Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
1.
Magn Reson Med ; 91(6): 2532-2545, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38321592

RESUMEN

PURPOSE: The increasing incidence of kidney diseases is a global concern, and current biomarkers and treatments are inadequate. Changes in renal tubule luminal volume fraction (TVF) serve as a rapid biomarker for kidney disease and improve understanding of renal (patho)physiology. This study uses the amplitude of the long T2 component as a surrogate for TVF in rats, by applying multiexponential analysis of the T2-driven signal decay to examine micromorphological changes in renal tissue. METHODS: Simulations were conducted to identify a low mean absolute error (MAE) protocol and an accelerated protocol customized for the in vivo study of T2 mapping of the rat kidney at 9.4 T. We then validated our bi-exponential approach in a phantom mimicking the relaxation properties of renal tissue. This was followed by a proof-of-principle demonstration using in vivo data obtained during a transient increase of renal pelvis and tubular pressure. RESULTS: Using the low MAE protocol, our approach achieved an accuracy of MAE < 1% on the mechanical phantom. The T2 mapping protocol customized for in vivo study achieved an accuracy of MAE < 3%. Transiently increasing pressure in the renal pelvis and tubules led to significant changes in TVF in renal compartments: ΔTVFcortex = 4.9%, ΔTVFouter_medulla = 4.5%, and ΔTVFinner_medulla = -14.6%. CONCLUSION: These results demonstrate that our approach is promising for research into quantitative assessment of renal TVF in in vivo applications. Ultimately, these investigations have the potential to help reveal mechanism in acute renal injury that may lead to chronic kidney disease, which will support research into renal disorders.


Asunto(s)
Lesión Renal Aguda , Insuficiencia Renal Crónica , Ratas , Animales , Imagen por Resonancia Magnética/métodos , Riñón/diagnóstico por imagen , Túbulos Renales/diagnóstico por imagen
2.
NMR Biomed ; 37(4): e5075, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38043545

RESUMEN

Renal pathologies often manifest as alterations in kidney size, providing a valuable avenue for employing dynamic parametric MRI as a means to derive kidney size measurements for the diagnosis, treatment, and monitoring of renal disease. Furthermore, this approach holds significant potential in supporting MRI data-driven preclinical investigations into the intricate mechanisms underlying renal pathophysiology. The integration of deep learning algorithms is crucial in achieving rapid and precise segmentation of the kidney from temporally resolved parametric MRI, facilitating the use of kidney size as a meaningful (pre)clinical biomarker for renal disease. To explore this potential, we employed dynamic parametric T2 mapping of the kidney in rats in conjunction with a custom-tailored deep dilated U-Net (DDU-Net) architecture. The architecture was trained, validated, and tested on manually segmented ground truth kidney data, with benchmarking against an analytical segmentation model and a self-configuring no new U-Net. Subsequently, we applied our approach to in vivo longitudinal MRI data, incorporating interventions that emulate clinically relevant scenarios in rats. Our approach achieved high performance metrics, including a Dice coefficient of 0.98, coefficient of determination of 0.92, and a mean absolute percentage error of 1.1% compared with ground truth. The DDU-Net enabled automated and accurate quantification of acute changes in kidney size, such as aortic occlusion (-8% ± 1%), venous occlusion (5% ± 1%), furosemide administration (2% ± 1%), hypoxemia (-2% ± 1%), and contrast agent-induced acute kidney injury (11% ± 1%). This approach can potentially be instrumental for the development of dynamic parametric MRI-based tools for kidney disorders, offering unparalleled insights into renal pathophysiology.


Asunto(s)
Aprendizaje Profundo , Compuestos Organofosforados , Triazoles , Animales , Ratas , Riñón/diagnóstico por imagen , Algoritmos , Imagen por Resonancia Magnética , Procesamiento de Imagen Asistido por Computador
3.
J Magn Reson Imaging ; 59(5): 1494-1513, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-37675919

