aortic injury, accelerates atherosclerosis in the remote brachiocephalic artery. The brachiocephalic artery atheroma had distinct histological features including increased plaque size, plaque permeability, necrotic core to collagen ratio, infiltration of more inflammatory monocyte subsets, and reduced collagen content. We also found that arterial inflammation following focal vascular injury evoked a prolonged systemic inflammatory response manifested as a persistent increase in serum IL-6 (interleukin 6). Finally, we demonstrate that 2 therapeutic interventions-pravastatin and minocycline-had distinct anti-inflammatory effects at the plaque and systemic level. CONCLUSIONS: We show for the first time that focal arterial inflammation in response to vascular injury enhances systemic vascular inflammation, accelerates remote atheroma progression and induces plaques more inflamed, lipid-rich, and collagen-poor in the absence of ischemic myocardial injury. This inflammatory cascade is modulated by pravastatin and minocycline treatments, which have anti-inflammatory effects at both plaque and systemic levels that mitigate atheroma progression.


Assuntos
Aortite/complicações , Aterosclerose/etiologia , Tronco Braquiocefálico/metabolismo , Mediadores da Inflamação/sangue , Placa Aterosclerótica , Animais , Anti-Inflamatórios/farmacologia , Aortite/sangue , Aortite/patologia , Aterosclerose/sangue , Aterosclerose/patologia , Aterosclerose/prevenção & controle , Tronco Braquiocefálico/efeitos dos fármacos , Tronco Braquiocefálico/patologia , Colágeno/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Interleucina-6/sangue , Metabolismo dos Lipídeos , Masculino , Camundongos Knockout para ApoE , Minociclina/farmacologia , Necrose , Pravastatina/farmacologia , Fatores de Tempo
9.
Sci Rep ; 10(1): 3231, 2020 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-32094414

RESUMO

Abdominal aortic aneurysm (AAA) remains a fatal disease. Its development encompasses a complex interplay between hemodynamic stimuli on and changes in the arterial wall. Currently available biomarkers fail to predict the risk of AAA rupture independent of aneurysm size. Therefore, novel biomarkers for AAA characterization are needed. In this study, we used a mouse model of AAA to investigate the potential of magnetic resonance imaging (MRI) with an albumin-binding probe to assess changes in vascular permeability at different stages of aneurysm growth. Two imaging studies were performed: a longitudinal study with follow-up and death as endpoint to predict rupture risk and a week-by-week study to characterize AAA development. AAAs, which eventually ruptured, demonstrated a significantly higher in vivo MR signal enhancement from the albumin-binding probe (p = 0.047) and a smaller nonenhancing thrombus area compared to intact AAAs (p = 0.001). The ratio of albumin-binding-probe enhancement of the aneurysm wall to size of nonenhancing-thrombus-area predicted AAA rupture with high sensitivity/specificity (100%/86%). More advanced aneurysms with higher vascular permeability demonstrated an increased uptake of the albumin-binding-probe. These results indicate that MRI with an albumin-binding probe may enable noninvasive assessment of vascular permeability in murine AAAs and prediction of rupture risk.


Assuntos
Albuminas/metabolismo , Aneurisma da Aorta Abdominal/diagnóstico , Aneurisma da Aorta Abdominal/fisiopatologia , Ruptura Aórtica/diagnóstico , Ruptura Aórtica/fisiopatologia , Permeabilidade Capilar , Diagnóstico por Imagem , Animais , Progressão da Doença , Gadolínio/análise , Terapia a Laser , Imageamento por Ressonância Magnética , Camundongos , Ligação Proteica , Fatores de Risco
10.
Magn Reson Med ; 79(1): 339-350, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28426162

