Abstract
Major advances in biomedical imaging have occurred over the last two decades and now allow many physiological, cellular and molecular processes to be imaged non-invasively in small animal models of cardiovascular disease. Many of these techniques can be also used in humans, providing pathophysiological context and helping to define the clinical relevance of the model. Ultrasound remains the most widely used approach and dedicated high frequency systems can obtain extremely detailed images in mice. Likewise, dedicated small animal tomographic systems have been developed for magnetic resonance (MR), positron-emission tomography (PET), fluorescence imaging and computed tomography in mice. In this article we review the use of ultrasound and PET in small animal models as well as emerging contrast mechanisms in magnetic resonance such as diffusion tensor imaging, hyperpolarized MR, chemical exchange saturation transfer imaging, MR-elastography and strain, arterial spin labeling and molecular imaging.
Subject Terms: Animal Models of Human Disease, Basic Science Research, Cardiovascular Disease, Imaging
Small animal models play a crucial role in cardiovascular investigation. The last two decades have witnessed major advances in the breadth and sophistication of molecular biology techniques to better understand and characterize small animal models of cardiovascular disease. Likewise, major developments have occurred in the techniques available to image these models noninvasively. We focus in this review on advances in ultrasound, magnetic resonance and positron emission tomography (PET) to image the left ventricle (LV) in small animal models. However, many of these techniques can also be used to image the right ventricle (RV) and the vascular system in mice, including models of arterial disease, valvular disease, aneurysm and thrombosis (see supplement).1–3 In addition to providing insights into disease pathophysiology, biomedical imaging in small animal models is playing an increasingly important role in the drug development process.4
Assessment of Cardiac Function
The evaluation of systolic and diastolic function in experimental models of cardiovascular disease has relied mainly on echocardiography, both transthoracic and transesophageal, and more recently on MRI. The ability to perform echocardiography in mice was first enabled by advances in the design of ultrasound transducers for clinical use.5 Due to the small size and the rapid heart rate of rodent’s hearts (LV long axis of approximately 5–8mm in an adult mouse, heart rate of 500–700bpm depending on the strain), echocardiography in these species requires high frequency probes (at least 7MHz in rats, 10–12MHz in mice) and high frame rates (at least 120Hz), to ensure sufficient spatial and temporal resolution. Technological advances in ultrasound have allowed the development of very high frequency probes (20–50MHz), with significantly better spatial resolution. The frame rates using very high frequency probes are acceptable; a very high frame rate (close to 1000Hz) can be obtained using ECG gating. This approach, however, requires the mouse to be perfectly still and have a stable EKG tracing, and cannot be done in awake or lightly sedated animals. While the use of ultra-high frequency systems is appealing,6 accurate insights into murine pathophysiology can be obtained on standard clinical ultrasound systems.7
One particularly crucial aspect in physiological studies, especially in rodents, is the role of sedation or anesthesia. All sedatives or anesthetics used affect cardiovascular function, generally with bradycardia and negative inotropy. Measures of systolic function are steeply and positively correlated with heart rate in mice,8 whereas LV volumes are negatively correlated. Additionally, most sedatives and anesthetics do not induce a stable hemodynamic state; although investigators should strive to acquire images at a similar heart rate, such a measure may not be sufficient to eliminate the variability of measurements. Finally, different genetic murine backgrounds may display different sensitivities to anesthesia; for example, mice on a FVB background tend to become less bradycardic with isoflurane than mice on a C57BL6 background.9
Ideally, echocardiography should be performed in awake mice; this requires training the mice and is not extensively performed. Ketamine, contrary to most sedatives, has a tachycardic effect, at least at low doses, but induces a very light sedation. 2,2,2-tribromoethanol has little cardiovascular effect, however, it may induce peritonitis when injected serially. Isoflurane is extensively used by investigators, and in spite of some individual and temporal variability, it may be an acceptable compromise. The impact of anesthesia in mice is discussed in more detail elsewhere.9 Since hemodynamic conditions are crucial to cardiac interpretation, the type and dose of sedation/anesthesia used, and the heart rate at which the images were obtained should ideally be reported.
Early papers took advantage of the high spatial resolution and sample rate of M Mode to validate the calculation of LV mass with necropsy measurements,5 and pharmacological manipulations to document changes in pressure-dimensions in mice.10, 11 The morphological and functional measurements derived from M Mode are extensively used to this day (Figure 1), and are often sufficient to report differences in LV dimensions, wall thickness or fractional shortening produced by a genetic modification or a model affecting global LV function. However, M Mode is not appropriate in any model involving regional wall motion abnormalities, such as ischemic models or myocarditis. Calculating the LV ejection fraction (EF) from the M-Mode is extensively done and reported, however, makes an assumption that the LV is a sphere (cubed formula). This assumption is not only incorrect, but the magnitude of the error also increases in large and remodeled LVs, making comparison between groups potentially fraught with error. This error can be avoided by using two-dimensional images and a modified Simpson’s rule approach to measuring EF.
Figure 1: Functional and hemodynamic imaging in mice.

(A) M-Mode obtained from a parasternal short axis view. Vertical scale: each bar 0.1mm, horizontal scale: each bar 0.1sec. (B) Parasternal Long axis view and (C) Parasternal short axis view, midpapillary level. LV: left ventricle, RV: right ventricle, AW: anterior wall (anteroseptum), PW: posterior wall (inferolateral). (D). Pulsed Doppler of the mitral inflow velocities obtained from an apical 4 chamber view. Vertical scale: each bar 30cm/s, horizontal scale: each bar 0.1sec. (E). Pulsed Doppler of the lateral mitral annular velocities obtained from an apical 4 chamber view. Vertical scale: each bar 6 cm/s, horizontal scale: each bar 0.1sec. (F) Two-dimensional view of a parasternal short axis view at the level of the aortic and pulmonary valves. Ao: aortic valve, PA: pulmonary artery, RA: right atrium, LA: left atrium, RVOT: right ventricular outflow tract. (G) correct positioning of the Doppler sample. (H) Doppler tracing of the flow at the pulmonary valve level. PAT: pulmonary acceleration time, PET: pulmonary ejection time.
The spatial resolution of two-dimensional imaging has improved using very high frequency probes (Figure 1) and allows precise measurement of 2D-derived LVEF. A single plane measurement in the parasternal long axis view using a single method of disks, an area-length method, or speckle tracking, correlates reasonably with MRI volumes and EF.12, 13 Acquiring and measuring three (base, mid, apex) or multiple short axis views, separated by a known distance may be more precise but is more time consuming.13–15 The 3D reconstruction of the LV from short axis views has been applied both to transthoracic and transesophageal echocardiography and can allow measurement of LV and RV volumes,14–18 however, they are time consuming, and rarely used. This method can also be useful in measuring a wall motion score index, or a percentage of akinetic myocardium, which reflects MI scar in non reperfused myocardial infarction models.19
Cine MRI of the murine heart can be performed on dedicated small animal scanners. It provides tomographic images with high spatial and temporal resolution, allowing the anatomy and function of both the LV and RV to be assessed.20, 21 The high flow of blood in the murine heart creates excellent contrast between the blood pool and myocardium via a spin-refreshment (inflow) effect. However, due to its cost, limited availability, and need for moderate to deep anesthesia, cardiac MRI is less frequently used to measure LV volumes and EF in mice than echocardiography. The strengths and limitations of several imaging modalities for the assessment of cardiovascular disease in mice are provided in Table 1.
Table 1:
Selection of Modality for Imaging Small Animal Models of Cardiovascular Disease.
| Ultrasound | MRI | SPECT/PET | Optical (In Vivo) | |
|---|---|---|---|---|
| Hardware | Point of care | Best performed with dedicated small animal scanners | Most systems are integrated with micro-CT scanners | Intravital microscopy, fluorescence tomography, optoacoustic systems |
| Complexity | Low complexity, no danger, no barriers to use | Requires significant expertise | Requires controlled environment and radio-pharmacy | Noninvasive tomographic imaging possible in NIR range. |
| Anesthesia/Sedation | Can be avoided | Required | Required | Required |
| Anatomy | Good | Excellent. No contrast agent needed. LV, RV, atria, aorta and PA all well visualized | Excellent if integrated with CT | Emerging |
| Function | Excellent, can be performed in awake animals (physiology not perturbed by sedation) | Excellent but requires anesthesia. LV and RV well visualized and quantified | ECG-Gated SPECT and PET feasible in mice | Volumetric optoacoustic techniques emerging |
| Strain | Excellent | Excellent, several mechanisms/techniques | - | Emerging |
| Perfusion | Excellent | Excellent | Gold Standard | Excellent |
| Viability | Not routine | Gold Standard - LGE | Gold Standard −18FDG | Emerging |
| Hemodynamics | Excellent | Excellent | - | Emerging |
| Elastic Properties | Emerging | Emerging | - | Emerging |
| Metabolism | - | Gold Standard | Gold Standard | Emerging |
| Microstructure | Not routine | Excellent | - | Excellent |
| Molecular Imaging (intra-vascular) | Excellent | Excellent | Excellent | Excellent |
| Molecular Imaging (extra-vascular) | - | Excellent | Excellent | Excellent |
CT = computed tomography, NIR = near infrared, PA = pulmonary artery, LGE = Late Gadolinium Enhancement, FDG = Fluorodeoxyglucose.
T1, T2 and Late Gadolinium Enhancement
The development of late gadolinium enhancement (LGE) has allowed infarct size in mice and rats to be precisely quantified.22, 23 Gadolinium chelates accumulate passively and transiently in the extracellular space and their detection is context specific. In an acute infarct LGE results from the disruption of the cardiomyocyte membrane,24 while in a chronic infarct LGE is produced by expansion of the extracellular space due to fibrosis.25 LGE in the murine heart correlates extremely well with TTC and histological stains for fibrosis.22, 23 The acquisition of LGE images in mice can be performed in one of two ways.23 The first involves the use of a 180° inversion prepulse followed by a delay (inversion time) for the magnetization in healthy myocardium to null. The rapid heart rate and flow of blood in the murine heart also allow LGE images to be obtained by acquiring cine images with a very high (T1-weighted) flip angle.23
T2-weighted MR imaging is sensitive to the presence of edema and has been used to measure the area-at-risk (AAR),26 although there has been some debate in the clinical literature about the exact pathophysiological significance of elevated T2 (transverse magnetic relaxation time).27 In the murine heart conventional T2-weighted imaging is challenging to perform. T2-preparation prepulses, however, have been used in mice to detect areas of edema,28, 29 which correlate well with the AAR by histology or microsphere injection.28, 29 The longitudinal relaxation time (T1) in the myocardium is also increased by edema and can be used to detect acute injury.30 In addition to measuring the AAR, T1 mapping in mice may also be able to provide a measure of microvascular function.31
While measuring the T2 of the myocardium in the murine heart is more challenging than in humans, T1 mapping is in many ways simpler. If the mouse’s heart rate is maintained above 500 beats per minute then the exponential T1 recovery curve can be sampled at least every 120ms to derive an accurate T1 value.24, 32 More sophisticated approaches, such as the modified Look-Locker Imaging or MOLLI sequence can be used in mice,33 but are vital in humans because of their slower heart rate. In clinical practice a normal reference range for T1 values must be acquired on each individual scanner.34 This is fortunately not necessary in small animal imaging provided the active and control groups are imaged with the same sequence and under the same experimental conditions.
The development of T1 mapping has been particularly useful in the characterization of murine models of pressure overload and cardiomyopathy.35–37 If T1 maps of the myocardium are acquired before the injection of gadolinium and at several time points after its injection, the extracellular volume fraction (ECV) of the myocardium can be derived.34 The ECV value is derived from the slope of R1 (1/T1) in the myocardium/R1 in the blood with a correction factor for hematocrit.34 The ECV of the murine heart is similar to humans (24–26%),35, 36 and is increased in the presence of LV fibrosis.35–37 It should be stressed, however, that an increase in ECV is not specific for fibrosis since ECV can also be increased by cell death and inflammation in acute injury and by chronic process such as amyloid.34
Imaging the Viscoelastic Properties of the Myocardium
In mice the assessment of diastole is hampered by the high heart rate and frequent fusion of the E and A wave of mitral inflow. There are no optimal ways to avoid this fusion without decreasing the heart rate below physiological values. Careful titration of isoflurane decreases the heart rate, however, inter-mice variability still exists and fusion can be observed in some mice with a heart rate as low as 400 bpm. Furthermore, the simultaneous negative inotropic effect of increasing isoflurane is itself variable. Ivabradine, an inhibitor of the If current, increases the duration of the spontaneous depolarization of the sinus node thus selectively decreases the heart rate without negative inotropic effects. In mice, ivabradine decreases the heart rate by as much as 20%.38–40 Echocardiography performed 6 hours after an intraperitoneal injection of 3μg/g body weight ivabradine (total of 2 injections in 24 hours) in a model of sepsis, reported a decrease in the heart rate from 601 (547–612) bpm to 447 (430–496) bpm, without changes in LV ejection fraction or anterior wall strain rate.38 However, intraperitoneal injection of ivabradine a few minutes prior to echocardiography is not recommended as this results in great heart rate variability, both between the mice and over time.
Hemodynamics in the murine heart can be readily assessed with Doppler imaging. Most investigators report the mitral inflow pattern (E/A), the amplitude of the medial mitral annular early diastolic velocity (e’), and the ratio of E/e’ as a parameter of left atrial pressure (Figure 1) to assess diastolic function by echo. Although e’ and E/e’ are important parameters of diastole, the analysis of diastolic dysfunction in humans also includes measurement of the left atrial volume index and of the tricuspid regurgitation maximum velocity.41 Rodents very rarely develop tricuspid regurgitation even in models of pulmonary hypertension. Investigators have reported left atrial volume index using the antero-posterior dimension in the parasternal long axis view and the mediolateral dimension in the parasternal short axis view, then calculating the LA volume using a prolate ellipse formula, and normalizing by body weight.42, 43 The LA volume index increased with age and displayed fair correlations with invasive measures of diastolic function.43 Other investigators have reported LA area measured on the 4-chamber view and demonstrated increases in this parameter in models of chronic volume overload (AAV fistula), DOCA-salt,44 and TAC.45
The measurement of hemodynamic indices in the murine heart is feasible with phase-contrast MRI,46, 47 however, MRI has been used more extensively to assess the material properties of the myocardium. MRI-based elastography of the heart is highly analogous to ultrasound-based shear wave elastography.48 An acoustic wave from an external device is directed towards the heart and its propagation through the myocardium can be used to calculate myocardial stiffness.49 In brief motion sensitizing magnetic gradients (small subsidiary magnetic fields) are applied along each Cartesian axis and phase difference images are acquired.49, 50 The term phase here refers to the phase (rotational angle) of the magnetization and not to the physiological phase of the cardiac cycle. A Fourier transform of the phase-difference image produces a complex wave image in each direction from which myocardial stiffness can be calculated. While highly accurate and very promising, the technique in mice is challenging and still fairly time consuming.50, 51
The stiffness of the myocardium can also be estimated using ultrashort TE (UTE) MRI,52 and a contrast mechanism known as T1-rho,53 without the need to inject an exogenous contrast agent. The echo-time (TE) in MRI refers to the time taken to refocus the magnetization and is not related to echocardiography. To detect the magnetization it must be flipped out of the longitudinal plane and into the transverse plane (orthogonal to the main magnetic field). Once in the transverse plane the magnetization decays exponentially with a time constant (T2) determined by the properties of the tissue. In stiff materials such as bone and tendon the T2 is extremely short and the signal cannot be detected with a conventional MR approach. UTE sequences have, therefore, been developed to image these tissues and can also be applied to detect fibrosis in the heart.52 In fibrotic/stiffer myocardium signal will be present in the UTE image but not in an image acquired with a conventional TE. In contrast, in normal myocardium substantial signal will be present in both images. This dual echo UTE (DUTE) approach has been implemented in mice with myocardial infarction with encouraging results.52
T1-rho is an advanced MR contrast mechanism that has been shown to be sensitive to cardiac fibrosis.53 Unlike conventional MRI, where a single short radiofrequency pulse is used to tip the magnetization from the longitudinal plane into the transverse plane, with T1-rho imaging a continuous radiofrequency pulse is applied parallel to the magnetization in the transverse plane.53 This technique, known as spin-locking, prevents the loss of magnetization via T2 decay. Instead, the magnetization undergoes precession in the transverse plane with a frequency in the kHz range. Molecular moieties with vibrational frequencies in the kHz range exchange energy with these protons, resulting in the decay of the T1-rho signal. This energy exchange mechanism occurs at a very different range of frequencies to those involved in T1 relaxation (recovery of longitudinal magnetization). In fibrotic myocardium T1-rho is prolonged and the technique may be able to detect both local and diffuse fibrosis.53 T1-rho imaging of the heat has been successfully performed in large animals and humans,54, 55 and its implementation in mice should be feasible.
Imaging of Myocardial Metabolism
Magnetic resonance can be performed on several atoms including protons (1H), fluorine-19 (19F), carbon-13 (13C) and phosphorus-31 (31P). Spectroscopy of (31P) can be used to quantify high-energy phosphates such as ATP and phosphocreatine,56, 57 and (13C) to characterize key intermediates in cardiac metabolism.58–62 The rate of ATP synthesis can even be imaged in the beating mouse heart.63 The abundance of (31P) and (13C) in in the body is far lower than protons, which limits their detection. However, this can be addressed using hyperpolarization,58 which dramatically increases the signal or polarization of these atoms. In brief, by cooling a suitable molecule such as pyruvate down to 4° Kelvin, while applying energy in the microwave range, the magnetic polarization can be increased by >10,000-fold.58 At this level of polarization (13C) images or spectra can be obtained rapidly and repeated longitudinally to provide a dynamic picture of metabolism (Figure 2). The principal limitation of the approach is that once the specimen is heated for injection the degree of hyperpolarization rapidly dissipates. Imaging must, therefore, be performed immediately after sample heating, which creates technical challenges and significant logistical complexity. Nevertheless, valuable insights into metabolism in the heart have been obtained with hyperpolarized MR in small animal models and humans.58–62
Figure 2: Imaging of myocardial metabolism.

(A) MRI of rat hearts after the injection of hyperpolarized (13C)-pyruvate. Maps of pyruvate and its downstream metabolites (bicarbonate, lactate, pyruvate hydrate, alanine) are shown with no suppression (NS) of the blood pool signal or with the suppression of flow (FS) in the blood pool. Fed rats injected with the pyruvate dehydrogenase kinase inhibitor (dichloroacetate, DCA) show an increase in myocardial bicarbonate. Reproduced with permission.59 (B) (13C)-lactate signal produced by macrophages infiltrating a healing infarct in a rat heart. When macrophages and monocytes are depleted a substantial reduction in signal is seen. Reproduced with permission.60 (C) Injection of hyper-polarized (13C)-fumarate in control and infarcted rats. Conversion to (13C)-malate is seen in the infarct. Reproduced with permission.61 (D) Chemical exchange saturation transfer (CEST) imaging of myocardial creatine content. In mice fed a high-fat diet the CEST creatine signal is decreased. Reproduced with permission.65
The proton pool can be divided into those protons that spin at the resonance frequency of water (on-resonance) and those that belong to chemical moieties such as amides that are shifted slightly off-resonance. There has been significant interest in exploiting the transfer of energy between these two pools using a technique known as chemical exchange saturation transfer (CEST) imaging.64 CEST detects the transfer of energy from the off-resonance to the on-resonance proton pool, which reduces the on-resonance signal. Off-resonance species of interest in the heart include creatine,64, 65 which could provide a proton-based assay of cardiac energetics and avoid the cost and complexity of (31P) and (13C) imaging. However, several technical challenges must be addressed when performing CEST imaging of the heart. Meticulous attention must be paid to achieving as uniform a main magnetic (B0) field as possible by positioning the heart at the isocenter of the magnet and paying close attention to field shimming. Notwithstanding the technical challenges, in vivo CEST imaging of the heart has been performed in small animal models,65 (Figure 2) and holds significant promise.
The imaging of myocardial metabolism with (11C)-based PET has been performed in mice,66 and is analogous to hyper-polarized MRI of (13C) with several important distinctions. The superb sensitivity of PET allows very low doses of a (11C)-labeled metabolite to be injected, which may better reflect normal physiology. However, kinetic analysis of the (11C) PET signal provides limited information on the flux of the molecule through various intermediates in a metabolic pathway. The cost and complexity of PET (cyclotron, radiopharmacy) also limit its broader application in mice and optical techniques, including fluorescence and Cerenkov luminescence, may allow some aspects of myocardial energetics to be assayed with significantly less complexity.67
Myocardial Perfusion
Anatomical imaging of the coronary arteries to detect congenital abnormalities or stenoses is feasible in mice using micro-CT,68 and perfusion imaging, using contrast echocardiography or MRI, can delineate the area-at-risk or infarction size in myocardial ischemia and infarction models.69–71 Investigators have used pulsed Doppler with a sample placed on the left coronary artery in the parasternal short axis or long axis view to measure coronary flow velocities, at rest and with vasodilators (adenosine or isoflurane), and calculate the coronary flow reserve.72–74 Semi-quantitative perfusion imaging has also been validated using myocardial contrast echocardiography.75, 76 The coronary reserve in healthy young wild-type mice is approximately 2.5, and is decreased in aged or obese mice.72, 76, 77 Myocardial perfusion imaging in mice using echocardiography has given excellent signal using probes at frequencies of 10–15 MHz, as these frequencies are close to the resonance frequency of commercially available microbubbles (Figure 3). Microbubbles can also be decorated with ligands to adhesion molecules, selectins and von Willebrand factor to detect endothelial abnormalities during reperfusion.78 Stressing the mouse heart can be challenging as cardiac sympathetic tone predominates at rest at room temperature.79, 80 Nevertheless, investigators have been able to unmask subtle functional cardiac abnormalities using stress echocardiography. Pharmacological stress, such as dobutamine has been used,76, 81 but recently echocardiography has also been performed immediately after peak exercise on a treadmill, which may provide more clinically relevant results.82, 83
Figure 3: Imaging of perfusion and strain in the murine heart.

(A, B) Measurement of perfusion in a healthy mouse with microbubbles. (A) Image acquired immediately after high energy pulses and (B) after replenishment of the myocardium with contrast. LV: left ventricle, RV: right ventricle, AW: anterior wall, PW: posterior wall. (C) Arterial spin labeling in mice fed a standard diet (SD) and a high fat high sucrose diet (HFHSD), which decreases myocardial perfusion. Reproduced with permission.91 (D) Arterial spin labeling in a mouse with a myocardial infarction showing a dramatic reduction of myocardial blood flow in the infarct zone. Reproduced with permission.32 (E) Echo microbubbles targeted to Von-Willebrand factor are retained in injured myocardium after ischemia-reperfusion. Reproduced with permission.78 Radial strain tracings obtained from the parasternal long axis view. The upper panel shows the endocardial and epicardial tracing, the lower panel the strain-time curve during one cardiac cycle. Horizontal scale: each bar 0.1sec. LV: left ventricle, RV: right ventricle, AW: anterior wall, PW: posterior wall. (G-I) DENSE imaging of mouse with myocardial infarct. (G) Late gadolinium enhancement of infarct zone. (H) Displacement map of each voxel in systole. Remote zone – black arrows, infarct zone red arrows. (I) The primary eigenvector (direction) of strain in the remote zone is radial, as indicated by the orientation of the black lines, but is highly perturbed in the infarct zone. (G-I) Reproduced with permission.105 (J) Circumferential strain by DENSE in an infarcted mouse is highly reduced in the infarct zone (arrows). Reproduced with permission.108
PET imaging provides several approaches to image myocardial perfusion including [13N]-ammonia, [82Rb]-Rubidium and [15O]-water. The radioactive half-life of [15O] is extremely short, which makes it suited only to highly specialized PET centers. [82Rb] is produced by a generator and does not require a cyclotron for its production, providing a useful option to sites without one. [82Rb] perfusion imaging has been successfully performed in rats with myocardial infarction and amiodarone therapy.84, 85 Likewise, [13N]-ammonia has been used to image perfusion in mice.86 [18F]-Flurpiridaz is a novel PET tracer that binds to mitochondrial complex 1 in a flow dependent manner, but its use in rodents has been limited.
The evaluation of myocardial perfusion with MRI can be performed using endogenous and exogenous contrast mechanisms. The two most widely used endogenous approaches involve arterial spin labeling (ASL) and BOLD (blood oxygen level dependent) imaging. The physical basis of BOLD lies in the paramagnetic properties of deoxyhemoglobin (deoxyHb), which serves as the endogenous tracer and affects T2 and T2*.87 An increase in myocardial work increases the production of deoxyHb but this is offset by an increase in myocardial blood flow/volume, which decrease deoxyHb concentration. The principal limitation of BOLD imaging lies in the relative (stress/rest ratio) nature of the measurement. The technique can detect an increase in myocardial blood flow in humans/large animals from a breath-hold induced rise in myocardial CO2,88, 89 but the feasibility of this in rodents is unclear.
ASL classically relies on the upstream inversion of protons, which affect T1 in a downstream tissue in a flow dependent manner.90 In the heart, however, the approach most widely used is to measure T1 in a section of the myocardium after applying slice-selective and non-selective inversion prepulses.90 In the slice-selective case only the spins in a thin slice of interest (~1mm thick) are inverted, whereas in the non-selective case the spins in the entire heart are. In the presence of robust myocardial blood flow a significant difference in T1 in the slice of interest will be seen with the selective vs. non-selective prepulses, allowing myocardial blood flow (ml/g/min) to be calculated (Figure 3).32, 90, 91 ASL in mice has shown that their resting myocardial blood flow is greater than 5ml/g/min (Figure 3),32, 91 which is substantially higher than humans. Fist pass perfusion with Gd in the rodent heart can be performed but requires the images to be acquired extremely rapidly. Standard techniques, such as Fermi deconvolution, can be used to derive a quantitative value of perfusion (ml/g/min).92
Myocardial Mechanics
Myocardial deformation indices (strain and strain rate) can be imaged by echocardiography (Figure 3) and have the ability to detect subtle and early abnormalities of systolic function and to predict subsequent major adverse cardiac events in a variety of cardiovascular pathologies. Tissue Doppler imaging (TDI) allows the high temporal sampling of tissue velocities in a direction parallel to the beam (radial direction on the parasternal views). Strain rate can then be integrated from the velocities.93 Radial strain rate obtained from TDI was the first parameter to be validated in mice using microsonometry,94 and was shown to predict mortality in a mouse model of doxorubicin cardiotoxicity.95 Speckle tracking follows the pixels on 2 dimensional images and allows the detection of myocardial motion independent of its direction. Strain can be integrated from myocardial motion.96 The temporal resolution of speckle tracking depends on the frame rate and the algorithm applied. Speckle tracking derived strain was also shown to decrease early and predict outcome in models of myocardial infarction, pressure overload and sepsis.97–100 The strain values reported in the literature for healthy 2 months old C57BL6 mice are quite comparable to those of humans (approximately −15 to −20% for longitudinal strain, 32–38% for radial strain, 29–30 for circumferential strain).97, 100 The strain rate reported using tissue Doppler imaging however is higher than that reported by speckle tracking (20–27/sec,94, 95, 101 vs. 8–9/sec,97, 100), raising the issue of an insufficient frame rate on the 2 dimensional images.102
Myocardial strain in rodents can be imaged using several MRI-based approaches. These include feature tracking, velocity-encoding, tagged cine imaging and a technique known as displacement encoded stimulated echo (DENSE) imaging (Figure 3). While feature tracking is extremely convenient,103 the most rigorous 3D approaches involve the use of tagging and DENSE.104–106 MR tagging involves magnetic labeling of protons in the transverse plane to eliminate their signal by a process known as saturation. The tags persist for several hundred milliseconds, which in a mouse is more than sufficient to measure strain across the entire cardiac cycle. The principal limitation of tagging is that the spatial resolution of the tag lines is substantially lower than the resolution routinely obtainable with MRI.107 The post-processing of tagged cines is also not straightforward and dedicated analysis software is required to do this.
DENSE imaging overcomes many of the limitations of MR tagging. It produces strain images of the heart at high spatial resolution and has been extensively validated in mice.105, 106 DENSE uses the stimulated echo refocusing mechanism (three successive 90° excitation pulses) and incorporates motion-sensitizing gradients after the first and third pulses. Of note, the polarity (direction) of the two motion-sensitizing gradients is opposite. If a proton remains stationary the effect of the first motion-sensitizing gradient on its phase (rotational angle of magnetization) will be completely reversed by the second motion-sensitizing gradient. However, if the proton undergoes translation between the two motion-sensitizing gradients the effects on its phase are not equal and a net phase difference results. The motion of the protons in each voxel can be calculated from the phase difference that accrues during the DENSE sequence in that voxel. 3D DENSE imaging has shown that the primary eigenvector (predominant direction) of myocardial strain is radial and that the secondary and tertiary eigenvectors of strain both have significant circumferential and longitudinal components (Figure 3).106 DENSE maps of myocardial strain can be easily co-registered with corresponding images of LGE, perfusion, metabolism and inflammation to provide a detailed phenotypic picture of the heart.108
Myocardial Microstructure
The diffusion of water in a tissue can be used to derive several useful metrics of its microstructure.109 Water diffuses most readily along the long axis of the cardiomyocytes and least freely orthogonal to them.110 Diffusion tensor imaging (DTI) can be used to resolve the preferred directions (eigenvectors) of diffusion in the heart and hence its microstructure.109, 110 Most DTI studies of the heart in mice and rats have been performed ex vivo,111–114 but with specialized high-performing gradient systems and sequences, the technique can be performed in vivo.29
Several useful scalar terms can be derived from the amount (eigenvalue) of diffusion along each eigenvector. The mean diffusivity (MD) reflects the average of the three eigenvalues and increases in the myocardium when there is an increase in its ECV and water content due to edema and/or cardiomyocyte death.29, 111 In mice serial in vivo DTI has shown that MD remains elevated for approximately 3 weeks after ischemia-reperfusion.29 The fractional anisotropy (FA) of the myocardium is determined from the ratios of the diffusion eigenvalues and reflects the degree of tissue anisotropy and coherence. FA is reduced in acute injury, often corresponding with an increase in MD,29, 111 but it is also perturbed in conditions associated with cardiomyocyte disarray such as hypertrophic cardiomyopathy (HCM).115
The orientation of cardiomyocytes in the heart is reflected by the primary eigenvector of the diffusion tensor and is often described in terms of its helix angle (HA).110 This is defined as the angle the primary eigenvector makes with the local radial plane of the heart (Figure 4).116 HA evolves from roughly 60° in the subendocardium to −60° in the subepicardium and is highly conserved across mammalian species.29, 113, 116 In all mammals the cardiomyocytes in the midmyocardial are circumferential while those in the subendocardium and subepicardium are oriented obliquely (Figure 4).110, 114, 116 The term HA is in some ways a misnomer since the cardiomyocytes in the heart are arranged in a 3D syncytium and do not form mechanically continuous helices, such as DNA. Nevertheless it is extremely useful to integrate the primary eigenvectors in each voxel into streamlines or tracts that provide a comprehensive 3D representation of cardiac microstructure.29, 113, 116, 117 These tracts can be classified by the HA in each segment or by the tractographic propagation angle (PA). The PA describes the angle between adjacent segments in a tract and is generally < 4°/voxel (Figure 4).117 Microstructural disorder causes PA to increase and the metric has been used to detect areas of myocardial injury and electrical instability with a high degree of accuracy.117
Figure 4: Microstructure of the heart.

The microstructure of the rodent heart can be characterized with diffusion tensor MRI. (A) The arrangement of cardiomyocytes in the heart can be described by the angle, termed the helix angle (HA), they make with the local radial plane of the ventricle. The HA in the heart ranges from 60° in the subendocardium to −60° in the subepicardium. Reproduced with permission.116 (B) The orientation of myocytes in the heart is highly ordered and allows them to be integrated into virtual streamlines or tracts. The propagation angle (PA) describes the angle between adjacent segments in these tracts and is <4°/voxel in most of the LV. Reproduced with permission.117 (C, D) Cardiomyocyte orientation tracts in the lateral wall of the rat and human heart, respectively, color-coded by HA. Myocardial microstructure is highly conserved across species and is extremely similar in rodent and human hearts. Reproduced with permission.116 (E) Cardiomyocyte orientation tracts in the mouse heart color-coded by PA. The PA in the LV is increased at the right ventricular insertion points but is otherwise fairly homogeneous. (F) Late gadolinium enhancement image of a mouse with a myocardial infarct. (G) PA in the infarct increases substantially and a PA threshold of 4° allows the infarct to be accurately detected. (E-G) Reproduced with permission.117 (H-K) Rat heart imaged during diastole and systole. The HA maps during diastole and systole are similar but the myocardial sheetlets, defined by the plane of the secondary and tertiary eigenvectors, undergo a radial re-orientation in systole. Reproduced with permission.112
Cardiomyocyte contraction results in radial thickening of the myocardium, which is the primary eigenvector (major direction) of myocardial strain.106 However, there are no radially oriented cardiomyocytes in the heart.110, 113 Rather, the radial yield of cardiomyocyte contraction is mediated via cardiomyocyte sheetlets, which consist of groups of 5–10 cardiomyocytes.118 The reorientation, extension and shearing of these sheets allows the heart to undergo radial strain during systole and diastole.118 The orientation of these sheets is given by the secondary eigenvector of the diffusion tensor and changes dynamically through the cardiac cycle.112, 119, 120 During diastole the sheets are oriented tangential to the surface of the heart and undergo a radial reorientation in systole (Figure 4).112, 119, 120 This can be quantified by following the orientation of the secondary eigenvector across the cardiac cycle. Because two mirror-image populations of sheets exist in the subendocardium and subepicardium the absolute value of the sheet angle (E2A) is used. In humans with dilated cardiomyopathy and HCM abnormalities in E2A have been detected in systole and diastole, respectively.120 DTI of the heart in small animals provides a wealth of information but remains a highly specialized technique and requires dedicated sequences and gradient systems to be performed in vivo.29
Imaging of Cell Death
In addition to LGE, manganese (Mn) based imaging has also been used to image cardiomyocyte viability.121, 122 Mn is paramagnetic and its uptake can be detected with MRI much like Gd. In the case of Mn, however, areas of viable myocardium take up the probe. Studies in rodents and large animals have shown that the addition of Mn imaging to LGE may allow areas of viable myocardium to be distinguished more accurately than LGE alone.121, 122 Molecular imaging approaches to image several forms of cell death including apoptosis, necrosis and autophagy have been developed and tested in the murine heart. Much of the initial work in apoptosis focused on the use of annexin-V to detect apoptosis with SPECT.123 Small animal imaging of apoptosis has now been performed by attaching radiotracers, iron-oxide nanoparticles and Gd-labeled liposomes to annexin-V (Figure 5).124–127
Figure 5: Molecular imaging of cell death.

(A) Detection of apoptosis in the murine heart in vivo with an annexin-decorated iron-oxide nanoparticle. Within a few hours of ischemia-reperfusion any unbound nanoparticles have washed out while phosphatidylserine-bound nanoparticles are retained and produce signal hypointensity on MRI. Reproduced with permission.127 (B) Detection of necrosis in a murine infarct with Gd-TO, a DNA-binding gadolinium chelate. The probe cannot cross an intact cell membrane but in the presence of necrotic cardiomyocytes can access and bind to nuclear/cell free DNA. (C) The TO moiety on the probe both binds to nucleic acids and provides a fluorescent readout, allowing the co-localization of Gd-TO with DAPI to be assessed by microscopy. (B-C) Reproduced with permission.24 (D) TO can be conjugated to a 40kD dextran to create an optical probe capable of detecting necrosis ex vivo in the murine heart. (E) Simultaneous injection of Dex-TO and a near-infrared annexin (AV-750) in mice with ischemia-reperfusion shows that areas of myocardium with low-moderate levels of annexin uptake do not lose cell integrity and may not fully execute the cell death cascade. (D-E) Reproduced with permission.128 (F) The injection of rapamycin (RAP) either before or after the onset of reperfusion significantly reduces the portion of the area-at-risk that becomes positive for annexin-uptake. Reproduced with permission.131 (G) Detection of transplant rejection (arrow) with 99Tc-duramycin. Reproduced with permission.129
The pathophysiological significance of annexin-V uptake has been closely studied in small animal models and is nuanced. The vast majority of the area-at-risk (AAR) following ischemia-reperfusion injury shows some uptake of annexin-V (Figure 5).128 However, only portions of the AAR with high levels of annexin uptake seem to fully execute the cell death cascade.128 Low levels of annexin-V uptake may, therefore, be a marker of ischemic injury rather than cell death. In addition, rupture of the cardiomyocyte membrane due to necrosis will also result in the uptake of annexin-V by phosphatidylserine on the inner side of the cell membrane. A dual contrast approach in which annexin is used with a marker of cell membrane rupture, such as LGE or antimyosin imaging, has been implemented to address this.126, 127 Several other ligands (C2-synaptotagmin, duramycin) have been developed to bind to phosphatidylserine on the surface of apoptotic cells and have been imaged in small animal models (Figure 5).129, 130 Many of the concerns and limitations of annexin-based imaging apply equally to these ligands.
Vital fluorochromes, most notably thiazole orange (TO), have also been modified to support in vivo imaging of necrosis in small animals (Figure 5).24 Gd-TO cannot cross an intact cell membrane and in the absence of necrosis is rapidly washed out of healthy myocardium. Access to nuclear DNA in ruptured cardiomycytes, or to cell-free DNA secreted into the interstitial space, results in Gd-TO binding and retention of the probe in the myocardium.24 The imaging of autophagy in vivo is challenging although a cathepsin-activatable near infrared fluorochrome has been used to image autophagy noninvasively in the heart.131 The near infrared nature of the probe allows fluorescence tomography of the heart to be performed non-invasively in mice.131, 132 However, the sensitivity of this fluorochrome for autophagy is moderate. More recently, a second-generation autophagy detecting nanoparticle, with extremely high sensitivity and specificity, has been generated to address this limitation.
Myocardial Inflammation
The imaging of myocardial inflammation is frequently performed by detecting the uptake of [18F]-fluorodeoxyglocose (FDG) by inflammatory cells in the heart. However, this approach relies on the complete suppression of glucose uptake by cardiomyocytes, which is challenging. Consequently, numerous probes have been developed to image myocardial inflammation with greater specificity. Iron-oxide nanoparticles are avidly taken up by macrophages and other cells in the reticulo-endothelial system (Figure 6).132 In the preclinical context a fluorochrome can be easily conjugated to the surface of the iron-oxide nanoparticle, which allows its uptake to be imaged at the whole organ level by MRI and fluorescence tomography, and at the cellular level with flow cytometry and microscopy.132 Several other MRI-based approaches have been used to image the infiltration of inflammatory cells into the heart including the use of Gd and [19F]-containing liposomes,108, 133–135 and nanoparticles labeled with both [19F] and [89Zr] (zirconium) (Figure 6).108, 133–136
Figure 6: Imaging of myocardial inflammation and remodeling.

Macrophages infiltrating healing myocardial infarcts (arrows) in the murine heart can be imaged with a variety of nanoparticles, which they avidly take up. (A) Iron-oxide nanoparticles, which produce signal hypointensity on the MR images. Reproduced with permission.132 (B) Gadolinium-loaded liposomes, which increase the longitudinal relaxation rate (R1) in the vicinity of their uptake. Reproduced with permission.108 (C) The signal from (19F)-loaded liposomes can be distinguished from the proton MRI signal allowing simultaneous (19F) and (1H) MRI to be performed. Reproduced with permission.133, 134 (D) [64Cu]-labeled macrin can be detected with micro-PET. (w = surgical wound in chest wall). Reproduced with permission.141 (E) Pre-labeling of myeloid cells with [89Zr] and (19F) nanoparticles. After myocardial infarction (MI) there is a rapid egress of myeloid cells from the liver (li) and spleen (sp). [89Zr] autoradiography shows a corresponding infiltration of these cells into the myocardium after MI or ischemia-reperfusion (IR). Reproduced with permission.136 (F-G) Imaging of macrophage infarct infiltration with a myeloperoxidase (MPO) activatable gadolinium-chelate. Activation of the probe in wildtype mice produces signal hyperintensity in the infarct, which is absent in the homozygous knockout mouse. Reproduced with permission.138 (H) MMP activation in mice after MI. The SPECT perfusion images (green) delineate the infarct zone, which shows significant uptake of the MMP-targeted probe (red). Reproduced with permission.147 (I) In vivo microPET of CXCR4 distribution in a murine infarct with corresponding ex vivo autoradiography below. Reproduced with permission.148 (J) Gadolinium chelate targeted to elastin is taken up in the anterior wall and apex of a healing murine infarct. Reproduced with permission.149 (K-L) Gadolinium chelate targeted to type 1 collagen accurately detects areas of fibrosis in a healed murine infarct. Reproduced with permission.25
The degradative enzymes released by inflammatory cells can also be directly imaged. Cathepsin-activatable fluorochromes, for instance, are robustly activated by neutrophils and macrophages.137 Another approach involves the detection of myeloperoxidase (MPO) secreted by inflammatory cells with a MPO-activatable Gd chelate (Figure 6).138 Several small molecule PET probes have been developed to image myocardial inflammation but have not proven superior to [18F]-FDG.139 However, a novel PET tracer, [18F-DHMT], was able to image the generation of reactive oxygen species in rodents treated with doxorubicin.140 In addition, the radiolabeled nanoparticle, [64Cu]-Macrin, appears to be highly promising. Preclinical use of the probe in mice showed that it could accurately detect myocardial inflammation (Figure 6).141 Ischemic injury also results in endothelial inflammation, which can be detected in mice with PET, MRI and targeted microbubbles.3, 78
Myocardial Healing, Angiogenesis and Fibrosis
Molecular imaging of angiogenesis using ligands to the αvβ3-integrin has been successfully performed in small animal models.142, 143 The uptake of these agents, however, is context specific. The RGD peptide, for instance, has also been reported to bind to αvβ3-integrin on the surface of myofibroblasts.144 The detection of fibroblast/myofibroblast activation can also be performed using radiolabeled fibroblast activation protein inhibitors (FAPI).145 Optical and SPECT-based approaches have been developed to image matrix metalloproteinases (MMPs) in the remodeling heart. The rate constant of MMP activation in the heart, however, may not be sufficient to support robust detection of the optical construct in vivo.146 In contrast, the SPECT based approach has been used with significant success (Figure 6).147 The probe consists of a chemical inhibitor to MMPs (2 and 9) conjugated to either [99Tc] or [111In] and has been used in models of myocardial infarction and anthracycline cardiotoxicity.140, 147 The role of CXCR4 in the remodeling heart is multifaceted and its imaging with PET in mice has provided valuable insights (Figure 6).148
Molecular imaging agents have been developed to directly image collagen and elastin during ventricular remodeling (Figure 6).25, 149 A peptide targeting type 1 collagen has been conjugated to Gd and used to specifically image fibrosis in infarcted mice.25 The highly reactive allysine groups on collagen, which cross link it and make it more resistant to breakdown, can be detected with an aldehyde (allysine) binding Gd chelate.150 This probe may be a marker of active fibrogenesis and provide a complementary readout to other collagen-targeted probes.
Myocardial Regeneration
Human studies involving the injection of bone-marrow derived and mesenchymal stem cells have been disappointing. Interestingly studies in small animal models using advanced imaging techniques did not show encouraging results. In one such study, serial in vivo diffusion tensor MRI tractography (DTI-tractography) was used to evaluate the impact of bone marrow derived mononuclear cells injected directly into mice three weeks after ischemia-reperfusion injury (Figure 7).29 Serial imaging before and after cell injection showed no restoration of myocardial anisotropy and microstructure in >90% of injected mice. Moreover, in some of the mice cell injection actually resulted in a loss of coherent microstructure (Figure 7).29
Figure 7. Diffusion tensor MRI and reporter imaging of cell therapy.

(A) Islets of surviving cardiomyocytes in a rat infarct create a network of residual tracts, which must be differentiated from regenerated myocardial tracts. Reproduced with permission.113 (B-C) DTI-tractography of the murine heart in vivo. Tracts intersecting a region-of-interest (red rectangle, inset) in the lateral wall are shown from a lateral and basal perspective, respectively. (D-I) Serial in vivo DTI tractography of mice with ischemic injury injected with bone marrow mononuclear cells. (D) In the majority of mice no response to cell injection was seen and a severe loss of tracts in the apical half of the ventricle persisted after injection. (E-G) However, in some mice cell injection was associated with the loss of tracts after injection. (E) Before injection coherent tracts (arrows) are seen in the anterior and inferolateral walls. (F) Imaging of the same mouse 7 days after injection reveals the loss of these tracts. (G) The transmural slope of helix angle in the inferolateral wall is normal before injection (blue curve) and severely perturbed (black curve) post-injection. (H) Cell injection produced an increase in mean diffusivity and a decrease in fractional anisotropy, which are signatures of injury or inflammation, rather than regeneration. (I) In >90% of the mice studied cell injection produced a negative (neutral or deleterious) response. (B-I) Reproduced with permission.29 (J) Serial bioluminescence imaging shows that bone marrow mononuclear cells injected into infarcted mice do not survive, with a vast reduction in signal from day 4 to day 21. Reproduced with permission.153 (K) Conversely embryonic stem cells, shown at weeks 3 and 4 by bioluminescence and PET reporter imaging, survive and grow (arrows) but distribute beyond the heart and form teratomas. Reproduced with permission.154
The labeling of stem cells with iron-oxide probes can be extremely useful in guiding their injection.151, 152 However, the persistence of the iron-oxide label in the myocardium does not equate with cell survival.152 Reporter techniques, in which the imaging readout relies on the active synthesis of a protein/enzyme by the cell, address this limitation (Figure 7).153, 154 Bioluminescent and PET reporter probes have been used to show that bone marrow mononuclear cells injected into infarcted myocardium survive extremely poorly,153 and conversely that embryonic cells survive extremely well but form teratomas (Figure 7).154
The Right Ventricle and Pulmonary Circulation
Right ventricular anatomy and function is difficult to assess using murine echocardiography due to its complex geometrical shape and small size. Although investigators have attempted to measure RV size, EF, and wall thickness using echocardiographic parasternal views,155–157 these parameters are obtained with more precision using MRI. The tricuspid annular velocity (S’) and tricuspid annular plane systolic excursion (TAPSE) can also be measured from the apical 4 chamber view and were decreased in a model of hypoxia and vascular endothelial growth factor inhibitor, and in heart failure.155, 156 Due to the paucity of tricuspid regurgitation in rodents, the measurement of pulmonary artery or right ventricular pressure using the maximum velocity of the tricuspid regurgitation jet is not feasible. Pulsed wave Doppler, however, has shown inverse correlations between systolic pulmonary artery pressure, measured by a pressure catheter in mice, and both the pulmonary acceleration time (PAT) and PAT/ejection time.158 These echocardiographic indices have been used widely in models of pulmonary hypertension.155, 159, 160 While most MRI based approaches in mice have focused on the imaging of the LV, many of the advanced MRI techniques, described above, can also be performed in the murine RV. However, others are limited by the thin nature of the RV wall. At the present time MRI of the murine RV is limited in most centers to measurements of RV volumes and function, including RV strain by feature tracking, with cine MRI.161
Future Prospects and Conclusion
Advances in biomedical imaging continue to occur at a rapid pace. The development of integrated multimodality platforms, such a PET-MR and optoacoustic systems, is opening up new possibilities in both the clinical and research domains. Many of the recently developed molecular imaging agents also incorporate a therapeutic capability (theranostic agents), which is ideally suited to testing in small animal models. Machine learning is playing an increasing role in biomedical imaging and will likely impact all areas of small animal imaging from acquisition to reconstruction and analysis. New approaches will also need to be developed to better integrate biomedical imaging data with genomic/metabolomic/proteomic data. Major opportunities for innovation will continue to exist at the interface of molecular biology and advanced biomedical imaging and will require a high degree of collaboration between the two communities to be fully realized.
Supplementary Material
Funding:
Funded in part by the following grants from the National Institutes of Health: R01HL159010, R01HL141563, R01HL109448 (DES) and R01HL131613, R01HL130539 (MSC)
Non-Standard Abbreviations and Acronyms
- T1
Longitudinal Magnetic Relaxation Time
- T1-Rho
Magnetic Relaxation Time with Spin-Locking
- UTE
Ultrashort Echo Time
- CEST
Chemical Exchange Saturation Transfer
- BOLD
Blood Oxygen Level Dependent
- ASL
Arterial Spin Labeling
- DENSE
Displacement Encoded Stimulated Echo
- DTI
Diffusion Tensor Imaging
- MD
Mean Diffusivity
- FA
Fractional Anisotropy
- HA
Helix Angle
- PA
Propagation Angle
- E2A
Absolute Value of Sheet Angle
- TO
Thiazole Orange
- MPO
Myeloperoxidase
- MMP
Matrix Metalloproteinase
- FAPI
Fibroblast Activation Protein Inhibitor
Footnotes
Disclosures: The Martinos Center for Biomedical Imaging has a research agreement with Siemens Medical, Erlangen Germany. Dr. Sosnovik has a research agreement with Collagen Medical, Boston MA.
References:
- 1.Dweck MR, Aikawa E, Newby DE, Tarkin JM, Rudd JH, Narula J and Fayad ZA. Noninvasive Molecular Imaging of Disease Activity in Atherosclerosis. Circ Res. 2016;119:330–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Osborn EA, Kessinger CW, Tawakol A and Jaffer FA. Metabolic and Molecular Imaging of Atherosclerosis and Venous Thromboembolism. J Nucl Med. 2017;58:871–877. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Nahrendorf M, Sosnovik DE, French BA, Swirski FK, Bengel F, Sadeghi MM, Lindner JR, Wu JC, Kraitchman DL, Fayad ZA and Sinusas AJ. Multimodality cardiovascular molecular imaging, Part II. Circ Cardiovasc Imaging. 2009;2:56–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Lindner JR and Link J. Molecular Imaging in Drug Discovery and Development. Circ Cardiovasc Imaging. 2018;11:e005355. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Manning WJ, Wei JY, Katz SE, Litwin SE and Douglas PS. In vivo assessment of LV mass in mice using high-frequency cardiac ultrasound: necropsy validation. Am J Physiol. 1994;266:H1672–5. [DOI] [PubMed] [Google Scholar]
- 6.Shung KK. High Frequency Ultrasonic Imaging. J Med Ultrasound. 2009;17:25–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Scherrer-Crosbie M and Thibault HB. Echocardiography in translational research: of mice and men. J Am Soc Echocardiogr. 2008;21:1083–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Chaves AA, Weinstein DM and Bauer JA. Non-invasive echocardiographic studies in mice: influence of anesthetic regimen. Life Sci. 2001;69:213–22. [DOI] [PubMed] [Google Scholar]
- 9.Pachon RE, Scharf BA, Vatner DE and Vatner SF. Best anesthetics for assessing left ventricular systolic function by echocardiography in mice. Am J Physiol Heart Circ Physiol. 2015;308:H1525–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Hoit BD, Khan ZU, Pawloski-Dahm CM and Walsh RA. In vivo determination of left ventricular wall stress-shortening relationship in normal mice. Am J Physiol. 1997;272:H1047–52. [DOI] [PubMed] [Google Scholar]
- 11.Williams RV, Lorenz JN, Witt SA, Hellard DT, Khoury PR and Kimball TR. End-systolic stress-velocity and pressure-dimension relationships by transthoracic echocardiography in mice. Am J Physiol. 1998;274:H1828–35. [DOI] [PubMed] [Google Scholar]
- 12.Bhan A, Sirker A, Zhang J, Protti A, Catibog N, Driver W, Botnar R, Monaghan MJ and Shah AM. High-frequency speckle tracking echocardiography in the assessment of left ventricular function and remodeling after murine myocardial infarction. Am J Physiol Heart Circ Physiol. 2014;306:H1371–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Heinen A, Raupach A, Behmenburg F, Holscher N, Flogel U, Kelm M, Kaisers W, Nederlof R, Huhn R and Godecke A. Echocardiographic Analysis of Cardiac Function after Infarction in Mice: Validation of Single-Plane Long-Axis View Measurements and the Bi-Plane Simpson Method. Ultrasound Med Biol. 2018;44:1544–1555. [DOI] [PubMed] [Google Scholar]
- 14.Russo I, Micotti E, Fumagalli F, Magnoli M, Ristagno G, Latini R and Staszewsky L. A novel echocardiographic method closely agrees with cardiac magnetic resonance in the assessment of left ventricular function in infarcted mice. Sci Rep. 2019;9:3580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Rutledge C, Cater G, McMahon B, Guo L, Nouraie SM, Wu Y, Villanueva F and Kaufman BA. Commercial 4-dimensional echocardiography for murine heart volumetric evaluation after myocardial infarction. Cardiovasc Ultrasound. 2020;18:9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Dawson D, Lygate CA, Saunders J, Schneider JE, Ye X, Hulbert K, Noble JA and Neubauer S. Quantitative 3-dimensional echocardiography for accurate and rapid cardiac phenotype characterization in mice. Circulation. 2004;110:1632–7. [DOI] [PubMed] [Google Scholar]
- 17.Scherrer-Crosbie M, Steudel W, Hunziker PR, Foster GP, Garrido L, Liel-Cohen N, Zapol WM and Picard MH. Determination of right ventricular structure and function in normoxic and hypoxic mice: a transesophageal echocardiographic study. Circulation. 1998;98:1015–21. [DOI] [PubMed] [Google Scholar]
- 18.Scherrer-Crosbie M, Steudel W, Hunziker PR, Liel-Cohen N, Ullrich R, Zapol WM and Picard MH. Three-dimensional echocardiographic assessment of left ventricular wall motion abnormalities in mouse myocardial infarction. J Am Soc Echocardiogr. 1999;12:834–40. [DOI] [PubMed] [Google Scholar]
- 19.Rodrigues AC, Hataishi R, Ichinose F, Bloch KD, Derumeaux G, Picard MH and Scherrer-Crosbie M. Relationship of systolic dysfunction to area at risk and infarction size after ischemia-reperfusion in mice. J Am Soc Echocardiogr. 2004;17:948–53. [DOI] [PubMed] [Google Scholar]
- 20.Wiesmann F, Frydrychowicz A, Rautenberg J, Illinger R, Rommel E, Haase A and Neubauer S. Analysis of right ventricular function in healthy mice and a murine model of heart failure by in vivo MRI. Am J Physiol Heart Circ Physiol. 2002;283:H1065–71. [DOI] [PubMed] [Google Scholar]
- 21.van Nierop BJ, van Assen HC, van Deel ED, Niesen LB, Duncker DJ, Strijkers GJ and Nicolay K. Phenotyping of left and right ventricular function in mouse models of compensated hypertrophy and heart failure with cardiac MRI. PLoS One. 2013;8:e55424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Yang Z, Berr SS, Gilson WD, Toufektsian MC and French BA. Simultaneous evaluation of infarct size and cardiac function in intact mice by contrast-enhanced cardiac magnetic resonance imaging reveals contractile dysfunction in noninfarcted regions early after myocardial infarction. Circulation. 2004;109:1161–7. [DOI] [PubMed] [Google Scholar]
- 23.Protti A, Sirker A, Shah AM and Botnar R. Late gadolinium enhancement of acute myocardial infarction in mice at 7T: cine-FLASH versus inversion recovery. J Magn Reson Imaging. 2010;32:878–86. [DOI] [PubMed] [Google Scholar]
- 24.Huang S, Chen HH, Yuan H, Dai G, Schuhle DT, Mekkaoui C, Ngoy S, Liao R, Caravan P, Josephson L and Sosnovik DE. Molecular MRI of acute necrosis with a novel DNA-binding gadolinium chelate: kinetics of cell death and clearance in infarcted myocardium. Circ Cardiovasc Imaging. 2011;4:729–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Helm PA, Caravan P, French BA, Jacques V, Shen L, Xu Y, Beyers RJ, Roy RJ, Kramer CM and Epstein FH. Postinfarction myocardial scarring in mice: molecular MR imaging with use of a collagen-targeting contrast agent. Radiology. 2008;247:788–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Friedrich MG, Abdel-Aty H, Taylor A, Schulz-Menger J, Messroghli D and Dietz R. The salvaged area at risk in reperfused acute myocardial infarction as visualized by cardiovascular magnetic resonance. J Am Coll Cardiol. 2008;51:1581–7. [DOI] [PubMed] [Google Scholar]
- 27.Friedrich MG, Kim HW and Kim RJ. T2-weighted imaging to assess post-infarct myocardium at risk. JACC Cardiovasc Imaging. 2011;4:1014–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Beyers RJ, Smith RS, Xu Y, Piras BA, Salerno M, Berr SS, Meyer CH, Kramer CM, French BA and Epstein FH. T(2) -weighted MRI of post-infarct myocardial edema in mice. Magn Reson Med. 2012;67:201–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Sosnovik DE, Mekkaoui C, Huang S, Chen HH, Dai G, Stoeck CT, Ngoy S, Guan J, Wang R, Kostis WJ, Jackowski MP, Wedeen VJ, Kozerke S and Liao R. Microstructural impact of ischemia and bone marrow-derived cell therapy revealed with diffusion tensor magnetic resonance imaging tractography of the heart in vivo. Circulation. 2014;129:1731–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Bulluck H, Hammond-Haley M, Fontana M, Knight DS, Sirker A, Herrey AS, Manisty C, Kellman P, Moon JC and Hausenloy DJ. Quantification of both the area-at-risk and acute myocardial infarct size in ST-segment elevation myocardial infarction using T1-mapping. J Cardiovasc Magn Reson. 2017;19:57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Cui SX and Epstein FH. MRI assessment of coronary microvascular endothelial nitric oxide synthase function using myocardial T1 mapping. Magn Reson Med. 2018;79:2246–2253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Vandsburger MH, Janiczek RL, Xu Y, French BA, Meyer CH, Kramer CM and Epstein FH. Improved arterial spin labeling after myocardial infarction in mice using cardiac and respiratory gated look-locker imaging with fuzzy C-means clustering. Magn Reson Med. 2010;63:648–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Nezafat M, Ramos IT, Henningsson M, Protti A, Basha T and Botnar RM. Improved segmented modified Look-Locker inversion recovery T1 mapping sequence in mice. PLoS One. 2017;12:e0187621. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Messroghli DR, Moon JC, Ferreira VM, Grosse-Wortmann L, He T, Kellman P, Mascherbauer J, Nezafat R, Salerno M, Schelbert EB, Taylor AJ, Thompson R, Ugander M, van Heeswijk RB and Friedrich MG. Clinical recommendations for cardiovascular magnetic resonance mapping of T1, T2, T2* and extracellular volume: A consensus statement by the Society for Cardiovascular Magnetic Resonance (SCMR) endorsed by the European Association for Cardiovascular Imaging (EACVI). J Cardiovasc Magn Reson. 2017;19:75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Coelho-Filho OR, Shah RV, Mitchell R, Neilan TG, Moreno H Jr., Simonson B, Kwong R, Rosenzweig A, Das S and Jerosch-Herold M Quantification of cardiomyocyte hypertrophy by cardiac magnetic resonance: implications for early cardiac remodeling. Circulation. 2013;128:1225–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Kwiecinski J, Lennen RJ, Gray GA, Borthwick G, Boswell L, Baker AH, Newby DE, Dweck MR and Jansen MA. Progression and regression of left ventricular hypertrophy and myocardial fibrosis in a mouse model of hypertension and concomitant cardiomyopathy. J Cardiovasc Magn Reson. 2020;22:57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Stuckey DJ, McSweeney SJ, Thin MZ, Habib J, Price AN, Fiedler LR, Gsell W, Prasad SK and Schneider MD. T(1) mapping detects pharmacological retardation of diffuse cardiac fibrosis in mouse pressure-overload hypertrophy. Circ Cardiovasc Imaging. 2014;7:240–9. [DOI] [PubMed] [Google Scholar]
- 38.Bedet A, Voiriot G, Ternacle J, Marcos E, Adnot S, Derumeaux G and Mekontso Dessap A. Heart Rate Control during Experimental Sepsis in Mice: Comparison of Ivabradine and beta-Blockers. Anesthesiology. 2020;132:321–329. [DOI] [PubMed] [Google Scholar]
- 39.Du XJ, Feng X, Gao XM, Tan TP, Kiriazis H and Dart AM. I(f) channel inhibitor ivabradine lowers heart rate in mice with enhanced sympathoadrenergic activities. Br J Pharmacol. 2004;142:107–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.O’Connor DM, Smith RS, Piras BA, Beyers RJ, Lin D, Hossack JA and French BA. Heart Rate Reduction With Ivabradine Protects Against Left Ventricular Remodeling by Attenuating Infarct Expansion and Preserving Remote-Zone Contractile Function and Synchrony in a Mouse Model of Reperfused Myocardial Infarction. J Am Heart Assoc. 2016;5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Nagueh SF, Smiseth OA, Appleton CP, Byrd BF 3rd, Dokainish H, Edvardsen T, Flachskampf FA, Gillebert TC, Klein AL, Lancellotti P, Marino P, Oh JK, Popescu BA and Waggoner AD. Recommendations for the Evaluation of Left Ventricular Diastolic Function by Echocardiography: An Update from the American Society of Echocardiography and the European Association of Cardiovascular Imaging. J Am Soc Echocardiogr. 2016;29:277–314. [DOI] [PubMed] [Google Scholar]
- 42.Granillo A, Pena CA, Pham T, Pandit LM and Taffet GE. Murine Echocardiography of Left Atrium, Aorta, and Pulmonary Artery. J Vis Exp. 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Medrano G, Hermosillo-Rodriguez J, Pham T, Granillo A, Hartley CJ, Reddy A, Osuna PM, Entman ML and Taffet GE. Left Atrial Volume and Pulmonary Artery Diameter Are Noninvasive Measures of Age-Related Diastolic Dysfunction in Mice. J Gerontol A Biol Sci Med Sci. 2016;71:1141–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Schnelle M, Catibog N, Zhang M, Nabeebaccus AA, Anderson G, Richards DA, Sawyer G, Zhang X, Toischer K, Hasenfuss G, Monaghan MJ and Shah AM. Echocardiographic evaluation of diastolic function in mouse models of heart disease. J Mol Cell Cardiol. 2018;114:20–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Richards DA, Aronovitz MJ, Calamaras TD, Tam K, Martin GL, Liu P, Bowditch HK, Zhang P, Huggins GS and Blanton RM. Distinct Phenotypes Induced by Three Degrees of Transverse Aortic Constriction in Mice. Sci Rep. 2019;9:5844. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Herold V, Herz S, Winter P, Gutjahr FT, Andelovic K, Bauer WR and Jakob PM. Assessment of local pulse wave velocity distribution in mice using k-t BLAST PC-CMR with semi-automatic area segmentation. J Cardiovasc Magn Reson. 2017;19:77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Braig M, Leupold J, Menza M, Russe M, Ko CW, Hennig J and von Elverfeldt D. Preclinical 4D-flow magnetic resonance phase contrast imaging of the murine aortic arch. PLoS One. 2017;12:e0187596. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Takaya Y, Nakamura K, Nakayama R, Ohtsuka H, Amioka N, Kondo M, Akazawa K, Ohno Y, Ichikawa K, Saito Y, Akagi S, Yoshida M, Miyoshi T and Ito H. Efficacy of shear wave elasticity for evaluating myocardial hypertrophy in hypertensive rats. Sci Rep. 2021;11:22812. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Khan S, Fakhouri F, Majeed W and Kolipaka A. Cardiovascular magnetic resonance elastography: A review. NMR Biomed. 2018;31:e3853. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Bayly PV and Garbow JR. Pre-clinical MR elastography: Principles, techniques, and applications. J Magn Reson. 2018;291:73–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Liu Y, Royston TJ, Klatt D and Lewandowski ED. Cardiac MR elastography of the mouse: Initial results. Magn Reson Med. 2016;76:1879–1886. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.van Nierop BJ, Bax NA, Nelissen JL, Arslan F, Motaal AG, de Graaf L, Zwanenburg JJ, Luijten PR, Nicolay K and Strijkers GJ. Assessment of Myocardial Fibrosis in Mice Using a T2*-Weighted 3D Radial Magnetic Resonance Imaging Sequence. PLoS One. 2015;10:e0129899. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Han Y, Liimatainen T, Gorman RC and Witschey WR. Assessing Myocardial Disease Using T1rho MRI. Curr Cardiovasc Imaging Rep. 2014;7:9248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Bustin A, Toupin S, Sridi S, Yerly J, Bernus O, Labrousse L, Quesson B, Rogier J, Haissaguerre M, van Heeswijk R, Jais P, Cochet H and Stuber M. Endogenous assessment of myocardial injury with single-shot model-based non-rigid motion-corrected T1 rho mapping. J Cardiovasc Magn Reson. 2021;23:119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Stoffers RH, Madden M, Shahid M, Contijoch F, Solomon J, Pilla JJ, Gorman JH 3rd, Gorman RC and Witschey WRT. Assessment of myocardial injury after reperfused infarction by T1rho cardiovascular magnetic resonance. J Cardiovasc Magn Reson. 2017;19:17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Maslov MY, Chacko VP, Stuber M, Moens AL, Kass DA, Champion HC and Weiss RG. Altered high-energy phosphate metabolism predicts contractile dysfunction and subsequent ventricular remodeling in pressure-overload hypertrophy mice. Am J Physiol Heart Circ Physiol. 2007;292:H387–91. [DOI] [PubMed] [Google Scholar]
- 57.ten Hove M, Lygate CA, Fischer A, Schneider JE, Sang AE, Hulbert K, Sebag-Montefiore L, Watkins H, Clarke K, Isbrandt D, Wallis J and Neubauer S. Reduced inotropic reserve and increased susceptibility to cardiac ischemia/reperfusion injury in phosphocreatine-deficient guanidinoacetate-N-methyltransferase-knockout mice. Circulation. 2005;111:2477–85. [DOI] [PubMed] [Google Scholar]
- 58.Schroeder MA, Clarke K, Neubauer S and Tyler DJ. Hyperpolarized magnetic resonance: a novel technique for the in vivo assessment of cardiovascular disease. Circulation. 2011;124:1580–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Lau AZ, Miller JJ, Robson MD and Tyler DJ. Simultaneous assessment of cardiac metabolism and perfusion using copolarized [1-(13) C]pyruvate and (13) C-urea. Magn Reson Med. 2017;77:151–158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Lewis AJM, Miller JJ, Lau AZ, Curtis MK, Rider OJ, Choudhury RP, Neubauer S, Cunningham CH, Carr CA and Tyler DJ. Noninvasive Immunometabolic Cardiac Inflammation Imaging Using Hyperpolarized Magnetic Resonance. Circ Res. 2018;122:1084–1093. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Miller JJ, Lau AZ, Nielsen PM, McMullen-Klein G, Lewis AJ, Jespersen NR, Ball V, Gallagher FA, Carr CA, Laustsen C, Botker HE, Tyler DJ and Schroeder MA. Hyperpolarized [1,4-(13)C2]Fumarate Enables Magnetic Resonance-Based Imaging of Myocardial Necrosis. JACC Cardiovasc Imaging. 2018;11:1594–1606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Cunningham CH, Lau JY, Chen AP, Geraghty BJ, Perks WJ, Roifman I, Wright GA and Connelly KA. Hyperpolarized 13C Metabolic MRI of the Human Heart: Initial Experience. Circ Res. 2016;119:1177–1182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Luptak I, Sverdlov AL, Panagia M, Qin F, Pimentel DR, Croteau D, Siwik DA, Ingwall JS, Bachschmid MM, Balschi JA and Colucci WS. Decreased ATP production and myocardial contractile reserve in metabolic heart disease. J Mol Cell Cardiol. 2018;116:106–114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Haris M, Singh A, Cai K, Kogan F, McGarvey J, Debrosse C, Zsido GA, Witschey WR, Koomalsingh K, Pilla JJ, Chirinos JA, Ferrari VA, Gorman JH, Hariharan H, Gorman RC and Reddy R. A technique for in vivo mapping of myocardial creatine kinase metabolism. Nat Med. 2014;20:209–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Pumphrey A, Yang Z, Ye S, Powell DK, Thalman S, Watt DS, Abdel-Latif A, Unrine J, Thompson K, Fornwalt B, Ferrauto G and Vandsburger M. Advanced cardiac chemical exchange saturation transfer (cardioCEST) MRI for in vivo cell tracking and metabolic imaging. NMR Biomed. 2016;29:74–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Croteau E, Renaud JM, Archer C, Klein R, DaSilva JN, Ruddy TD, Beanlands RS and deKemp RA. beta2-adrenergic stress evaluation of coronary endothelial-dependent vasodilator function in mice using (11)C-acetate micro-PET imaging of myocardial blood flow and oxidative metabolism. EJNMMI Res. 2014;4:68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Panagia M, Chen HH, Croteau D, Iris Chen YC, Ran C, Luptak I, Josephson L, Colucci WS and Sosnovik DE. Multiplexed Optical Imaging of Energy Substrates Reveals That Left Ventricular Hypertrophy Is Associated With Brown Adipose Tissue Activation. Circ Cardiovasc Imaging. 2018;11:e007007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Sawall S, Beckendorf J, Amato C, Maier J, Backs J, Vande Velde G, Kachelriess M and Kuntz J. Coronary micro-computed tomography angiography in mice. Sci Rep. 2020;10:16866. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Pong T, Scherrer-Crosbie M, Atochin DN, Bloch KD and Huang PL. Phosphomimetic modulation of eNOS improves myocardial reperfusion and mimics cardiac postconditioning in mice. PLoS One. 2014;9:e85946. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Scherrer-Crosbie M, Rodrigues AC, Hataishi R and Picard MH. Infarct size assessment in mice. Echocardiography. 2007;24:90–6. [DOI] [PubMed] [Google Scholar]
- 71.Scherrer-Crosbie M, Steudel W, Ullrich R, Hunziker PR, Liel-Cohen N, Newell J, Zaroff J, Zapol WM and Picard MH. Echocardiographic determination of risk area size in a murine model of myocardial ischemia. Am J Physiol. 1999;277:H986–92. [DOI] [PubMed] [Google Scholar]
- 72.Hartley CJ, Reddy AK, Michael LH, Entman ML, Chintalagattu V, Khakoo AY and Taffet GE. Coronary flow reserve in mice: effects of age, coronary disease, and vascular loading. Annu Int Conf IEEE Eng Med Biol Soc. 2010;2010:3780–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Wikstrom J, Gronros J and Gan LM. Adenosine induces dilation of epicardial coronary arteries in mice: relationship between coronary flow velocity reserve and coronary flow reserve in vivo using transthoracic echocardiography. Ultrasound Med Biol. 2008;34:1053–62. [DOI] [PubMed] [Google Scholar]
- 74.You J, Wu J, Ge J and Zou Y. Comparison between adenosine and isoflurane for assessing the coronary flow reserve in mouse models of left ventricular pressure and volume overload. Am J Physiol Heart Circ Physiol. 2012;303:H1199–207. [DOI] [PubMed] [Google Scholar]
- 75.Raher MJ, Thibault H, Poh KK, Liu R, Halpern EF, Derumeaux G, Ichinose F, Zapol WM, Bloch KD, Picard MH and Scherrer-Crosbie M. In vivo characterization of murine myocardial perfusion with myocardial contrast echocardiography: validation and application in nitric oxide synthase 3 deficient mice. Circulation. 2007;116:1250–7. [DOI] [PubMed] [Google Scholar]
- 76.Raher MJ, Thibault HB, Buys ES, Kuruppu D, Shimizu N, Brownell AL, Blake SL, Rieusset J, Kaneki M, Derumeaux G, Picard MH, Bloch KD and Scherrer-Crosbie M. A short duration of high-fat diet induces insulin resistance and predisposes to adverse left ventricular remodeling after pressure overload. Am J Physiol Heart Circ Physiol. 2008;295:H2495–502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Naresh NK, Butcher JT, Lye RJ, Chen X, Isakson BE, Gan LM, Kramer CM, Annex BH and Epstein FH. Cardiovascular magnetic resonance detects the progression of impaired myocardial perfusion reserve and increased left-ventricular mass in mice fed a high-fat diet. J Cardiovasc Magn Reson. 2016;18:53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Ozawa K, Packwood W, Varlamov O, Qi Y, Xie A, Wu MD, Ruggeri Z, Lopez JA and Lindner JR. Molecular Imaging of VWF (von Willebrand Factor) and Platelet Adhesion in Postischemic Impaired Microvascular Reflow. Circ Cardiovasc Imaging. 2018;11:e007913. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Gehrmann J, Hammer PE, Maguire CT, Wakimoto H, Triedman JK and Berul CI. Phenotypic screening for heart rate variability in the mouse. Am J Physiol Heart Circ Physiol. 2000;279:H733–40. [DOI] [PubMed] [Google Scholar]
- 80.Janssen B, Debets J, Leenders P and Smits J. Chronic measurement of cardiac output in conscious mice. Am J Physiol Regul Integr Comp Physiol. 2002;282:R928–35. [DOI] [PubMed] [Google Scholar]
- 81.Li Z, Li Y, Zhang L, Zhang X, Sullivan R, Ai X, Szeto C, Cai A, Liu L, Xiao W, Li Q, Ge S and Chen X. Reduced Myocardial Reserve in Young X-Linked Muscular Dystrophy Mice Diagnosed by Two-Dimensional Strain Analysis Combined with Stress Echocardiography. J Am Soc Echocardiogr. 2017;30:815–827 e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Pan G, Munukutla S, Kar A, Gardinier J, Thandavarayan RA and Palaniyandi SS. Type-2 diabetic aldehyde dehydrogenase 2 mutant mice (ALDH 2*2) exhibiting heart failure with preserved ejection fraction phenotype can be determined by exercise stress echocardiography. PLoS One. 2018;13:e0195796. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Roh JD, Houstis N, Yu A, Chang B, Yeri A, Li H, Hobson R, Lerchenmuller C, Vujic A, Chaudhari V, Damilano F, Platt C, Zlotoff D, Lee RT, Shah R, Jerosch-Herold M and Rosenzweig A. Exercise training reverses cardiac aging phenotypes associated with heart failure with preserved ejection fraction in male mice. Aging Cell. 2020;19:e13159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Ghotbi AA, Clemmensen A, Kyhl K, Follin B, Hasbak P, Engstrom T, Ripa RS and Kjaer A. Rubidium-82 PET imaging is feasible in a rat myocardial infarction model. J Nucl Cardiol. 2019;26:798–809. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Bentsen S, Bang LE, Hasbak P, Kjaer A and Ripa RS. Amiodarone attenuates cardiac Rubidium-82 in consecutive PET/CT scans in a rodent model. J Nucl Cardiol. 2021. [DOI] [PubMed] [Google Scholar]
- 86.Hess A, Nekolla SG, Meier M, Bengel FM and Thackeray JT. Accuracy of cardiac functional parameters measured from gated radionuclide myocardial perfusion imaging in mice. J Nucl Cardiol. 2020;27:1317–1327. [DOI] [PubMed] [Google Scholar]
- 87.Wacker CM, Hartlep AW, Pfleger S, Schad LR, Ertl G and Bauer WR. Susceptibility-sensitive magnetic resonance imaging detects human myocardium supplied by a stenotic coronary artery without a contrast agent. J Am Coll Cardiol. 2003;41:834–40. [DOI] [PubMed] [Google Scholar]
- 88.Yang HJ, Oksuz I, Dey D, Sykes J, Klein M, Butler J, Kovacs MS, Sobczyk O, Cokic I, Slomka PJ, Bi X, Li D, Tighiouart M, Tsaftaris SA, Prato FS, Fisher JA and Dharmakumar R. Accurate needle-free assessment of myocardial oxygenation for ischemic heart disease in canines using magnetic resonance imaging. Sci Transl Med. 2019;11. [DOI] [PubMed] [Google Scholar]
- 89.Fischer K, Guensch DP and Friedrich MG. Response of myocardial oxygenation to breathing manoeuvres and adenosine infusion. Eur Heart J Cardiovasc Imaging. 2015;16:395–401. [DOI] [PubMed] [Google Scholar]
- 90.Kober F, Jao T, Troalen T and Nayak KS. Myocardial arterial spin labeling. J Cardiovasc Magn Reson. 2016;18:22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Abdesselam I, Pepino P, Troalen T, Macia M, Ancel P, Masi B, Fourny N, Gaborit B, Giannesini B, Kober F, Dutour A and Bernard M. Time course of cardiometabolic alterations in a high fat high sucrose diet mice model and improvement after GLP-1 analog treatment using multimodal cardiovascular magnetic resonance. J Cardiovasc Magn Reson. 2015;17:95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Naresh NK, Chen X, Roy RJ, Antkowiak PF, Annex BH and Epstein FH. Accelerated dual-contrast first-pass perfusion MRI of the mouse heart: development and application to diet-induced obese mice. Magn Reson Med. 2015;73:1237–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Sutherland GR, Di Salvo G, Claus P, D’Hooge J and Bijnens B. Strain and strain rate imaging: a new clinical approach to quantifying regional myocardial function. J Am Soc Echocardiogr. 2004;17:788–802. [DOI] [PubMed] [Google Scholar]
- 94.Sebag IA, Handschumacher MD, Ichinose F, Morgan JG, Hataishi R, Rodrigues AC, Guerrero JL, Steudel W, Raher MJ, Halpern EF, Derumeaux G, Bloch KD, Picard MH and Scherrer-Crosbie M. Quantitative assessment of regional myocardial function in mice by tissue Doppler imaging: comparison with hemodynamics and sonomicrometry. Circulation. 2005;111:2611–6. [DOI] [PubMed] [Google Scholar]
- 95.Neilan TG, Jassal DS, Perez-Sanz TM, Raher MJ, Pradhan AD, Buys ES, Ichinose F, Bayne DB, Halpern EF, Weyman AE, Derumeaux G, Bloch KD, Picard MH and Scherrer-Crosbie M. Tissue Doppler imaging predicts left ventricular dysfunction and mortality in a murine model of cardiac injury. Eur Heart J. 2006;27:1868–75. [DOI] [PubMed] [Google Scholar]
- 96.Gorcsan J 3rd, and Tanaka H Echocardiographic assessment of myocardial strain. J Am Coll Cardiol. 2011;58:1401–13. [DOI] [PubMed] [Google Scholar]
- 97.Bauer M, Cheng S, Jain M, Ngoy S, Theodoropoulos C, Trujillo A, Lin FC and Liao R. Echocardiographic speckle-tracking based strain imaging for rapid cardiovascular phenotyping in mice. Circ Res. 2011;108:908–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Chaudhury A, Wanek A, Ponnalagu D, Singh H and Kohut A. Use of Speckle Tracking Echocardiography to Detect Induced Regional Strain Changes in the Murine Myocardium by Acoustic Radiation Force. J Cardiovasc Imaging. 2021;29:147–157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Deddens JC, Feyen DA, Zwetsloot PP, Brans MA, Siddiqi S, van Laake LW, Doevendans PA and Sluijter JP. Targeting chronic cardiac remodeling with cardiac progenitor cells in a murine model of ischemia/reperfusion injury. PLoS One. 2017;12:e0173657. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Hoffman M, Kyriazis ID, Lucchese AM, de Lucia C, Piedepalumbo M, Bauer M, Schulze PC, Bonios MJ, Koch WJ and Drosatos K. Myocardial Strain and Cardiac Output are Preferable Measurements for Cardiac Dysfunction and Can Predict Mortality in Septic Mice. J Am Heart Assoc. 2019;8:e012260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Ternacle J, Wan F, Sawaki D, Surenaud M, Pini M, Mercedes R, Ernande L, Audureau E, Dubois-Rande JL, Adnot S, Hue S, Czibik G and Derumeaux G. Short-term high-fat diet compromises myocardial function: a radial strain rate imaging study. Eur Heart J Cardiovasc Imaging. 2017;18:1283–1291. [DOI] [PubMed] [Google Scholar]
- 102.Ferferieva V, Van den Bergh A, Claus P, Jasaityte R, La Gerche A, Rademakers F, Herijgers P and D’Hooge J. Assessment of strain and strain rate by two-dimensional speckle tracking in mice: comparison with tissue Doppler echocardiography and conductance catheter measurements. Eur Heart J Cardiovasc Imaging. 2013;14:765–73. [DOI] [PubMed] [Google Scholar]
- 103.Hammouda K, Khalifa F, Abdeltawab H, Elnakib A, Giridharan GA, Zhu M, Ng CK, Dassanayaka S, Kong M, Darwish HE, Mohamed TMA, Jones SP and El-Baz A. A New Framework for Performing Cardiac Strain Analysis from Cine MRI Imaging in Mice. Sci Rep. 2020;10:7725. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Chuang JS, Zemljic-Harpf A, Ross RS, Frank LR, McCulloch AD and Omens JH. Determination of three-dimensional ventricular strain distributions in gene-targeted mice using tagged MRI. Magn Reson Med. 2010;64:1281–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Gilson WD, Yang Z, French BA and Epstein FH. Measurement of myocardial mechanics in mice before and after infarction using multislice displacement-encoded MRI with 3D motion encoding. Am J Physiol Heart Circ Physiol. 2005;288:H1491–7. [DOI] [PubMed] [Google Scholar]
- 106.Zhong X, Gibberman LB, Spottiswoode BS, Gilliam AD, Meyer CH, French BA and Epstein FH. Comprehensive cardiovascular magnetic resonance of myocardial mechanics in mice using three-dimensional cine DENSE. J Cardiovasc Magn Reson. 2011;13:83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Hankiewicz JH and Lewandowski ED. Improved cardiac tagging resolution at ultra-high magnetic field elucidates transmural differences in principal strain in the mouse heart and reduced stretch in dilated cardiomyopathy. J Cardiovasc Magn Reson. 2007;9:883–90. [DOI] [PubMed] [Google Scholar]
- 108.Naresh NK, Xu Y, Klibanov AL, Vandsburger MH, Meyer CH, Leor J, Kramer CM, French BA and Epstein FH. Monocyte and/or macrophage infiltration of heart after myocardial infarction: MR imaging by using T1-shortening liposomes. Radiology. 2012;264:428–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Mekkaoui C, Reese TG, Jackowski MP, Bhat H and Sosnovik DE. Diffusion MRI in the heart. NMR Biomed. 2017;30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Scollan DF, Holmes A, Winslow R and Forder J. Histological validation of myocardial microstructure obtained from diffusion tensor magnetic resonance imaging. Am J Physiol. 1998;275:H2308–18. [DOI] [PubMed] [Google Scholar]
- 111.Chen J, Song SK, Liu W, McLean M, Allen JS, Tan J, Wickline SA and Yu X. Remodeling of cardiac fiber structure after infarction in rats quantified with diffusion tensor MRI. Am J Physiol Heart Circ Physiol. 2003;285:H946–54. [DOI] [PubMed] [Google Scholar]
- 112.Hales PW, Schneider JE, Burton RA, Wright BJ, Bollensdorff C and Kohl P. Histo-anatomical structure of the living isolated rat heart in two contraction states assessed by diffusion tensor MRI. Prog Biophys Mol Biol. 2012;110:319–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Sosnovik DE, Wang R, Dai G, Wang T, Aikawa E, Novikov M, Rosenzweig A, Gilbert RJ and Wedeen VJ. Diffusion spectrum MRI tractography reveals the presence of a complex network of residual myofibers in infarcted myocardium. Circ Cardiovasc Imaging. 2009;2:206–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Teh I, McClymont D, Burton RA, Maguire ML, Whittington HJ, Lygate CA, Kohl P and Schneider JE. Resolving Fine Cardiac Structures in Rats with High-Resolution Diffusion Tensor Imaging. Sci Rep. 2016;6:30573. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Ariga R, Tunnicliffe EM, Manohar SG, Mahmod M, Raman B, Piechnik SK, Francis JM, Robson MD, Neubauer S and Watkins H. Identification of Myocardial Disarray in Patients With Hypertrophic Cardiomyopathy and Ventricular Arrhythmias. J Am Coll Cardiol. 2019;73:2493–2502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Mekkaoui C, Huang S, Chen HH, Dai G, Reese TG, Kostis WJ, Thiagalingam A, Maurovich-Horvat P, Ruskin JN, Hoffmann U, Jackowski MP and Sosnovik DE. Fiber architecture in remodeled myocardium revealed with a quantitative diffusion CMR tractography framework and histological validation. J Cardiovasc Magn Reson. 2012;14:70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Mekkaoui C, Jackowski MP, Kostis WJ, Stoeck CT, Thiagalingam A, Reese TG, Reddy VY, Ruskin JN, Kozerke S and Sosnovik DE. Myocardial Scar Delineation Using Diffusion Tensor Magnetic Resonance Tractography. J Am Heart Assoc. 2018;7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.LeGrice IJ, Takayama Y and Covell JW. Transverse shear along myocardial cleavage planes provides a mechanism for normal systolic wall thickening. Circ Res. 1995;77:182–93. [DOI] [PubMed] [Google Scholar]
- 119.Dou J, Tseng WY, Reese TG and Wedeen VJ. Combined diffusion and strain MRI reveals structure and function of human myocardial laminar sheets in vivo. Magn Reson Med. 2003;50:107–13. [DOI] [PubMed] [Google Scholar]
- 120.Nielles-Vallespin S, Khalique Z, Ferreira PF, de Silva R, Scott AD, Kilner P, McGill LA, Giannakidis A, Gatehouse PD, Ennis D, Aliotta E, Al-Khalil M, Kellman P, Mazilu D, Balaban RS, Firmin DN, Arai AE and Pennell DJ. Assessment of Myocardial Microstructural Dynamics by In Vivo Diffusion Tensor Cardiac Magnetic Resonance. J Am Coll Cardiol. 2017;69:661–676. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Spath NB, Lilburn DML, Gray GA, Le Page LM, Papanastasiou G, Lennen RJ, Janiczek RL, Dweck MR, Newby DE, Yang PC, Jansen MA and Semple SI. Manganese-Enhanced T1 Mapping in the Myocardium of Normal and Infarcted Hearts. Contrast Media Mol Imaging. 2018;2018:9641527. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Dash R, Chung J, Ikeno F, Hahn-Windgassen A, Matsuura Y, Bennett MV, Lyons JK, Teramoto T, Robbins RC, McConnell MV, Yeung AC, Brinton TJ, Harnish PP and Yang PC. Dual manganese-enhanced and delayed gadolinium-enhanced MRI detects myocardial border zone injury in a pig ischemia-reperfusion model. Circ Cardiovasc Imaging. 2011;4:574–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Narula J, Acio ER, Narula N, Samuels LE, Fyfe B, Wood D, Fitzpatrick JM, Raghunath PN, Tomaszewski JE, Kelly C, Steinmetz N, Green A, Tait JF, Leppo J, Blankenberg FG, Jain D and Strauss HW. Annexin-V imaging for noninvasive detection of cardiac allograft rejection. Nat Med. 2001;7:1347–52. [DOI] [PubMed] [Google Scholar]
- 124.Sosnovik DE, Schellenberger EA, Nahrendorf M, Novikov MS, Matsui T, Dai G, Reynolds F, Grazette L, Rosenzweig A, Weissleder R and Josephson L. Magnetic resonance imaging of cardiomyocyte apoptosis with a novel magneto-optical nanoparticle. Magn Reson Med. 2005;54:718–24. [DOI] [PubMed] [Google Scholar]
- 125.Hiller KH, Waller C, Nahrendorf M, Bauer WR and Jakob PM. Assessment of cardiovascular apoptosis in the isolated rat heart by magnetic resonance molecular imaging. Mol Imaging. 2006;5:115–21. [PubMed] [Google Scholar]
- 126.Sarda-Mantel L, Michel JB, Rouzet F, Martet G, Louedec L, Vanderheyden JL, Hervatin F, Raguin O, Vrigneaud JM, Khaw BA and Le Guludec D. (99m)Tc-annexin V and (111)In-antimyosin antibody uptake in experimental myocardial infarction in rats. Eur J Nucl Med Mol Imaging. 2006;33:239–45. [DOI] [PubMed] [Google Scholar]
- 127.Sosnovik DE, Garanger E, Aikawa E, Nahrendorf M, Figuiredo JL, Dai G, Reynolds F, Rosenzweig A, Weissleder R and Josephson L. Molecular MRI of cardiomyocyte apoptosis with simultaneous delayed-enhancement MRI distinguishes apoptotic and necrotic myocytes in vivo: potential for midmyocardial salvage in acute ischemia. Circ Cardiovasc Imaging. 2009;2:460–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Chen HH, Yuan H, Cho H, Feng Y, Ngoy S, Kumar AT, Liao R, Chao W, Josephson L and Sosnovik DE. Theranostic Nucleic Acid Binding Nanoprobe Exerts Anti-inflammatory and Cytoprotective Effects in Ischemic Injury. Theranostics. 2017;7:814–825. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Chaudhry F, Adapoe M, Johnson KW, Narula N, Shekhar A, Kawai H, Horwitz JK, Liu J, Li Y, Pak KY, Mattis J, Moreira AL, Levy PD, Strauss HW, Petrov A, Heeger PS and Narula J. Molecular Imaging of Cardiac Allograft Rejection: Targeting Apoptosis With Radiolabeled Duramycin. JACC Cardiovasc Imaging. 2020;13:1438–1441. [DOI] [PubMed] [Google Scholar]
- 130.Liu Z, Zhao M, Zhu X, Furenlid LR, Chen YC and Barrett HH. In vivo dynamic imaging of myocardial cell death using 99mTc-labeled C2A domain of synaptotagmin I in a rat model of ischemia and reperfusion. Nucl Med Biol. 2007;34:907–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Chen HH, Mekkaoui C, Cho H, Ngoy S, Marinelli B, Waterman P, Nahrendorf M, Liao R, Josephson L and Sosnovik DE. Fluorescence tomography of rapamycin-induced autophagy and cardioprotection in vivo. Circ Cardiovasc Imaging. 2013;6:441–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Sosnovik DE, Nahrendorf M, Deliolanis N, Novikov M, Aikawa E, Josephson L, Rosenzweig A, Weissleder R and Ntziachristos V. Fluorescence tomography and magnetic resonance imaging of myocardial macrophage infiltration in infarcted myocardium in vivo. Circulation. 2007;115:1384–91. [DOI] [PubMed] [Google Scholar]
- 133.Ye YX, Basse-Lusebrink TC, Arias-Loza PA, Kocoski V, Kampf T, Gan Q, Bauer E, Sparka S, Helluy X, Hu K, Hiller KH, Boivin-Jahns V, Jakob PM, Jahns R and Bauer WR. Monitoring of monocyte recruitment in reperfused myocardial infarction with intramyocardial hemorrhage and microvascular obstruction by combined fluorine 19 and proton cardiac magnetic resonance imaging. Circulation. 2013;128:1878–88. [DOI] [PubMed] [Google Scholar]
- 134.Flogel U, Ding Z, Hardung H, Jander S, Reichmann G, Jacoby C, Schubert R and Schrader J. In vivo monitoring of inflammation after cardiac and cerebral ischemia by fluorine magnetic resonance imaging. Circulation. 2008;118:140–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.van Heeswijk RB, De Blois J, Kania G, Gonzales C, Blyszczuk P, Stuber M, Eriksson U and Schwitter J. Selective in vivo visualization of immune-cell infiltration in a mouse model of autoimmune myocarditis by fluorine-19 cardiac magnetic resonance. Circ Cardiovasc Imaging. 2013;6:277–84. [DOI] [PubMed] [Google Scholar]
- 136.Senders ML, Meerwaldt AE, van Leent MMT, Sanchez-Gaytan BL, van de Voort JC, Toner YC, Maier A, Klein ED, Sullivan NAT, Sofias AM, Groenen H, Faries C, Oosterwijk RS, van Leeuwen EM, Fay F, Chepurko E, Reiner T, Duivenvoorden R, Zangi L, Dijkhuizen RM, Hak S, Swirski FK, Nahrendorf M, Perez-Medina C, Teunissen AJP, Fayad ZA, Calcagno C, Strijkers GJ and Mulder WJM. Probing myeloid cell dynamics in ischaemic heart disease by nanotracer hot-spot imaging. Nat Nanotechnol. 2020;15:398–405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Nahrendorf M, Sosnovik DE, Waterman P, Swirski FK, Pande AN, Aikawa E, Figueiredo JL, Pittet MJ and Weissleder R. Dual channel optical tomographic imaging of leukocyte recruitment and protease activity in the healing myocardial infarct. Circ Res. 2007;100:1218–25. [DOI] [PubMed] [Google Scholar]
- 138.Nahrendorf M, Sosnovik D, Chen JW, Panizzi P, Figueiredo JL, Aikawa E, Libby P, Swirski FK and Weissleder R. Activatable magnetic resonance imaging agent reports myeloperoxidase activity in healing infarcts and noninvasively detects the antiinflammatory effects of atorvastatin on ischemia-reperfusion injury. Circulation. 2008;117:1153–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Thackeray JT, Bankstahl JP, Wang Y, Korf-Klingebiel M, Walte A, Wittneben A, Wollert KC and Bengel FM. Targeting post-infarct inflammation by PET imaging: comparison of (68)Ga-citrate and (68)Ga-DOTATATE with (18)F-FDG in a mouse model. Eur J Nucl Med Mol Imaging. 2015;42:317–27. [DOI] [PubMed] [Google Scholar]
- 140.Boutagy NE, Wu J, Cai Z, Zhang W, Booth CJ, Kyriakides TC, Pfau D, Mulnix T, Liu Z, Miller EJ, Young LH, Carson RE, Huang Y, Liu C and Sinusas AJ. In Vivo Reactive Oxygen Species Detection With a Novel Positron Emission Tomography Tracer, (18)F-DHMT, Allows for Early Detection of Anthracycline-Induced Cardiotoxicity in Rodents. JACC Basic Transl Sci. 2018;3:378–390. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Nahrendorf M, Hoyer FF, Meerwaldt AE, van Leent MMT, Senders ML, Calcagno C, Robson PM, Soultanidis G, Perez-Medina C, Teunissen AJP, Toner YC, Ishikawa K, Fish K, Sakurai K, van Leeuwen EM, Klein ED, Sofias AM, Reiner T, Rohde D, Aguirre AD, Wojtkiewicz G, Schmidt S, Iwamoto Y, Izquierdo-Garcia D, Caravan P, Swirski FK, Weissleder R and Mulder WJM. Imaging Cardiovascular and Lung Macrophages With the Positron Emission Tomography Sensor (64)Cu-Macrin in Mice, Rabbits, and Pigs. Circ Cardiovasc Imaging. 2020;13:e010586. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Makowski MR, Rischpler C, Ebersberger U, Keithahn A, Kasel M, Hoffmann E, Rassaf T, Kessler H, Wester HJ, Nekolla SG, Schwaiger M and Beer AJ. Multiparametric PET and MRI of myocardial damage after myocardial infarction: correlation of integrin alphavbeta3 expression and myocardial blood flow. Eur J Nucl Med Mol Imaging. 2021;48:1070–1080. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Meoli DF, Sadeghi MM, Krassilnikova S, Bourke BN, Giordano FJ, Dione DP, Su H, Edwards DS, Liu S, Harris TD, Madri JA, Zaret BL and Sinusas AJ. Noninvasive imaging of myocardial angiogenesis following experimental myocardial infarction. J Clin Invest. 2004;113:1684–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.van den Borne SW, Isobe S, Verjans JW, Petrov A, Lovhaug D, Li P, Zandbergen HR, Ni Y, Frederik P, Zhou J, Arbo B, Rogstad A, Cuthbertson A, Chettibi S, Reutelingsperger C, Blankesteijn WM, Smits JF, Daemen MJ, Zannad F, Vannan MA, Narula N, Pitt B, Hofstra L and Narula J. Molecular imaging of interstitial alterations in remodeling myocardium after myocardial infarction. J Am Coll Cardiol. 2008;52:2017–28. [DOI] [PubMed] [Google Scholar]
- 145.Varasteh Z, Mohanta S, Robu S, Braeuer M, Li Y, Omidvari N, Topping G, Sun T, Nekolla SG, Richter A, Weber C, Habenicht A, Haberkorn UA and Weber WA. Molecular Imaging of Fibroblast Activity After Myocardial Infarction Using a (68)Ga-Labeled Fibroblast Activation Protein Inhibitor, FAPI-04. J Nucl Med. 2019;60:1743–1749. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Chen J, Tung CH, Allport JR, Chen S, Weissleder R and Huang PL. Near-infrared fluorescent imaging of matrix metalloproteinase activity after myocardial infarction. Circulation. 2005;111:1800–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Su H, Spinale FG, Dobrucki LW, Song J, Hua J, Sweterlitsch S, Dione DP, Cavaliere P, Chow C, Bourke BN, Hu XY, Azure M, Yalamanchili P, Liu R, Cheesman EH, Robinson S, Edwards DS and Sinusas AJ. Noninvasive targeted imaging of matrix metalloproteinase activation in a murine model of postinfarction remodeling. Circulation. 2005;112:3157–67. [DOI] [PubMed] [Google Scholar]
- 148.Werner RA, Hess A, Koenig T, Diekmann J, Derlin T, Melk A, Thackeray JT, Bauersachs J and Bengel FM. Molecular imaging of inflammation crosstalk along the cardio-renal axis following acute myocardial infarction. Theranostics. 2021;11:7984–7994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Wildgruber M, Bielicki I, Aichler M, Kosanke K, Feuchtinger A, Settles M, Onthank DC, Cesati RR, Robinson SP, Huber AM, Rummeny EJ, Walch AK and Botnar RM. Assessment of myocardial infarction and postinfarction scar remodeling with an elastin-specific magnetic resonance agent. Circ Cardiovasc Imaging. 2014;7:321–9. [DOI] [PubMed] [Google Scholar]
- 150.Akam EA, Abston E, Rotile NJ, Slattery HR, Zhou IY, Lanuti M and Caravan P. Improving the reactivity of hydrazine-bearing MRI probes for in vivo imaging of lung fibrogenesis. Chem Sci. 2020;11:224–231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Kraitchman DL, Heldman AW, Atalar E, Amado LC, Martin BJ, Pittenger MF, Hare JM and Bulte JW. In vivo magnetic resonance imaging of mesenchymal stem cells in myocardial infarction. Circulation. 2003;107:2290–3. [DOI] [PubMed] [Google Scholar]
- 152.Azene N, Fu Y, Maurer J and Kraitchman DL. Tracking of stem cells in vivo for cardiovascular applications. J Cardiovasc Magn Reson. 2014;16:7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Sheikh AY, Huber BC, Narsinh KH, Spin JM, van der Bogt K, de Almeida PE, Ransohoff KJ, Kraft DL, Fajardo G, Ardigo D, Ransohoff J, Bernstein D, Fischbein MP, Robbins RC and Wu JC. In vivo functional and transcriptional profiling of bone marrow stem cells after transplantation into ischemic myocardium. Arterioscler Thromb Vasc Biol. 2012;32:92–102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Cao F, Lin S, Xie X, Ray P, Patel M, Zhang X, Drukker M, Dylla SJ, Connolly AJ, Chen X, Weissman IL, Gambhir SS and Wu JC. In vivo visualization of embryonic stem cell survival, proliferation, and migration after cardiac delivery. Circulation. 2006;113:1005–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Dayeh NR, Tardif JC, Shi Y, Tanguay M, Ledoux J and Dupuis J. Echocardiographic validation of pulmonary hypertension due to heart failure with reduced ejection fraction in mice. Sci Rep. 2018;8:1363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Kang Y, Zhang G, Huang EC, Huang J, Cai J, Cai L, Wang S and Keller BB. Sulforaphane prevents right ventricular injury and reduces pulmonary vascular remodeling in pulmonary arterial hypertension. Am J Physiol Heart Circ Physiol. 2020;318:H853–H866. [DOI] [PubMed] [Google Scholar]
- 157.Zhong G, Li Y, Li H, Sun W, Cao D, Li J, Zhao D, Song J, Jin X, Song H, Yuan X, Wu X, Li Q, Xu Q, Kan G, Cao H, Ling S and Li Y. Simulated Microgravity and Recovery-Induced Remodeling of the Left and Right Ventricle. Front Physiol. 2016;7:274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Thibault HB, Kurtz B, Raher MJ, Shaik RS, Waxman A, Derumeaux G, Halpern EF, Bloch KD and Scherrer-Crosbie M. Noninvasive assessment of murine pulmonary arterial pressure: validation and application to models of pulmonary hypertension. Circ Cardiovasc Imaging. 2010;3:157–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Das M, Boerma M, Goree JR, Lavoie EG, Fausther M, Gubrij IB, Pangle AK, Johnson LG and Dranoff JA. Pathological changes in pulmonary circulation in carbon tetrachloride (CCl4)-induced cirrhotic mice. PLoS One. 2014;9:e96043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Kimura D, Saravia J, Jaligama S, McNamara I, Vu LD, Sullivan RD, Mancarella S, You D and Cormier SA. New mouse model of pulmonary hypertension induced by respiratory syncytial virus bronchiolitis. Am J Physiol Heart Circ Physiol. 2018;315:H581–H589. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Dufva MJ, Boehm M, Ichimura K, Truong U, Qin X, Tabakh J, Hunter KS, Ivy D, Spiekerkoetter E and Kheyfets VO. Pulmonary arterial banding in mice may be a suitable model for studies on ventricular mechanics in pediatric pulmonary arterial hypertension. J Cardiovasc Magn Reson. 2021;23:66. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