RESUMEN

Owing to the increasing prevalence of diabetic mellitus, diabetic kidney disease (DKD) is presently the leading cause of chronic kidney disease and end-stage renal disease worldwide. Early identification and disease interception is of paramount clinical importance for DKD management. However, current diagnostic, disease monitoring and prognostic tools are not satisfactory, due to their low sensitivity, low specificity, or invasiveness. Magnetic resonance imaging (MRI) is noninvasive and offers a host of contrast mechanisms that are sensitive to pathophysiological changes and risk factors associated with DKD. MRI tissue characterization involves structural and functional information including renal morphology (kidney volume (TKV) and parenchyma thickness using T1- or T2-weighted MRI), renal microstructure (diffusion weighted imaging, DWI), renal tissue oxygenation (blood oxygenation level dependent MRI, BOLD), renal hemodynamics (arterial spin labeling and phase contrast MRI), fibrosis (DWI) and abdominal or perirenal fat fraction (Dixon MRI). Recent (pre)clinical studies demonstrated the feasibility and potential value of DKD evaluation with MRI. Recognizing this opportunity, this review outlines key concepts and current trends in renal MRI technology for furthering our understanding of the mechanisms underlying DKD and for supplementing clinical decision-making in DKD. Progress in preclinical MRI of DKD is surveyed, and challenges for clinical translation of renal MRI are discussed. Future directions of DKD assessment and renal tissue characterization with (multi)parametric MRI are explored. Opportunities for discovery and clinical break-through are discussed including biological validation of the MRI findings, large-scale population studies, standardization of DKD protocols, the synergistic connection with data science to advance comprehensive texture analysis, and the development of smart and automatic data analysis and data visualization tools to further the concepts of virtual biopsy and personalized DKD precision medicine. We hope that this review will convey this vision and inspire the reader to become pioneers in noninvasive assessment and management of DKD with MRI. LEVEL OF EVIDENCE: 1 TECHNICAL EFFICACY: Stage 2.


Asunto(s)
Diabetes Mellitus , Nefropatías Diabéticas , Insuficiencia Renal Crónica , Humanos , Nefropatías Diabéticas/diagnóstico por imagen , Riñón/patología , Imagen por Resonancia Magnética/métodos , Pruebas de Función Renal/métodos , Insuficiencia Renal Crónica/patología
4.
J Magn Reson Imaging ; 58(6): 1660-1679, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37243378

RESUMEN

Renal diseases pose a significant socio-economic burden on healthcare systems. The development of better diagnostics and prognostics is well-recognized as a key strategy to resolve these challenges. Central to these developments are MRI biomarkers, due to their potential for monitoring of early pathophysiological changes, renal disease progression or treatment effects. The surge in renal MRI involves major cross-domain initiatives, large clinical studies, and educational programs. In parallel with these translational efforts, the need for greater (patho)physiological specificity remains, to enable engagement with clinical nephrologists and increase the associated health impact. The ISMRM 2022 Member Initiated Symposium (MIS) on renal MRI spotlighted this issue with the goal of inspiring more solutions from the ISMRM community. This work is a summary of the MIS presentations devoted to: 1) educating imaging scientists and clinicians on renal (patho)physiology and demands from clinical nephrologists, 2) elucidating the connection of MRI parameters with renal physiology, 3) presenting the current state of leading MR surrogates in assessing renal structure and functions as well as their next generation of innovation, and 4) describing the potential of these imaging markers for providing clinically meaningful renal characterization to guide or supplement clinical decision making. We hope to continue momentum of recent years and introduce new entrants to the development process, connecting (patho)physiology with (bio)physics, and conceiving new clinical applications. We envision this process to benefit from cross-disciplinary collaboration and analogous efforts in other body organs, but also to maximally leverage the unique opportunities of renal physiology. LEVEL OF EVIDENCE: 1 TECHNICAL EFFICACY STAGE: 2.


Asunto(s)
Enfermedades Renales , Riñón , Humanos , Riñón/diagnóstico por imagen , Imagen por Resonancia Magnética/métodos , Enfermedades Renales/diagnóstico por imagen , Nefronas , Pruebas de Función Renal
5.
Magn Reson Med ; 85(1): 334-345, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32710578