RESUMO

PURPOSE: Develop a framework for efficient free-breathing simultaneous whole-heart coronary magnetic resonance angiography (CMRA) and cardiac positron emission tomography (PET) on a 3 Tesla PET-MR system. METHODS: An acquisition that enables nonrigid motion correction of both CMRA and PET has been developed. The proposed method estimates translational motion from low-resolution 2D MR image navigators acquired at each heartbeat and 3D nonrigid respiratory motion between different respiratory bins from the CMRA data itself. Estimated motion is used for correcting the CMRA as well as the emission and attenuation PET data sets to the same respiratory position. The CMRA approach was studied in 10 healthy subjects and compared for both left and right coronary arteries (LCA, RCA) against a reference scan with diaphragmatic navigator gating and tracking. The PET-CMRA approach was tested in 5 oncology patients with 18 F-FDG myocardial uptake. PET images were compared against uncorrected and gated PET reconstructions. RESULTS: For the healthy subjects, no statistically significant differences in vessel length and sharpness (P > 0.01) were observed between the proposed approach and the reference acquisition with navigator gating and tracking, although data acquisition was significantly shorter. The proposed approach improved CMRA vessel sharpness by 37.9% and 49.1% (LCA, RCA) and vessel length by 48.0% and 36.7% (LCA, RCA) in comparison with no motion correction for all the subjects. Motion-corrected PET images showed improved sharpness of the myocardium compared to uncorrected reconstructions and reduced noise compared to gated reconstructions. CONCLUSION: Feasibility of a new respiratory motion-compensated simultaneous cardiac PET-CMRA acquisition has been demonstrated. Magn Reson Med 79:339-350, 2018. © 2017 The Authors Magnetic Resonance in Medicine published by Wiley Periodicals, Inc. on behalf of International Society for Magnetic Resonance in Medicine. This is an open access article under the terms of the Creative Commons Attribution License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.


Assuntos
Vasos Coronários/diagnóstico por imagem , Coração/diagnóstico por imagem , Angiografia por Ressonância Magnética , Miocárdio/patologia , Tomografia por Emissão de Pósitrons , Adulto , Eletrocardiografia , Fluordesoxiglucose F18/química , Voluntários Saudáveis , Humanos , Processamento de Imagem Assistida por Computador , Pessoa de Meia-Idade , Movimento (Física) , Reprodutibilidade dos Testes , Respiração
11.
Nanotheranostics ; 1(2): 186-195, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29071187

RESUMO

A novel dual-imaging cisplatin-carrying molecular cargo capable of performing simultaneous optical and MR imaging is reported herein. This long-lasting MRI contrast agent (r1 relaxivity of 23.4 mM-1s-1 at 3T, 25 oC) is a photo-activated cisplatin prodrug (PtGdL) which enables real-time monitoring of anti-cancer efficacy. PtGdL is a model for monitoring the drug delivery and anti-cancer efficacy by MRI with a much longer retention time (24 hours) in several organs such as renal cortex and spleen than GdDOTA and its motif control GdL. Upon complete release of cisplatin, all PtGdL is converted to GdL enabling subsequent MRI analyses of therapy efficacy within its reasonably short clearance time of 4 hours. There is also responsive fluorescence enhancement for monitoring by photon-excitation.

12.
J Cardiovasc Magn Reson ; 18(1): 66, 2016 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-27716273

RESUMO

BACKGROUND: The use of gadolinium contrast agents in cardiovascular magnetic resonance is well-established and serves to improve both vascular imaging as well as enable late gadolinium enhancement (LGE) imaging for tissue characterization. Currently, gadofosveset trisodium, an intravascular contrast agent, combined with a three-dimensional inversion recovery balanced steady state free precession (3D IR bSSFP) sequence, is commonly used in pediatric cardiac imaging and yields excellent vascular imaging, but cannot be used for late gadolinium enhancement. Gadofosveset use remains limited in clinical practice, and manufacture was recently halted, thus an alternative is needed to allow 3D IR bSSFP and LGE in the same study. METHODS: Here we propose a protocol to give a bolus of 0.1 mL/kg = 0.1 mmol/kg gadobutrol (GADAVIST/GADOVIST) for time-resolved magnetic resonance angiography (MRA). Subsequently, 0.1 mmol/kg is diluted up to 5 or 7.5 mL with saline and then loaded into intravenous tubing connected to the patient. A 0.5 mL short bolus is infused, then a slow infusion is given at 0.02 or 0.03 mL/s. Image navigated (iNAV) 3D IR bSSFP imaging is initiated 45-60 s after the initiation of the infusion, with a total image acquisition time of ~5 min. If necessary, LGE imaging using phase sensitive inversion recovery reconstruction (PSIR) is performed at 10 min after the infusion is initiated. RESULTS: We have successfully performed the above protocol with good image quality on 10 patients with both time-resolved MRA and 3D IR bSSFP iNAV imaging. Our initial attempts to use pencil beam respiratory navigation failed due to signal labeling in the liver by the navigator. We have also performed 2D PSIR LGE successfully, with both LGE positive and LGE negative results. CONCLUSION: A bolus of gadobutrol, followed later by a slow infusion, allows time-resolved MRA, 3D IR bSSFP using the iNAV navigation technique, and LGE imaging, all in a single study with a single contrast agent.


Assuntos
Meios de Contraste/administração & dosagem , Angiografia Coronária/métodos , Cardiopatias Congênitas/diagnóstico por imagem , Coração/diagnóstico por imagem , Angiografia por Ressonância Magnética/métodos , Imagem Cinética por Ressonância Magnética/métodos , Compostos Organometálicos/administração & dosagem , Coração/fisiopatologia , Cardiopatias Congênitas/fisiopatologia , Humanos , Interpretação de Imagem Assistida por Computador , Imageamento Tridimensional , Infusões Intravenosas , Injeções Intravenosas , Valor Preditivo dos Testes , Estudos Retrospectivos , Fatores de Tempo
13.
Eur Radiol ; 26(3): 910-20, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26139316

RESUMO

UNLABELLED: Molecular imaging aims to improve the identification and characterization of pathological processes in vivo by visualizing the underlying biological mechanisms. Molecular imaging techniques are increasingly used to assess vascular inflammation, remodeling, cell migration, angioneogenesis and apoptosis. In cardiovascular diseases, molecular magnetic resonance imaging (MRI) offers new insights into the in vivo biology of pathological vessel wall processes of the coronary and carotid arteries and the aorta. This includes detection of early vascular changes preceding plaque development, visualization of unstable plaques and assessment of response to therapy. The current review focuses on recent developments in the field of molecular MRI to characterise different stages of atherosclerotic vessel wall disease. A variety of molecular MR-probes have been developed to improve the non-invasive detection and characterization of atherosclerotic plaques. Specifically targeted molecular probes allow for the visualization of key biological steps in the cascade leading to the development of arterial vessel wall lesions. Early detection of processes which lead to the development of atherosclerosis and the identification of vulnerable atherosclerotic plaques may enable the early assessment of response to therapy, improve therapy planning, foster the prevention of cardiovascular events and may open the door for the development of patient-specific treatment strategies. KEY POINTS: Targeted MR-probes allow the characterization of atherosclerosis on a molecular level. Molecular MRI can identify in vivo markers for the differentiation of stable and unstable plaques. Visualization of early molecular changes has the potential to improve patient-individualized risk-assessment.


Assuntos
Aterosclerose/diagnóstico , Sondas Moleculares , Artérias Carótidas/patologia , Doenças das Artérias Carótidas/patologia , Doenças das Artérias Carótidas/fisiopatologia , Endotélio Vascular/fisiopatologia , Fibrina/análise , Previsões , Humanos , Lipídeos/análise , Macrófagos/patologia , Angiografia por Ressonância Magnética/métodos , Imagem Molecular/métodos , Neovascularização Patológica/patologia , Neovascularização Patológica/fisiopatologia , Peptídeo Hidrolases/análise , Placa Aterosclerótica/patologia , Placa Aterosclerótica/fisiopatologia , Remodelação Vascular/fisiologia
14.
IEEE Trans Med Imaging ; 34(11): 2258-70, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25935031

RESUMO

Combining Positron Emission Tomography (PET) with Magnetic Resonance Imaging (MRI) results in a promising hybrid molecular imaging modality as it unifies the high sensitivity of PET for molecular and cellular processes with the functional and anatomical information from MRI. Digital Silicon Photomultipliers (dSiPMs) are the digital evolution in scintillation light detector technology and promise high PET SNR. DSiPMs from Philips Digital Photon Counting (PDPC) were used to develop a preclinical PET/RF gantry with 1-mm scintillation crystal pitch as an insert for clinical MRI scanners. With three exchangeable RF coils, the hybrid field of view has a maximum size of 160 mm × 96.6 mm (transaxial × axial). 0.1 ppm volume-root-mean-square B 0-homogeneity is kept within a spherical diameter of 96 mm (automatic volume shimming). Depending on the coil, MRI SNR is decreased by 13% or 5% by the PET system. PET count rates, energy resolution of 12.6% FWHM, and spatial resolution of 0.73 mm (3) (isometric volume resolution at isocenter) are not affected by applied MRI sequences. PET time resolution of 565 ps (FWHM) degraded by 6 ps during an EPI sequence. Timing-optimized settings yielded 260 ps time resolution. PET and MR images of a hot-rod phantom show no visible differences when the other modality was in operation and both resolve 0.8-mm rods. Versatility of the insert is shown by successfully combining multi-nuclei MRI ((1)H/(19)F) with simultaneously measured PET ((18)F-FDG). A longitudinal study of a tumor-bearing mouse verifies the operability, stability, and in vivo capabilities of the system. Cardiac- and respiratory-gated PET/MRI motion-capturing (CINE) images of the mouse heart demonstrate the advantage of simultaneous acquisition for temporal and spatial image registration.


Assuntos
Imageamento por Ressonância Magnética/métodos , Imagem Molecular/métodos , Tomografia por Emissão de Pósitrons/métodos , Animais , Desenho de Equipamento , Feminino , Fluordesoxiglucose F18 , Camundongos , Camundongos Endogâmicos BALB C , Imagem Multimodal , Imagens de Fantasmas
15.
Artigo em Inglês | MEDLINE | ID: mdl-25873720

RESUMO

BACKGROUND: Despite the beneficial effects of vascular interventions, these procedures may damage the endothelium leading to increased vascular permeability and remodeling. Re-endothelialization of the vessel wall, with functionally and structurally intact cells, is controlled by endothelial nitric oxide synthase (NOS3) and is crucial for attenuating adverse effects after injury. We investigated the applicability of the albumin-binding MR contrast agent, gadofosveset, to noninvasively monitor focal changes in vascular permeability and remodeling, after injury, in NOS3-knockout (NOS3(-/-)) and wild-type (WT) mice in vivo. METHODS AND RESULTS: WT and NOS3(-/-) mice were imaged at 7, 15, and 30 days after aortic denudation or sham-surgery. T1 mapping (R1=1/T1, s(-1)) and delayed-enhanced MRI were used as measurements of vascular permeability (R1) and remodeling (vessel wall enhancement, mm(2)) after gadofosveset injection, respectively. Denudation resulted in higher vascular permeability and vessel wall enhancement 7 days after injury in both strains compared with sham-operated animals. However, impaired re-endothelialization and increased neovascularization in NOS3(-/-) mice resulted in significantly higher R1 at 15 and 30 days post injury compared with WT mice that showed re-endothelialization and lack of neovascularization (R1 [s(-1)]=15 days: NOS3 (-/-)4.02 [interquartile range, IQR, 3.77-4.41] versus WT2.39 [IQR, 2.35-2.92]; 30 days: NOS3 (-/-)4.23 [IQR, 3.94-4.68] versus WT2.64 [IQR, 2.33-2.80]). Similarly, vessel wall enhancement was higher in NOS3(-/-) but recovered in WT mice (area [mm(2)]=15 days: NOS3 (-/-)5.20 [IQR, 4.68-6.80] versus WT2.13 [IQR, 0.97-3.31]; 30 days: NOS3 (-/-)7.35 [IQR, 5.66-8.61] versus WT1.60 [IQR, 1.40-3.18]). Ex vivo histological studies corroborated the MRI findings. CONCLUSIONS: We demonstrate that increased vascular permeability and remodeling, after injury, can be assessed noninvasively using an albumin-binding MR contrast agent and may be used as surrogate markers for evaluating the healing response of the vessel wall after injury.


Assuntos
Permeabilidade Capilar , Meios de Contraste , Endotélio Vascular/lesões , Gadolínio , Angiografia por Ressonância Magnética , Compostos Organometálicos , Remodelação Vascular , Animais , Permeabilidade Capilar/fisiologia , Endotélio Vascular/metabolismo , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Modelos Animais , Óxido Nítrico Sintase Tipo III , Remodelação Vascular/fisiologia , Vasodilatação/fisiologia
16.
Br J Pharmacol ; 172(14): 3548-64, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25824964

RESUMO

BACKGROUND AND PURPOSE: There are conflicting data regarding whether netrin-1 retards or accelerates atherosclerosis progression, as it can lead either to monocyte repulsion from or retention within plaques depending on its cellular source. We investigated the effect of aspirin, which is widely used in cardiovascular prophylaxis, on the synthesis of different isoforms of netrin-1 by endothelial cells under pro-inflammatory conditions, and defined the net effect of aspirin-dependent systemic modulation of netrin-1 on atherosclerosis progression. EXPERIMENTAL APPROACH: Netrin-1 synthesis was studied in vitro using human endothelial cells stimulated with TNF-α, with or without aspirin treatment. In vivo experiments were conducted in ApoE(-/-) mice fed with a high-fat diet (HFD), receiving either aspirin or clopidogrel. KEY RESULTS: TNF-α-induced NF-κB activation up-regulated the nuclear isoform of netrin-1, while simultaneously reducing secreted netrin-1. Down-regulation of the secreted isoform compromised the chemorepellent action of the endothelium against monocyte chemotaxis. Aspirin counteracted TNF-α-mediated effects on netrin-1 synthesis by endothelial cells through COX-dependent inhibition of NF-κB and concomitant histone hyperacetylation. Administration of aspirin to ApoE(-/-) mice on HFD increased blood and arterial wall levels of netrin-1 independently of its effects on platelets, accompanied by reduced plaque size and content of monocytes/macrophages, compared with untreated or clopidogrel-treated mice. In vivo blockade of netrin-1 enhanced monocyte plaque infiltration in aspirin-treated ApoE(-/-) mice. CONCLUSIONS AND IMPLICATIONS: Aspirin counteracts down-regulation of secreted netrin-1 induced by pro-inflammatory stimuli in endothelial cells. The aspirin-dependent increase of netrin-1 in ApoE(-/-) mice exerts anti-atherogenic effects by preventing arterial accumulation of monocytes.


Assuntos
Aspirina/farmacologia , Histonas/metabolismo , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Monócitos/citologia , Monócitos/efeitos dos fármacos , Fatores de Crescimento Neural/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Acetilação/efeitos dos fármacos , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/metabolismo , Aspirina/administração & dosagem , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Masculino , Camundongos , Camundongos Knockout , Monócitos/metabolismo , Fatores de Crescimento Neural/biossíntese , Netrina-1 , Isoformas de Proteínas/biossíntese , Isoformas de Proteínas/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Proteínas Supressoras de Tumor/biossíntese
17.
Arterioscler Thromb Vasc Biol ; 34(6): 1193-1198, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24723557

RESUMO

OBJECTIVE: Deep venous thrombosis is a major health problem. Thrombolytic therapies are effective in recanalizing the veins and preventing post-thrombotic complications, but there is no consensus on selection criteria. The aim of this study was to investigate a fibrin-specific MRI contrast agent (EP-2104R) for the accurate quantification of thrombus' fibrin content in vivo and for the identification of thrombus suitable for thrombolysis. APPROACH AND RESULTS: Venous thrombosis was induced in the inferior vena cava of 8- to 10-week-old male BALB/C mice and MRI performed 2, 4, 7, 10, 14, and 21 days later. Eighteen mice were scanned at each time point pre and 2 hours post injection of EP-2104R (8.0 µmol/kg) with 12 mice at each time point used to correlate fibrin contrast uptake with thrombus' histological stage and fibrin content. Six mice at each time point were immediately subjected to intravascular thrombolytic therapy (10 mg/kg of tissue-type plasminogen activator). Mice were imaged to assess response to lytic therapy 24 hours after thrombolytic treatment. Two mice at each time point were scanned post injection of 0.2 mmol/kg of Gd-DTPA (gadolinium with diethylenetriaminepentacetate, Magnevist, Schering AG, Berlin, Germany) for control purpose. Contrast uptake was correlated positively with the fibrin content of the thrombus measured by Western blotting (R(2)=0.889; P<0.001). Thrombus relaxation rate (R1) post contrast and the change in visualized thrombus size on late gadolinium enhancement inversion recovery MRI pre-EP-2104R and post-EP-2104R injection were the best predictors for successful thrombolysis (area under the curve, 0.989 [95% confidence interval, 0.97-1.00] and 0.994 [95% confidence interval, 0.98-1.00] respectively). CONCLUSIONS: MRI with a fibrin-specific contrast agent accurately estimates thrombus fibrin content in vivo and identifies thrombi that are amenable for thrombolysis.


Assuntos
Fibrina/análise , Imageamento por Ressonância Magnética/métodos , Terapia Trombolítica , Trombose Venosa/diagnóstico , Animais , Gadolínio , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Peptídeos , Trombose Venosa/tratamento farmacológico , Trombose Venosa/metabolismo
18.
J Magn Reson Imaging ; 39(3): 598-608, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24006053

RESUMO

PURPOSE: To investigate a very small iron-oxide particle (VSOP) in a mouse model of acute ischemia-reperfusion to access the mechanism of such particles in areas of myocardial inflammation. MATERIALS AND METHODS: Animals were injected with VSOP at several time points, in a mouse model of acute myocardial infarction (MI), before and after MI. MRI was used to localize areas of VSOP enhancement, evaluate VSOP areas extension, and determine the related T2* values. Histology, electron microscopy, macrophage counting, and Evan's Blue staining were also performed. RESULTS: We found that areas of VSOP uptake decreased from 1 to 8 days post-MI while the related T2* values increased. T2* and VSOP areas, defined from MRI data, correlated well between 1 and 3 days post-MI but not at 7 days after injection. Histological analysis and electron microscopy showed colocalization of macrophages with areas of VSOP staining. However, there was no correlation between number of macrophages and the extension of the VSOP areas achieved by MR. We found that only areas of increased permeability (assessed by Evan's Blue staining) showed colocalization of macrophages and VSOP uptake. CONCLUSION: This study demonstrates that VSOP allows the assessment of myocardial inflammation associated with increased permeability during infarct healing in a mouse model of ischemia-reperfusion.


Assuntos
Compostos Férricos/farmacologia , Imageamento por Ressonância Magnética/métodos , Infarto do Miocárdio/diagnóstico , Reperfusão Miocárdica/métodos , Animais , Meios de Contraste , Modelos Animais de Doenças , Feminino , Inflamação/patologia , Injeções Intravenosas , Camundongos , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/patologia , Infarto do Miocárdio/cirurgia , Distribuição Aleatória , Valores de Referência , Sensibilidade e Especificidade
19.
Circulation ; 128(7): 729-736, 2013 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-23820077

RESUMO

BACKGROUND: The magnetic resonance longitudinal relaxation time (T1) changes with thrombus age in humans. In this study, we investigate the possible mechanisms that give rise to the T1 signal in venous thrombi and whether changes in T1 relaxation time are informative of the susceptibility to lysis. METHODS AND RESULTS: Venous thrombosis was induced in the vena cava of BALB/C mice, and temporal changes in T1 relaxation time correlated with thrombus composition. The mean T1 relaxation time of thrombus was shortest at 7 days following thrombus induction and returned to that of blood as the thrombus resolved. T1 relaxation time was related to thrombus methemoglobin formation and further processing. Studies in inducible nitric oxide synthase (iNOS(-/-))-deficient mice revealed that inducible nitric oxide synthase mediates oxidation of erythrocyte lysis-derived iron to paramagnetic Fe3+, which causes thrombus T1 relaxation time shortening. Studies using chemokine receptor-2-deficient mice (Ccr2(-/-)) revealed that the return of the T1 signal to that of blood is regulated by removal of Fe3+ by macrophages that accumulate in the thrombus during its resolution. Quantification of T1 relaxation time was a good predictor of successful thrombolysis with a cutoff point of <747 ms having a sensitivity and specificity to predict successful lysis of 83% and 94%, respectively. CONCLUSIONS: The source of the T1 signal in the thrombus results from the oxidation of iron (released from the lysis of trapped erythrocytes in the thrombus) to its paramagnetic Fe3+ form. Quantification of T1 relaxation time appears to be a good predictor of the success of thrombolysis.


Assuntos
Fibrinólise/fisiologia , Ferro/metabolismo , Imageamento por Ressonância Magnética , Trombose Venosa/patologia , Animais , Endotélio Vascular/lesões , Eritrócitos/química , Humanos , Ligadura , Macrófagos/fisiologia , Masculino , Espectrometria de Massas , Metemoglobina/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Óxido Nítrico Sintase Tipo II/deficiência , Óxido Nítrico Sintase Tipo II/fisiologia , Oxirredução , Receptores CCR2/deficiência , Receptores CCR2/fisiologia , Fatores de Tempo , Veia Cava Inferior/patologia , Trombose Venosa/etiologia , Trombose Venosa/metabolismo
20.
Circ Cardiovasc Imaging ; 6(3): 433-440, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23564561

RESUMO

BACKGROUND: Deep vein thrombosis remains a major health problem necessitating accurate diagnosis. Thrombolysis is associated with significant morbidity and is effective only for the treatment of unorganized thrombus. We tested the feasibility of in vivo magnetization transfer (MT) and diffusion-weighted magnetic resonance imaging to detect thrombus organization in a murine model of deep vein thrombosis. METHODS AND RESULTS: Deep vein thrombosis was induced in the inferior vena cava of male BALB/C mice. Magnetic resonance imaging was performed at days 1, 7, 14, 21, and 28 after thrombus induction using MT, diffusion-weighted, inversion-recovery, and T1-mapping protocols. Delayed enhancement and T1 mapping were repeated 2 hours after injection of a fibrin contrast agent. Finally, excised thrombi were used for histology. We found that MT and diffusion-weighted imaging can detect histological changes associated with thrombus aging. MT rate (MTR) maps and percentage of MT rate (%MTR) allowed visualization and quantification of the thrombus protein content, respectively. The %MTR increased with thrombus organization and was significantly higher at days 14, 21, and 28 after thrombus induction (days 1, 7, 14, 21, 28: %MTR=2483±451, 2079±1210, 7029±2490, 10 295±4356, 32 994±25 449; PANOVA<0.05). There was a significant positive correlation between the %MTR and the histological protein content of the thrombus (r=0.70; P<0.05). The apparent diffusion coefficient was lower in erythrocyte-rich and collagen-rich thrombus (0.72±0.10 and 0.69±0.05 [×10(-3) mm(2)/s]). Thrombus at days 7 and 14 had the highest apparent diffusion coefficient values (0.95±0.09 and 1.10±0.18 [×10(-3) mm(2)/s]). CONCLUSIONS: MT and diffusion-weighted magnetic resonance imaging sequences are promising for the staging of thrombus composition and could be useful in guiding medical intervention.


Assuntos
Imagem de Difusão por Ressonância Magnética , Veia Cava Inferior/patologia , Trombose Venosa/diagnóstico , Animais , Colágeno/sangue , Meios de Contraste , Modelos Animais de Doenças , Eritrócitos/metabolismo , Estudos de Viabilidade , Fibrina/metabolismo , Gadolínio , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Peptídeos , Valor Preditivo dos Testes , Sensibilidade e Especificidade , Fatores de Tempo , Veia Cava Inferior/metabolismo , Trombose Venosa/sangue , Trombose Venosa/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA