Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Junjie Chen is active.

Publication


Featured researches published by Junjie Chen.


The FASEB Journal | 2007

19F magnetic resonance imaging for stem/progenitor cell tracking with multiple unique perfluorocarbon nanobeacons

Kathryn C. Partlow; Junjie Chen; Jason A. Brant; Anne M. Neubauer; Todd E. Meyerrose; Michael H. Creer; Jan A. Nolta; Shelton D. Caruthers; Gregory M. Lanza; Samuel A. Wickline

MRI has been employed to elucidate the migratory behavior of stem/progenitor cells noninva‐sively in vivo with traditional proton (1H) imaging of iron oxide nanoparticle‐labeled cells. Alternatively, we demonstrate that fluorine (19F) MRI of cells labeled with different types of liquid perfluorocarbon (PFC) nanoparticles produces unique and sensitive cell markers distinct from any tissue background signal. To define the utility for cell tracking, mononuclear cells harvested from human umbilical cord blood were grown under proendothelial conditions and labeled with nanoparticles composed of two distinct PFC cores (perfluorooctylbromide and perfluoro‐15‐crown‐5 ether). The sensitivity for detecting and imaging labeled cells was defined on 11.7T (research) and 1.5T (clinical) scanners. Stem/progenitor cells (CD34+CD133+CD31+) readily internalized PFC nanoparticles without aid of adjunctive labeling techniques, and cells remained functional in vivo. PFC‐labeled cells exhibited distinct 19F signals and were readily detected after both local and intravenous injection. PFC nanoparticles provide an unequivocal and unique signature for stem/progenitor cells, enable spatial cell localization with 19F MRI, and permit quantification and detection of multiple fluorine signatures via 19F MR spectroscopy. This method should facilitate longitudinal investigation of cellular events in vivo for multiple cell types simultaneously.—Partlow, K. C., Chen, J., Brant, J. A., Neubauer, A. M., Meyerrose, T. E., Creer, M. H., Nolta, J. A., Caruthers, S. D., Lanza, G. M., Wickline, S. A. 19F magnetic resonance imaging for stem/progenitor cell tracking with multiple unique perfluorocarbon nanobeacons. FASEB J. 21, 1647–1654 (2007)


Magnetic Resonance in Medicine | 2000

High-resolution MRI characterization of human thrombus using a novel fibrin-targeted paramagnetic nanoparticle contrast agent

Xin Yu; Sheng-Kwei Song; Junjie Chen; Michael J. Scott; Ralph J. Fuhrhop; Christopher S. Hall; Patrick J. Gaffney; Samuel A. Wickline; Gregory M. Lanza

In this study, the sensitivity of a novel fibrin‐targeted contrast agent for fibrin detection was defined in vitro on human thrombus. The contrast agent was a lipid‐encapsulated perfluorocarbon nanoparticle with numerous Gd‐DTPA complexes incorporated into the outer surface. After binding to fibrin clots, scanning electron microscopy of treated clots revealed dense accumulation of nanoparticles on the clot surfaces. Fibrin clots with sizes ranging from 0.5–7.0 mm were imaged at 4.7 T with or without treatment with the targeted contrast agent. Regardless of sizes, untreated clots were not detectable by T1‐weighted MRI, while targeted contrast agent dramatically improved the detectability of all clots. Decreases in T1 and T2 relaxation times (20–40%) were measured relative to the surrounding media and the control clots. These results suggest the potential for sensitive and specific detection of microthrombi that form on the intimal surfaces of unstable atherosclerotic plaque. Magn Reson Med 44:867–872, 2000.


Magnetic Resonance in Medicine | 2003

Improved molecular imaging contrast agent for detection of human thrombus

Patrick M. Winter; Shelton D. Caruthers; Xin Yu; Sheng-Kwei Song; Junjie Chen; Brad Miller; Jeff W. M. Bulte; J. David Robertson; Patrick J. Gaffney; Samuel A. Wickline; Gregory M. Lanza

Molecular imaging of microthrombus within fissures of unstable atherosclerotic plaques requires sensitive detection with a thrombus‐specific agent. Effective molecular imaging has been previously demonstrated with fibrin‐targeted Gd‐DTPA‐bis‐oleate (BOA) nanoparticles. In this study, the relaxivity of an improved fibrin‐targeted paramagnetic formulation, Gd‐DTPA‐phosphatidylethanolamine (PE), was compared with Gd‐DTPA‐BOA at 0.05‐4.7 T. Ion‐ and particle‐based r1 relaxivities (1.5 T) for Gd‐DTPA‐PE (33.7 (s*mM)‐1 and 2.48 × 106 (s*mM)‐1, respectively) were about twofold higher than for Gd‐DTPA‐BOA, perhaps due to faster water exchange with surface gadolinium. Gd‐DTPA‐PE nanoparticles bound to thrombus surfaces via anti‐fibrin antibodies (1H10) induced 72% ± 5% higher change in R1 values at 1.5 T (ΔR1 = 0.77 ± 0.02 1/s) relative to Gd‐DTPA‐BOA (ΔR1 = 0.45 ± 0.02 1/s). These studies demonstrate marked improvement in a fibrin‐specific molecular imaging agent that might allow sensitive, early detection of vascular microthrombi, the antecedent to stroke and heart attack. Magn Reson Med 50:411–416, 2003.


Magnetic Resonance in Medicine | 2006

Targeted PARACEST nanoparticle contrast agent for the detection of fibrin.

Patrick M. Winter; Kejia Cai; Junjie Chen; Christopher Adair; Garry E. Kiefer; Phillip S. Athey; Patrick J. Gaffney; Carolyn E. Buff; J. David Robertson; Shelton D. Caruthers; Samuel A. Wickline; Gregory M. Lanza

A lipid‐encapsulated perfluorocarbon nanoparticle molecular imaging contrast agent that utilizes a paramagnetic chemical exchange saturation transfer (PARACEST) chelate is presented. PARACEST agents are ideally suited for molecular imaging applications because one can switch the contrast on and off at will simply by adjusting the pulse sequence parameters. This obviates the need for pre‐ and postinjection images to define contrast agent binding. Spectroscopy (4.7T) of PARACEST nanoparticles revealed a bound water peak at 52 ppm, in agreement with results from the water‐soluble chelate. Imaging of control nanoparticles showed no appreciable contrast, while PARACEST nanoparticles produced >10% signal enhancement. PARACEST nanoparticles were targeted to clots via antifibrin antibodies and produced a contrast‐to‐noise ratio (CNR) of 10 at the clot surface. Magn Reson Med, 2006.


Circulation | 2009

Resolution of Established Cardiac Hypertrophy and Fibrosis and Prevention of Systolic Dysfunction in a Transgenic Rabbit Model of Human Cardiomyopathy Through Thiol-Sensitive Mechanisms

Raffaella Lombardi; Gabriela Rodriguez; Suet Nee Chen; Crystal M. Ripplinger; Wenwen Li; Junjie Chen; James T. Willerson; Sandro Betocchi; Samuel A. Wickline; Igor R. Efimov; Ali J. Marian

Background— Cardiac hypertrophy, the clinical hallmark of hypertrophic cardiomyopathy (HCM), is a major determinant of morbidity and mortality not only in HCM but also in a number of cardiovascular diseases. There is no effective therapy for HCM and generally for cardiac hypertrophy. Myocardial oxidative stress and thiol-sensitive signaling molecules are implicated in pathogenesis of hypertrophy and fibrosis. We posit that treatment with N-acetylcysteine, a precursor of glutathione, the largest intracellular thiol pool against oxidative stress, could reverse cardiac hypertrophy and fibrosis in HCM. Methods and Results— We treated 2-year-old β-myosin heavy-chain Q403 transgenic rabbits with established cardiac hypertrophy and preserved systolic function with N-acetylcysteine or a placebo for 12 months (n=10 per group). Transgenic rabbits in the placebo group had cardiac hypertrophy, fibrosis, systolic dysfunction, increased oxidized to total glutathione ratio, higher levels of activated thiol-sensitive active protein kinase G, dephosphorylated nuclear factor of activated T cells (NFATc1) and phospho-p38, and reduced levels of glutathiolated cardiac α-actin. Treatment with N-acetylcysteine restored oxidized to total glutathione ratio, normalized levels of glutathiolated cardiac α-actin, reversed cardiac and myocyte hypertrophy and interstitial fibrosis, reduced the propensity for ventricular arrhythmias, prevented cardiac dysfunction, restored myocardial levels of active protein kinase G, and dephosphorylated NFATc1 and phospho-p38. Conclusions— Treatment with N-acetylcysteine, a safe prodrug against oxidation, reversed established cardiac phenotype in a transgenic rabbit model of human HCM. Because there is no effective pharmacological therapy for HCM and given that hypertrophy, fibrosis, and cardiac dysfunction are common and major predictors of clinical outcomes, the findings could have implications in various cardiovascular disorders.


Magnetic Resonance in Medicine | 2008

Gadolinium-modulated 19F signals from perfluorocarbon nanoparticles as a new strategy for molecular imaging.

Anne M. Neubauer; Jacob W Myerson; Shelton D. Caruthers; Franklin D. Hockett; Patrick M. Winter; Junjie Chen; Patrick J. Gaffney; J. David Robertson; Gregory M. Lanza; Samuel A. Wickline

Recent advances in the design of fluorinated nanoparticles for molecular magnetic resonance imaging (MRI) have enabled specific detection of 19F nuclei, providing unique and quantifiable spectral signatures. However, a pressing need for signal enhancement exists because the total 19F in imaging voxels is often limited. By directly incorporating a relaxation agent, gadolinium (Gd), into the lipid monolayer that surrounds the perfluorocarbon (PFC), a marked augmentation of the 19F signal from 200‐nm nanoparticles was achieved. This design increases the magnetic relaxation rate of the 19F nuclei fourfold at 1.5 T and effects a 125% increase in signal—an effect that is maintained when they are targeted to human plasma clots. By varying the surface concentration of Gd, the relaxation effect can be quantitatively modulated to tailor particle properties. This novel strategy dramatically improves the sensitivity and range of 19F MRI/MRS and forms the basis for designing contrast agents capable of sensing their surface chemistry. Magn Reson Med 60:1066–1072, 2008.


Wiley Interdisciplinary Reviews-nanomedicine and Nanobiotechnology | 2010

Quantitative magnetic resonance fluorine imaging: today and tomorrow

Junjie Chen; Gregory M. Lanza; Samuel A. Wickline

Fluorine (19F) is a promising moiety for quantitative magnetic resonance imaging (MRI). It possesses comparable magnetic resonance (MR) sensitivity to proton (1H) but exhibits no tissue background signal, allowing specific and selective assessment of the administrated 19F-containing compounds in vivo. Additionally, the MR spectra of 19F-containing compounds exhibited a wide range of chemical shifts (>200 ppm). Therefore, both MR parameters (e.g., spin-lattice relaxation rate R1) and the absolute quantity of molecule can be determined with 19F MRI for unbiased assessment of tissue physiology and pathology. This article reviews quantitative 19F MRI applications for mapping tumor oxygenation, assessing molecular expression in vascular diseases, and tracking labeled stem cells.


Circulation Research | 2007

Enhanced transmural fiber rotation and connexin 43 heterogeneity are associated with an increased upper limit of vulnerability in a transgenic rabbit model of human hypertrophic cardiomyopathy

Crystal M. Ripplinger; Wenwen Li; Jennifer Hadley; Junjie Chen; Florence Rothenberg; Raffaella Lombardi; Samuel A. Wickline; Ali J. Marian; Igor R. Efimov

Human hypertrophic cardiomyopathy, characterized by cardiac hypertrophy and myocyte disarray, is the most common cause of sudden cardiac death in the young. Hypertrophic cardiomyopathy is often caused by mutations in sarcomeric genes. We sought to determine arrhythmia propensity and underlying mechanisms contributing to arrhythmia in a transgenic (TG) rabbit model (&bgr;-myosin heavy chain–Q403) of human hypertrophic cardiomyopathy. Langendorff-perfused hearts from TG (n=6) and wild-type (WT) rabbits (n=6) were optically mapped. The upper and lower limits of vulnerability, action potential duration (APD) restitution, and conduction velocity were measured. The transmural fiber angle shift was determined using diffusion tensor MRI. The transmural distribution of connexin 43 was quantified with immunohistochemistry. The upper limit of vulnerability was significantly increased in TG versus WT hearts (13.3±2.1 versus 7.4±2.3 V/cm; P=3.2e−5), whereas the lower limits of vulnerability were similar. APD restitution, conduction velocities, and anisotropy were also similar. Left ventricular transmural fiber rotation was significantly higher in TG versus WT hearts (95.6±10.9° versus 79.2±7.8°; P=0.039). The connexin 43 density was significantly increased in the mid-myocardium of TG hearts compared with WT (5.46±2.44% versus 2.68±0.77%; P=0.024), and similar densities were observed in the endo- and epicardium. Because a nearly 2-fold increase in upper limit of vulnerability was observed in the TG hearts without significant changes in APD restitution, conduction velocity, or the anisotropy ratio, we conclude that structural remodeling may underlie the elevated upper limit of vulnerability in human hypertrophic cardiomyopathy.


Journal of Cardiovascular Magnetic Resonance | 2008

Detection and quantification of angiogenesis in experimental valve disease with integrin-targeted nanoparticles and 19-fluorine MRI/MRS

Emily A. Waters; Junjie Chen; John S. Allen; Huiying Zhang; Gregory M. Lanza; Samuel A. Wickline

BackgroundAngiogenesis is a critical early feature of atherosclerotic plaque development and may also feature prominently in the pathogenesis of aortic valve stenosis. It has been shown that MRI can detect and quantify specific molecules of interest expressed in cardiovascular disease and cancer by measuring the unique fluorine signature of appropriately targeted perfluorocarbon (PFC) nanoparticles. In this study, we demonstrated specific binding of ανβ3 integrin targeted nanoparticles to neovasculature in a rabbit model of aortic valve disease. We also showed that fluorine MRI could be used to detect and quantify the development of neovasculature in the excised aortic valve leaflets.MethodsNew Zealand White rabbits consumed a cholesterol diet for ~180 days and developed aortic valve thickening, inflammation, and angiogenesis mimicking early human aortic valve disease. Rabbits (n = 7) were treated with ανβ3 integrin targeted PFC nanoparticles or control untargeted PFC nanoparticles (n = 6). Competitive inhibition in vivo of nanoparticle binding (n = 4) was tested by pretreatment with targeted nonfluorinated nanoparticles followed 2 hours later by targeted PFC nanoparticles. 2 hours after treatment, aortic valves were excised and 19F MRS was performed at 11.7T. Integrated 19F spectral peaks were compared using a one-way ANOVA and Hsus MCB (multiple comparisons with the best) post hoc t test. In 3 additional rabbits treated with ανβ3 integrin targeted PFC nanoparticles, 19F spectroscopy was performed on a 3.0T clinical scanner. The presence of angiogenesis was confirmed by immunohistochemistry.ResultsValves of rabbits treated with targeted PFC nanoparticles had 220% more fluorine signal than valves of rabbits treated with untargeted PFC nanoparticles (p < 0.001). Pretreatment of rabbits with targeted oil-based nonsignaling nanoparticles reduced the fluorine signal by 42% due to competitive inhibition, to a level not significantly different from control animals. Nanoparticles were successfully detected in all samples scanned at 3.0T. PECAM endothelial staining and ανβ3 integrin staining revealed the presence of neovasculature within the valve leaflets.ConclusionIntegrin-targeted PFC nanoparticles specifically detect early angiogenesis in sclerotic aortic valves of cholesterol fed rabbits. These techniques may be useful for assessing atherosclerotic components of preclinical aortic valve disease in patients and could assist in defining efficacy of medical therapies.


Magnetic Resonance in Medicine | 2004

Harmonic phase MR tagging for direct quantification of Lagrangian strain in rat hearts after myocardial infarction.

Wei Liu; Junjie Chen; Songbai Ji; J. Stacy Allen; Philip V. Bayly; Samuel A. Wickline; Xin Yu

The utility of harmonic phase (HARP) analysis was recently demonstrated in humans and large animals as a technique for rapid and automatic analysis of tagged magnetic resonance images. In the current study, the applicability and accuracy of HARP analysis for automatic strain quantification in small animals were investigated. A validation study was performed on seven postinfarct rats and seven age‐matched controls. A method for direct computation of 2D Lagrangian strain fields from spatial derivatives of HARP images was also developed in this paper. The results of HARP analysis were evaluated by comparison with those of homogeneous strain analysis employing finite element method and manual tag tracking. Both methods were validated with simulated digital images. Compared to conventional homogeneous strain analysis, HARP analysis yielded similar results in the assessment of regional strain patterns in both control and infarct rats. Both methods detected a reduction in maximal stretch and shortening in infarct rats. Our results suggest that HARP analysis can also be applied to quantify alterations in regional myocardial wall motion in small animals. Magn Reson Med 52:1282–1290, 2004.

Collaboration


Dive into the Junjie Chen's collaboration.

Top Co-Authors

Avatar

Samuel A. Wickline

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Gregory M. Lanza

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Huiying Zhang

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Shelton D. Caruthers

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Sheng-Kwei Song

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Lei Zhang

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Xiaoxia Yang

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Lingzhi Hu

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

John S. Allen

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Qing Wang

Washington University in St. Louis

View shared research outputs
Researchain Logo
Decentralizing Knowledge