RESUMEN

PURPOSE: Examine the feasibility of characterizing the regulation of renal oxygenation using high-temporal-resolution monitoring of the T2∗ response to a step-like oxygenation stimulus. METHODS: For T2∗ mapping, multi-echo gradient-echo imaging was used (temporal resolution = 9 seconds). A step-like renal oxygenation challenge was applied involving sequential exposure to hyperoxia (100% O2 ), hypoxia (10% O2 + 90% N2 ), and hyperoxia (100% O2 ). In vivo experiments were performed in healthy rats (N = 10) and in rats with bilateral ischemia-reperfusion injury (N = 4). To assess the step response of renal oxygenation, a second-order exponential model was used (model parameters: amplitude [A], time delay [Δt], damping constant [D], and period of the oscillation [T]) for renal cortex, outer stripe of the outer medulla, inner stripe of the outer medulla, and inner medulla. RESULTS: The second-order exponential model permitted us to model the exponential T2∗ recovery and the superimposed T2∗ oscillation following renal oxygenation stimulus. The in vivo experiments revealed a difference in Douter medulla between healthy controls (D < 1, indicating oscillatory recovery) and ischemia-reperfusion injury (D > 1, reflecting aperiodic recovery). The increase in Douter medulla by a factor of 3.7 (outer stripe of the outer medulla) and 10.0 (inner stripe of the outer medulla) suggests that this parameter might be rather sensitive to (patho)physiological oxygenation changes. CONCLUSION: This study demonstrates the feasibility of monitoring the dynamic oxygenation response of renal tissues to a step-like oxygenation challenge using high-temporal-resolution T2∗ mapping. Our results suggest that the implemented system analysis approach may help to unlock questions regarding regulation of renal oxygenation, with the ultimate goal of providing imaging means for diagnostics and therapy of renal diseases.


Asunto(s)
Hiperoxia , Daño por Reperfusión , Animales , Hiperoxia/diagnóstico por imagen , Hipoxia , Riñón/diagnóstico por imagen , Corteza Renal/diagnóstico por imagen , Médula Renal/diagnóstico por imagen , Imagen por Resonancia Magnética , Oxígeno , Ratas
6.
MAGMA ; 33(1): 121-130, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31797228

RESUMEN

OBJECTIVE: Design, implementation, evaluation and application of a quadrature birdcage radiofrequency (RF) resonator tailored for renal and cardiac sodium (23Na) magnetic resonance imaging (MRI) in rats at 9.4 T. MATERIALS AND METHODS: A low pass birdcage resonator (16 rungs, din = 62 mm) was developed. The transmission field (B1+) was examined with EMF simulations. The scattering parameter (S-parameter) and the quality factor (Q-factor) were measured. For experimental validation B1+-field maps were acquired with the double-angle method. In vivo sodium imaging of the heart (spatial resolution: (1 × 1 × 5) mm3) and kidney (spatial resolution: (1 × 1 × 10) mm3) was performed with a FLASH technique. RESULTS: The RF resonator exhibits RF characteristics, transmission field homogeneity and penetration that afford 23Na MR in vivo imaging of the kidney and heart at 9.4 T. For the renal cortex and medulla a SNRs of 8 and 13 were obtained and a SNRs of 14 and 15 were observed for the left and right ventricle. DISCUSSION: These initial results obtained in vivo in rats using the quadrature birdcage volume RF resonator for 23Na MRI permit dedicated studies on experimental models of cardiac and renal diseases, which would contribute to translational research of the cardiorenal syndrome.


Asunto(s)
Riñón/diagnóstico por imagen , Imagen por Resonancia Magnética/instrumentación , Isótopos de Sodio , Animales , Calibración , Diseño de Equipo , Corazón/diagnóstico por imagen , Ventrículos Cardíacos/diagnóstico por imagen , Miocardio , Fantasmas de Imagen , Ondas de Radio , Ratas , Relación Señal-Ruido , Transductores , Investigación Biomédica Traslacional
7.
MAGMA ; 33(1): 199-215, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31768797

RESUMEN

Harmonization of acquisition and analysis protocols is an important step in the validation of BOLD MRI as a renal biomarker. This harmonization initiative provides technical recommendations based on a consensus report with the aim to move towards standardized protocols that facilitate clinical translation and comparison of data across sites. We used a recently published systematic review paper, which included a detailed summary of renal BOLD MRI technical parameters and areas of investigation in its supplementary material, as the starting point in developing the survey questionnaires for seeking consensus. Survey data were collected via the Delphi consensus process from 24 researchers on renal BOLD MRI exam preparation, data acquisition, data analysis, and interpretation. Consensus was defined as ≥ 75% unanimity in response. Among 31 survey questions, 14 achieved consensus resolution, 12 showed clear respondent preference (65-74% agreement), and 5 showed equal (50/50%) split in opinion among respondents. Recommendations for subject preparation, data acquisition, processing and reporting are given based on the survey results and review of the literature. These technical recommendations are aimed towards increased inter-site harmonization, a first step towards standardization of renal BOLD MRI protocols across sites. We expect this to be an iterative process updated dynamically based on progress in the field.


Asunto(s)
Riñón/diagnóstico por imagen , Imagen por Resonancia Magnética/tendencias , Animales , Biomarcadores/metabolismo , Consenso , Técnica Delphi , Humanos , Riñón/metabolismo , Imagen por Resonancia Magnética/normas , Reproducibilidad de los Resultados , Relación Señal-Ruido , Encuestas y Cuestionarios , Investigación Biomédica Traslacional/tendencias
8.
Magn Reson Med ; 82(6): 2343-2356, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31257651

RESUMEN

PURPOSE: Cardiorenal syndrome describes disorders of the heart and the kidneys in which a dysfunction of 1 organ induces a dysfunction in the other. This work describes the design, evaluation, and application of a 4/4-channel hydrogen-1/sodium (1 H/23 Na) RF array tailored for cardiorenal MRI at 7.0 Tesla (T) for a better physiometabolic understanding of cardiorenal syndrome. METHODS: The dual-frequency RF array is composed of a planar posterior section and a modestly curved anterior section, each section consisting of 2 loop elements tailored for 23 Na MR and 2 loopole-type elements customized for 1 H MR. Numerical electromagnetic field and specific absorption rate simulations were carried out. Transmission field ( B1+ ) uniformity was optimized and benchmarked against electromagnetic field simulations. An in vivo feasibility study was performed. RESULTS: The proposed array exhibits sufficient RF characteristics, B1+ homogeneity, and penetration depth to perform 23 Na MRI of the heart and kidney at 7.0 T. The mean B1+ field for sodium in the heart is 7.7 ± 0.8 µT/√kW and in the kidney is 6.9 ± 2.3 µT/√kW. The suitability of the RF array for 23 Na MRI was demonstrated in healthy subjects (acquisition time for 23 Na MRI: 18 min; nominal isotropic spatial resolution: 5 mm [kidney] and 6 mm [heart]). CONCLUSION: This work provides encouragement for further explorations into densely packed multichannel transceiver arrays tailored for 23 Na MRI of the heart and kidney. Equipped with this technology, the ability to probe sodium concentration in the heart and kidney in vivo using 23 Na MRI stands to make a critical contribution to deciphering the complex interactions between both organs.


Asunto(s)
Corazón/diagnóstico por imagen , Procesamiento de Imagen Asistido por Computador/métodos , Riñón/diagnóstico por imagen , Imagen por Resonancia Cinemagnética , Isótopos de Sodio/química , Campos Electromagnéticos , Estudios de Factibilidad , Femenino , Humanos , Masculino , Fantasmas de Imagen , Protones , Ondas de Radio , Reproducibilidad de los Resultados , Torso/diagnóstico por imagen , Transductores
9.
Neuroimage ; 172: 562-574, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29421323

RESUMEN

The neuromatrix, or "pain matrix", is a network of cortical brain areas which is activated by noxious as well as salient somatosensory stimulation. This has been studied in mice and humans using blood oxygenation level-dependent (BOLD) fMRI. Here we demonstrate that BOLD effects observed in the murine neuromatrix in response to salient somatosensory stimuli are prone to reflect mean arterial blood pressure (MABP) changes, rather than neural activity. We show that a standard electrostimulus typically used in murine somatosensory fMRI can induce substantial elevations in MABP. Equivalent drug-induced MABP changes - without somatosensory stimulation - evoked BOLD patterns in the neuromatrix strikingly similar to those evoked by electrostimulation. This constitutes a serious caveat for murine fMRI. The regional specificity of these BOLD patterns can be attributed to the co-localization of the neuromatrix with large draining veins. Based on these findings we propose a cardiovascular support mechanism whereby abrupt elevations in MABP provide additional energy supply to the neuromatrix and other essential brain areas in fight-or-flight situations.


Asunto(s)
Presión Sanguínea/fisiología , Encéfalo/fisiopatología , Nocicepción/fisiología , Dolor Nociceptivo/fisiopatología , Animales , Mapeo Encefálico/métodos , Estimulación Eléctrica , Potenciales Evocados Somatosensoriales/fisiología , Procesamiento de Imagen Asistido por Computador , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos C57BL
10.
Magn Reson Med ; 80(2): 672-684, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29327365

RESUMEN

PURPOSE: The aim of this study was to achieve millimeter spatial resolution sodium in vivo MRI of the human eye at 7 T using a dedicated six-channel transceiver array. We present a detailed description of the radiofrequency coil design, along with electromagnetic field and specific absorption ratio simulations, data validation, and in vivo application. METHODS: Electromagnetic field and specific absorption ratio simulations were performed. Transmit field uniformity was optimized by using a multi-objective genetic algorithm. Transmit field mapping was conducted using a phase-sensitive method. An in vivo feasibility study was carried out with 3-dimensional density-adapted projection reconstruction imaging technique. RESULTS: Measured transmit field distribution agrees well with the one obtained from simulations. The specific absorption ratio simulations confirm that the radiofrequency coil is safe for clinical use. Our radiofrequency coil is light and conforms to an average human head. High spatial resolution (nominal 1.4 and 1.0 mm isotropic) sodium in vivo images of the human eye were acquired within scan times suitable for clinical applications (∼ 10 min). CONCLUSIONS: Three most important eye compartments in the context of sodium physiology were clearly delineated in all of the images: the vitreous humor, the aqueous humor, and the lens. Our results provide encouragement for further clinical studies. The implications for research into eye diseases including ocular melanoma, cataract, and glaucoma are discussed. Magn Reson Med 80:672-684, 2018. © 2018 International Society for Magnetic Resonance in Medicine.


Asunto(s)
Ojo/diagnóstico por imagen , Imagen por Resonancia Magnética/instrumentación , Imagen por Resonancia Magnética/métodos , Sodio/química , Adulto , Diseño de Equipo , Femenino , Humanos , Masculino , Fantasmas de Imagen
11.
Magn Reson Med ; 77(6): 2381-2389, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-27342430

RESUMEN

PURPOSE: Myocardial effective relaxation time T2* is commonly regarded as a surrogate for myocardial tissue oxygenation. However, it is legitimate to assume that there are multiple factors that influence T2*. To this end, this study investigates the relationship between T2* and cardiac macromorphology given by left ventricular (LV) wall thickness and left ventricular radius, and provides interpretation of the results in the physiological context. METHODS: High spatio-temporally resolved myocardial CINE T2* mapping was performed in 10 healthy volunteers using a 7.0 Tesla (T) full-body MRI system. Ventricular septal wall thickness, left ventricular inner radius, and T2* were analyzed. Macroscopic magnetic field changes were elucidated using cardiac phase-resolved magnetic field maps. RESULTS: Ventricular septal T2* changes periodically over the cardiac cycle, increasing in systole and decreasing in diastole. Ventricular septal wall thickness and T2* showed a significant positive correlation, whereas the inner LV radius and T2* were negatively correlated. The effect of macroscopic magnetic field gradients on T2* can be considered minor in the ventricular septum. CONCLUSION: Our findings suggest that myocardial T2* is related to tissue blood volume fraction. Temporally resolved T2* mapping could be beneficial for myocardial tissue characterization and for understanding cardiac (patho)physiology in vivo. Magn Reson Med 77:2381-2389, 2017. © 2016 International Society for Magnetic Resonance in Medicine.


Asunto(s)
Ventrículos Cardíacos/anatomía & histología , Ventrículos Cardíacos/diagnóstico por imagen , Interpretación de Imagen Asistida por Computador/métodos , Imagen por Resonancia Magnética/métodos , Función Ventricular Izquierda/fisiología , Adulto , Femenino , Humanos , Masculino , Tamaño de los Órganos/fisiología , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
12.
Am J Physiol Renal Physiol ; 306(8): F864-72, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24431205

RESUMEN

Iodinated contrast media (CM) have adverse effects that may result in contrast-induced acute kidney injury. Oxidative stress is believed to play a role in CM-induced kidney injury. We test the hypothesis that oxidative stress and reduced nitric oxide in tubules are consequences of CM-induced direct cell damage and that increased local oxidative stress may increase tubuloglomerular feedback. Rat thick ascending limbs (TAL) were isolated and perfused. Superoxide and nitric oxide were quantified using fluorescence techniques. Cell death rate was estimated using propidium iodide and trypan blue. The function of macula densa and tubuloglomerular feedback responsiveness were measured in isolated, perfused juxtaglomerular apparatuses (JGA) of rabbits. The expression of genes related to oxidative stress and the activity of superoxide dismutase (SOD) were investigated in the renal medulla of rats that received CM. CM increased superoxide concentration and reduced nitric oxide bioavailability in TAL. Propidium iodide fluorescence and trypan blue uptake increased more in CM-perfused TAL than in controls, indicating increased rate of cell death. There were no marked acute changes in the expression of genes related to oxidative stress in medullary segments of Henle's loop. SOD activity did not differ between CM and control groups. The tubuloglomerular feedback in isolated JGA was increased by CM. Tubular cell damage and accompanying oxidative stress in our model are consequences of CM-induced direct cell damage, which also modifies the tubulovascular interaction at the macula densa, and may therefore contribute to disturbances of renal perfusion and filtration.


Asunto(s)
Medios de Contraste/efectos adversos , Aparato Yuxtaglomerular/efectos de los fármacos , Túbulos Renales/efectos de los fármacos , Asa de la Nefrona/efectos de los fármacos , Ácidos Triyodobenzoicos/efectos adversos , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/fisiopatología , Animales , Disponibilidad Biológica , Muerte Celular/efectos de los fármacos , Retroalimentación Fisiológica/efectos de los fármacos , Técnicas In Vitro , Aparato Yuxtaglomerular/fisiología , Túbulos Renales/metabolismo , Asa de la Nefrona/metabolismo , Masculino , Óxido Nítrico/metabolismo , Óxido Nítrico/farmacocinética , Estrés Oxidativo/efectos de los fármacos , Perfusión , Conejos , Ratas , Superóxidos/metabolismo , Transcriptoma/efectos de los fármacos
13.
Invest Radiol ; 59(2): 150-164, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38157437

RESUMEN

OBJECTIVES: After the administration of gadolinium-based contrast agents (GBCAs), residual gadolinium (Gd) has been detected in a few distinct morphological structures of the central nervous system (CNS). However, a systematic, comprehensive, and quantitative analysis of the spatial Gd distribution in the entire brain is not yet available. The first aim of this study is to provide this analysis in healthy rats after administration of high GBCA doses. The second aim is to assess the spatial distributions and possible Gd colocalizations of endogenous iron (Fe), manganese (Mn), and phosphorus (P). In addition, the presence of Gd in proximity to blood vessels was assessed by immunohistochemistry. MATERIALS AND METHODS: Male rats were randomly assigned to 3 groups (n = 3/group): saline (control), gadodiamide (linear GBCA), and gadobutrol (macrocyclic GBCA) with cumulative Gd doses of 14.4 mmol/kg of body mass. Five weeks after the last administration, the brains were collected and cryosectioned. The spatial distributions of Gd, Fe, Mn, and P were analyzed in a total of 130 sections, each covering the brain in 1 of the 3 perpendicular anatomical orientations, using laser ablation coupled with inductively coupled plasma mass spectrometry. Quantitative spatial element maps were generated, and the concentrations of Gd, Fe, and Mn were measured in 31 regions of interest covering various distinct CNS structures. Correlation analyses were performed to test for possible colocalization of Gd, Fe, and Mn. The spatial proximity of Gd and blood vessels was studied using metal-tagged antibodies against von Willebrand factor with laser ablation coupled with inductively coupled plasma mass spectrometry. RESULTS: After administration of linear gadodiamide, high Gd concentrations were measured in many distinct structures of the gray matter. This involved structures previously reported to retain Gd after linear GBCA, such as the deep cerebellar nuclei or the globus pallidus, but also structures that had not been reported so far including the dorsal subiculum, the retrosplenial cortex, the superior olivary complex, and the inferior colliculus. The analysis in all 3 orientations allowed the localization of Gd in specific subregions and layers of certain structures, such as the hippocampus and the primary somatosensory cortex. After macrocyclic gadobutrol, the Gd tissue concentration was significantly lower than after gadodiamide. Correlation analyses of region of interest concentrations of Gd, Fe, and Mn revealed no significant colocalization of Gd with endogenous Fe or Mn in rats exposed to either GBCA. Immunohistochemistry revealed a colocalization of Gd traces with vascular endothelium in the deep cerebellar nuclei after gadobutrol, whereas the majority of Gd was found outside the vasculature after gadodiamide. CONCLUSIONS: In rats exposed to gadodiamide but not in rats exposed to gadobutrol, high Gd concentrations were measured in various distinct CNS structures, and structures not previously reported were identified to contain Gd, including specific subregions and layers with different cytoarchitecture and function. Knowledge of these distinct spatial patterns may pave the way for tailored functional neurological testing. Signs for the localization of the remaining Gd in the vascular endothelium were prominent for gadobutrol but not gadodiamide. The results also indicate that local transmetalation with endogenous Fe or Mn is unlikely to explain the spatial patterns of Gd deposition in the brain, which argues against a general role of these metals in local transmetalation and release of Gd ions in the CNS.


Asunto(s)
Gadolinio , Compuestos Organometálicos , Ratas , Masculino , Animales , Manganeso , Hierro , Fósforo , Gadolinio DTPA , Medios de Contraste , Encéfalo/diagnóstico por imagen
16.
Eur Heart J ; 33(16): 2007-15, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22267241

RESUMEN

In general, iodinated contrast media (CM) are tolerated well, and CM use is steadily increasing. Acute kidney injury is the leading life-threatening side effect of CM. Here, we highlight endpoints used to assess CM-induced acute kidney injury (CIAKI), CM types, risk factors, and CIAKI prevention. Moreover, we put forward a unifying theory as to how CIAKI comes about; the kidney medulla's unique hyperosmolar environment concentrates CM in the tubules and vasculature. Highly concentrated CM in the tubules and vessels increases fluid viscosity. Thus, flow through medullary tubules and vessels decreases. Reducing the flow rate will increase the contact time of cytotoxic CM with the tubular epithelial cells and vascular endothelium, and thereby damage cells and generate oxygen radicals. As a result, medullary vasoconstriction takes place, causing hypoxia. Moreover, the glomerular filtration rate declines due to congestion of highly viscous tubular fluid. Effective prevention aims at reducing the medullary concentration of CM, thereby diminishing fluid viscosity. This is achieved by generous hydration using isotonic electrolyte solutions. Even forced diuresis may prove efficient if accompanied by adequate volume supplementation. Limiting the CM dose is the most effective measure to diminish fluid viscosity and to reduce cytotoxic effects.


Asunto(s)
Lesión Renal Aguda/inducido químicamente , Medios de Contraste/efectos adversos , Compuestos de Yodo/efectos adversos , Lesión Renal Aguda/fisiopatología , Lesión Renal Aguda/prevención & control , Animales , Viscosidad Sanguínea/fisiología , Hipoxia de la Célula/efectos de los fármacos , Diuréticos/uso terapéutico , Relación Dosis-Respuesta a Droga , Tasa de Filtración Glomerular/fisiología , Humanos , Médula Renal/irrigación sanguínea , Concentración Osmolar , Ratas , Factores de Riesgo , Vasoconstricción/efectos de los fármacos
17.
Acta Physiol (Oxf) ; 237(2): e13868, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-35993768

RESUMEN

AIM: Tissue hypoxia is an early key feature of acute kidney injury. Assessment of renal oxygenation using magnetic resonance imaging (MRI) markers T2 and T2 * enables insights into renal pathophysiology. This assessment can be confounded by changes in the blood and tubular volume fractions, occurring upon pathological insults. These changes are mirrored by changes in kidney size (KS). Here, we used dynamic MRI to monitor KS for physiological interpretation of T2 * and T2 changes in acute pathophysiological scenarios. METHODS: KS was determined from T2 *, T2 mapping in rats. Six interventions that acutely alter renal tissue oxygenation were performed directly within the scanner, including interventions that change the blood and/or tubular volume. A biophysical model was used to estimate changes in O2 saturation of hemoglobin from changes in T2 * and KS. RESULTS: Upon aortic occlusion KS decreased; this correlated with a decrease in T2 *, T2 . Upon renal vein occlusion KS increased; this negatively correlated with a decrease in T2 *, T2 . Upon simultaneous occlusion of both vessels KS remained unchanged; there was no correlation with decreased T2 *, T2 . Hypoxemia induced mild reductions in KS and T2 *, T2 . Administration of an X-ray contrast medium induced sustained KS increase, with an initial increase in T2 *, T2 followed by a decrease. Furosemide caused T2 *, T2 elevation and a minor increase in KS. Model calculations yielded physiologically plausible calibration ratios for T2 *. CONCLUSION: Monitoring KS allows physiological interpretation of acute renal oxygenation changes obtained by T2 *, T2 . KS monitoring should accompany MRI-oximetry, for new insights into renal pathophysiology and swift translation into human studies.


Asunto(s)
Lesión Renal Aguda , Riñón , Ratas , Humanos , Animales , Imagen por Resonancia Magnética/métodos , Furosemida/farmacología , Hipoxia , Lesión Renal Aguda/diagnóstico por imagen , Lesión Renal Aguda/patología , Oxígeno
18.
Quant Imaging Med Surg ; 13(12): 8336-8349, 2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-38106319

RESUMEN

Background: Rhabdomyolysis (RM)-induced acute kidney injury (AKI) is a common renal disease with low survival rate and inadequate prognosis. In this study, we investigate the feasibility of chemical exchange saturation transfer (CEST) magnetic resonance imaging (MRI) for assessing the progression of RM-induced AKI in a mouse model. Methods: AKI was induced in C57BL/6J mice via intramuscular injection of 7.5 mL/kg glycerol (n=30). Subsequently, serum creatinine (SCr), blood urea nitrogen (BUN), and hematoxylin-eosin (HE) and Masson staining, were performed. Longitudinal CEST-MRI was conducted on days 1, 3, 7, 15, and 30 after AKI induction using a 7.0-T MRI system. CEST-MRI quantification parameters including magnetization transfer ratio (MTR), MTR asymmetric analysis (MTRasym), apparent amide proton transfer (APT*), and apparent relayed nuclear Overhauser effect (rNOE*) were used to investigate the feasibility of detecting RM-induced renal damage. Results: Significant increases of SCr and BUN demonstrated established AKI. The HE staining revealed various degrees of tubular damage, and Masson staining indicted an increase in the degree of fibrosis in the injured kidneys. Among CEST parameters, the cortical MTR presented a significant difference, and it also showed the best diagnostic performance for AKI [area under the receiver operating characteristic curve (AUC) =0.915] and moderate negative correlations with SCr and BUN. On the first day of renal damage, MTR was significantly reduced in cortex (22.7%±0.04%, P=0.013), outer stripe of outer medulla (24.7%±1.6%, P<0.001), and inner stripe of outer medulla (27.0%±1.5%, P<0.001) compared to the control group. Longitudinally, MTR increased steadily with AKI progression. Conclusions: The MTR obtained from CEST-MRI is sensitive to the pathological changes in RM-induced AKI, indicating its potential clinical utility for the assessment of kidney diseases.

19.
Quant Imaging Med Surg ; 13(6): 3973-3987, 2023 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-37284101

RESUMEN

Background: Diabetic kidney disease (DKD) is the leading cause of end-stage renal disease (ESRD). There are unmet needs for noninvasive diagnosis and prognosis prediction of DKD in clinical practice. This study examines the diagnostic and prognostic value of magnetic resonance (MR) markers of renal compartment volume and the apparent diffusion coefficient (ADC) for mild, moderate, and severe DKD. Methods: This study was registered at the Chinese Clinical Trial Registry Center (registration number: ChiCTR-RRC-17012687). Sixty-seven DKD patients were prospectively randomly enrolled and underwent clinical examination and diffusion-weighted magnetic resonance imaging (DW-MRI). Patients with comorbidities that affected renal volumes or components were excluded. Ultimately, 52 DKD patients were included in the cross-sectional analysis. The ADC in the renal cortex (ADCcortex), ADC in the renal medulla (ADCmedulla) and difference between ADCcortex and ADCmedulla (ΔADC) were measured using a twelve-layer concentric objects (TLCO) approach. Renal compartment volumes of the parenchyma and pelvis were derived from T2-weighted MRI. Due to lost contact or ESRD diagnosed before follow-up (n=14), only 38 DKD patients remained for follow-up (median period =8.25 years) to investigate the correlations between MR markers and renal outcomes. The primary outcomes were the composite of doubling of the primary serum creatinine concentration or ESRD. Results: ADCcortex presented superior performance in discriminating DKD with normal and declined estimated glomerular filtration rate (eGFR) over ADCmedulla, ΔADC and renal compartment volumes with an AUC of 0.904 (sensitivity of 83% and specificity of 91%) and was moderately correlated with the clinical biomarkers eGFR and proteinuria (P<0.05). The Cox survival analysis demonstrated that ADCcortex rather than ΔADC is a predictor of renal outcomes with a hazard ratio of 3.4 (95% CI: 1.1-10.2, P<0.05) independent of baseline eGFR and proteinuria. Conclusions: ADCcortex is a valuable imaging marker for the diagnosis and prediction of renal function decline in DKD.

SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda