Network


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

Hotspot


Dive into the research topics where Amanda A. Riley is active.

Publication


Featured researches published by Amanda A. Riley.


Science Translational Medicine | 2014

The HMGB1-RAGE axis mediates traumatic brain injury–induced pulmonary dysfunction in lung transplantation

Daniel J. Weber; Adam Gracon; Matthew S. Ripsch; Amanda J. Fisher; Bo M. Cheon; Pankita H. Pandya; Ragini Vittal; Maegan L. Capitano; Youngsook Kim; Yohance M. Allette; Amanda A. Riley; Brian P. McCarthy; Paul R. Territo; Gary D. Hutchins; Hal E. Broxmeyer; George E. Sandusky; Fletcher A. White; David S. Wilkes

Traumatic brain injury induces acute lung injury that negatively impacts the physiology of the donor lung before and after lung transplantation. Sounding the Alarm for RAGE Only 20% of lungs are transplantable because traumatic brain injury, a major cause of death in organ doors, may induce acute lung injury. High-mobility group box-1 (HMGB1) release from the injured brain likely contributes to acute lung injury in donors by preferentially interacting with receptor for advanced glycation end products (RAGE) in the lung. Blocking the HMGB1-RAGE axis improves lung function in murine donors with traumatic brain injury and after transplant. In translational studies, lungs sourced from donors with high HMGB1 levels had worse pulmonary function after transplant. Targeting the HMGB1-RAGE axis may increase the number of lungs available for transplantation and improve patient outcomes. Traumatic brain injury (TBI) results in systemic inflammatory responses that affect the lung. This is especially critical in the setting of lung transplantation, where more than half of donor allografts are obtained postmortem from individuals with TBI. The mechanism by which TBI causes pulmonary dysfunction remains unclear but may involve the interaction of high-mobility group box-1 (HMGB1) protein with the receptor for advanced glycation end products (RAGE). To investigate the role of HMGB1 and RAGE in TBI-induced lung dysfunction, RAGE-sufficient (wild-type) or RAGE-deficient (RAGE−/−) C57BL/6 mice were subjected to TBI through controlled cortical impact and studied for cardiopulmonary injury. Compared to control animals, TBI induced systemic hypoxia, acute lung injury, pulmonary neutrophilia, and decreased compliance (a measure of the lungs’ ability to expand), all of which were attenuated in RAGE−/− mice. Neutralizing systemic HMGB1 induced by TBI reversed hypoxia and improved lung compliance. Compared to wild-type donors, lungs from RAGE−/− TBI donors did not develop acute lung injury after transplantation. In a study of clinical transplantation, elevated systemic HMGB1 in donors correlated with impaired systemic oxygenation of the donor lung before transplantation and predicted impaired oxygenation after transplantation. These data suggest that the HMGB1-RAGE axis plays a role in the mechanism by which TBI induces lung dysfunction and that targeting this pathway before transplant may improve recipient outcomes after lung transplantation.


The Journal of Nuclear Medicine | 2017

Characterization of 11C-GSK1482160 for Targeting the P2X7 Receptor as a Biomarker for Neuroinflammation

Paul R. Territo; Jill A. Meyer; Jonathan Peters; Amanda A. Riley; Brian P. McCarthy; Mingzhang Gao; Min Wang; Mark A. Green; Qi-Huang Zheng; Gary D. Hutchins

The purinergic receptor subtype 7 (P2X7R) represents a novel molecular target for imaging neuroinflammation via PET. GSK1482160, a potent P2X7R antagonist, has high receptor affinity, high blood–brain barrier penetration, and the ability to be radiolabeled with 11C. We report the initial physical and biologic characterization of this novel ligand. Methods: 11C-GSK1482160 was synthesized according to published methods. Cell density studies were performed on human embryonic kidney cell lines expressing human P2X7R (HEK293-hP2X7R) and underwent Western blotting, an immunofluorescence assay, and radioimmunohistochemistry analysis using P2X7R polyclonal antibodies. Receptor density and binding potential were determined by saturation and association–disassociation kinetics, respectively. Peak immune response to lipopolysaccharide treatment in mice was determined in time course studies and analyzed via Iba1 and P2X7R Western blotting and Iba1 immunohistochemistry. Whole-animal biodistribution studies were performed on saline- or lipopolysaccharide-treated mice at 15, 30, and 60 min after radiotracer administration. Dynamic in vivo PET/CT was performed on the mice at 72 h after administration of saline, lipopolysaccharide, or lipopolysaccharide + blocking, and 2-compartment, 5-parameter tracer kinetic modeling of brain regions was performed. Results: P2X7R changed linearly with concentrations or cell numbers. For high-specific-activity 11C-GSK1482160, receptor density and Kd were 1.15 ± 0.12 nM and 3.03 ± 0.10 pmol/mg, respectively, in HEK293-hP2X7R membranes. Association constant kon, dissociation constant koff, and binding potential (kon/koff) in HEK293-hP2X7R cells were 0.2312 ± 0.01542 min−1⋅nM−1, 0.2547 ± 0.0155 min−1, and 1.0277 ± 0.207, respectively. Whole-brain Iba1 expression in lipopolysaccharide-treated mice peaked by 72 h on immunohistochemistry, and Western blot analysis of P2X7R for saline- and lipopolysaccharide-treated brain sections showed a respective 1.8- and 1.7-fold increase in signal enhancement at 72 h. Biodistribution of 11C-GSK1482160 in saline- and lipopolysaccharide-treated mice at 72 h was statistically significant across all tissues studied. In vivo dynamic 11C-GSK1482160 PET/CT of mice at 72 h after administration of saline, lipopolysaccharide, or lipopolysaccharide + blocking showed a 3.2-fold increase and 97% blocking by 30 min. The total distribution volumes for multiple cortical regions and the hippocampus showed statistically significant increases and were blocked by an excess of authentic standard GSK1482160. Conclusion: The current study provides compelling data that support the suitability of 11C-GSK1482160 as a radioligand targeting P2X7R, a biomarker of neuroinflammation.


Journal of Bone and Mineral Research | 2015

In Vivo UTE‐MRI Reveals Positive Effects of Raloxifene on Skeletal‐Bound Water in Skeletally Mature Beagle Dogs

Matthew R. Allen; Paul R. Territo; Chen Lin; Scott Persohn; Lei Jiang; Amanda A. Riley; Brian P. McCarthy; Christopher L. Newman; David B. Burr; Gary D. Hutchins

Raloxifene positively affects mechanical properties of the bone matrix in part through modification of skeletal‐bound water. The goal of this study was to determine if raloxifene‐induced alterations in skeletal hydration could be measured in vivo using ultra‐short echotime magnetic resonance imaging (UTE‐MRI). Twelve skeletally mature female beagle dogs (n = 6/group) were treated for 6 months with oral doses of saline vehicle (VEH, 1 mL/kg/d) or raloxifene (RAL, 0.5 mg/kg/d). After 6 months of treatment, all animals underwent in vivo UTE‐MRI of the proximal tibial cortical bone. UTE‐MRI signal intensity versus echotime curves were analyzed by fitting a double exponential to determine the short and long relaxation times of water with the bone (dependent estimations of bound and free water, respectively). Raloxifene‐treated animals had significantly higher bound water (+14%; p = 0.05) and lower free water (–20%) compared with vehicle‐treated animals. These data provide the first evidence that drug‐induced changes in skeletal hydration can be noninvasively assessed using UTE‐MRI.


PLOS ONE | 2014

Multimodality Imaging Methods for Assessing Retinoblastoma Orthotopic Xenograft Growth and Development

Timothy W. Corson; Brian C. Samuels; Andrea A. Wenzel; Anna J. Geary; Amanda A. Riley; Brian P. McCarthy; Helmut Hanenberg; Barbara J. Bailey; Pamela I. Rogers; Karen E. Pollok; Gangaraju Rajashekhar; Paul R. Territo

Genomic studies of the pediatric ocular tumor retinoblastoma are paving the way for development of targeted therapies. Robust model systems such as orthotopic xenografts are necessary for testing such therapeutics. One system involves bioluminescence imaging of luciferase-expressing human retinoblastoma cells injected into the vitreous of newborn rat eyes. Although used for several drug studies, the spatial and temporal development of tumors in this model has not been documented. Here, we present a new model to allow analysis of average luciferin flux () through the tumor, a more biologically relevant parameter than peak bioluminescence as traditionally measured. Moreover, we monitored the spatial development of xenografts in the living eye. We engineered Y79 retinoblastoma cells to express a lentivirally-delivered enhanced green fluorescent protein-luciferase fusion protein. In intravitreal xenografts, we assayed bioluminescence and computed , as well as documented tumor growth by intraocular optical coherence tomography (OCT), brightfield, and fluorescence imaging. In vivo bioluminescence, ex vivo tumor size, and ex vivo fluorescent signal were all highly correlated in orthotopic xenografts. By OCT, xenografts were dense and highly vascularized, with well-defined edges. Small tumors preferentially sat atop the optic nerve head; this morphology was confirmed on histological examination. In vivo, in xenografts showed a plateau effect as tumors became bounded by the dimensions of the eye. The combination of modeling and in vivo intraocular imaging allows both quantitative and high-resolution, non-invasive spatial analysis of this retinoblastoma model. This technique will be applied to other cell lines and experimental therapeutic trials in the future.


Journal of Neuroscience Methods | 2011

Assessment of IP injection of [18F]fallypride for behavioral neuroimaging in rats

Karmen K. Yoder; Bruce H. Mock; Qi Huang Zheng; Brian P. McCarthy; Amanda A. Riley; Gary D. Hutchins

Great progress has been made toward using small animal PET to assess neurochemical changes during behavior. [(18)F]fallypride (FAL) is a D(2)/D(3) antagonist that is sensitive to changes in endogenous dopamine, and, in theory, could be used to assess changes in dopamine during behavioral paradigms. Tail vein injections of tracer require restraint in awake animals, and catheter implantation is invasive and can cause logistical problems. Thus, administering tracer with i.p. injections (which are well-tolerated by rodents) would be preferable. The purpose of this study was to determine whether i.p. injection of FAL would produce striatal uptake similar to that seen with traditional i.v. tail vein injection protocols. Four male Sprague-Dawley rats underwent i.p. injection of FAL, followed by a 30-min uptake and subsequent dynamic image acquisition on the IndyPET III small animal scanner. Three of these rats also received traditional dynamic scanning with i.v. FAL injection via a tail vein. Two rats that received i.p. injection had moderate striatal uptake, with striatum/cerebellum ratios (SUVR) that were only ∼20% lower than ratios from i.v. scans. Two other rats had little to no uptake; SUVR values were ∼70% lower than i.v. SUVR. These latter two animals showed heavy bone uptake, evidence of defluorination of FAL. The results of this pilot study suggest that it may be possible to achieve striatal uptake of FAL after i.p. injection. However, this was not seen consistently across animals. Future studies are needed to validate, and then to optimize, the use of i.p. FAL for behavioral imaging protocols.


American Journal of Physiology-renal Physiology | 2016

Novel application of complementary imaging techniques to examine in vivo glucose metabolism in the kidney

Takashi Hato; Allon N. Friedman; Henry E. Mang; Zoya Plotkin; Shataakshi Dube; Gary D. Hutchins; Paul R. Territo; Brian P. McCarthy; Amanda A. Riley; Kumar Pichumani; Craig R. Malloy; Robert A. Harris; Pierre C. Dagher; Timothy A. Sutton

The metabolic status of the kidney is a determinant of injury susceptibility and a measure of progression for many disease processes; however, noninvasive modalities to assess kidney metabolism are lacking. In this study, we employed positron emission tomography (PET) and intravital multiphoton microscopy (MPM) to assess cortical and proximal tubule glucose tracer uptake, respectively, following experimental perturbations of kidney metabolism. Applying dynamic image acquisition PET with 2-18fluoro-2-deoxyglucose (18F-FDG) and tracer kinetic modeling, we found that an intracellular compartment in the cortex of the kidney could be distinguished from the blood and urine compartments in animals. Given emerging literature that the tumor suppressor protein p53 is an important regulator of cellular metabolism, we demonstrated that PET imaging was able to discern a threefold increase in cortical 18F-FDG uptake following the pharmacological inhibition of p53 in animals. Intravital MPM with the fluorescent glucose analog 2-[N-(7-nitrobenz-2-oxa-1,3-diazol-4-yl)amino]-2-deoxyglucose (2-NBDG) provided increased resolution and corroborated these findings at the level of the proximal tubule. Extending our observation of p53 inhibition on proximal tubule glucose tracer uptake, we demonstrated by intravital MPM that pharmacological inhibition of p53 diminishes mitochondrial potential difference. We provide additional evidence that inhibition of p53 alters key metabolic enzymes regulating glycolysis and increases intermediates of glycolysis. In summary, we provide evidence that PET is a valuable tool for examining kidney metabolism in preclinical and clinical studies, intravital MPM is a powerful adjunct to PET in preclinical studies of metabolism, and p53 inhibition alters basal kidney metabolism.


Pancreas | 2015

Longitudinal Bioluminescence Imaging of Primary Versus Abdominal Metastatic Tumor Growth in Orthotopic Pancreatic Tumor Models in NSG Mice.

Harlan E. Shannon; Melissa L. Fishel; Jingwu Xie; Dongsheng Gu; Brian P. McCarthy; Amanda A. Riley; Anthony L. Sinn; Jayne M. Silver; Kacie M. Peterman; Mark R. Kelley; Helmut Hanenberg; Murray Korc; Karen E. Pollok; Paul R. Territo

Objectives The purpose of the present study was to develop and validate noninvasive bioluminescence imaging methods for differentially monitoring primary and abdominal metastatic tumor growth in mouse orthotopic models of pancreatic cancer. Methods A semiautomated maximum entropy segmentation method was implemented for the primary tumor region of interest, and a rule-based method for manually drawing a region of interest for the abdominal metastatic region was developed for monitoring tumor growth in orthotopic models of pancreatic cancer. The 2 region-of-interest methods were validated by having 2 observers independently segment Panc-1 tumors, and the results were compared with the number of mesenteric lymph node nodules and histopathologic assessment of liver metastases. The findings were extended to orthotopic tumors of the more metastatic MIA PaCa-2 and AsPC-1 cells where separate groups of animals were implanted with different numbers of cells. Results The results demonstrated that the segmentation methods were highly reliable, reproducible, and robust and allowed statistically significant discrimination in the growth rates of primary and abdominal metastatic tumors of different cell lines implanted with different numbers of cells. Conclusions The present results demonstrate that primary tumors and abdominal metastatic foci in orthotopic pancreatic cancer models can be reliably quantified separately and noninvasively over time with bioluminescence imaging.


Alzheimers & Dementia | 2018

THE MODEL-AD CONSORTIUM PRECLINICAL TESTING PIPELINE: PHARMACOKINETICS AND PHARMACODYNAMICS OF PROPHYLACTIC TREATMENT WITH LEVETIRACETAM IN THE 5XFAD MOUSE MODEL OF ALZHEIMER’S DISEASE

Stacey J. Sukoff Rizzo; Sara K. Quinney; Kristen D. Onos; Kelly J. Keezer; David R. Jones; Andrea R. Masters; Ingrid F. Metzger; Jill A. Meyer; Jonathan Peters; Scott C. Persohn; Brian P. McCarthy; Amanda A. Riley; Michael Sasner; Gareth R. Howell; Harriett Williams; Adrian L. Oblak; Bruce T. Lamb; Paul R. Territo

restrictive barrier properties controlled by tight junctions and polarized expression of selective transporters, the endothelial cells that form the BBB effectively regulates movement of metabolites and nutrients between blood and brain parenchyma. Any changes in the BBB may impair the clearance of neurotoxic molecules allowing their accumulation and deposition in brain parenchyma and vasculature, leading to neuronal dysfunction and degeneration, and contribute to the onset and progression of Alzheimer’s disease (AD). In AD and CAA, accumulation of amyloid-b (Ab) on microvessels results in a rupture of vessels wall and cerebral hemorrhage, which contribute to, and aggravate, dementia. Accumulation of Ab depends on the imbalance between the production and clearance of Ab. Several pathways for Ab clearance from the brain have been reported including transport across the BBB and enzymatic degradation. Despite our understanding of the pathways responsible for BBB dysfunction and clearance of Ab, the availability of drugs to treat CAA or AD remains lacking. Identifying strategies to rectify BBB integrity and function, and maximize clearance of Ab from the brain is of high clinical importance for the development of interventions, which prevent or delay onset of CAA and AD.Methods:In my lab, we developed a novel BBB model consisting of cerebrovascular endothelial cells and high-throughput screening (HTS) methodologies to screen for hit compounds that ameliorate Ab induced increases in endothelial cell permeability and enhance Ab clearance. Identified hits were then tested in vivo in AD mouse model for BBB tightness, Ab brain levels and Ab related pathology. Results:Multiple hit compounds were identified from the screening that were ranked for their potencies. Most potent compounds were in vivo evaluated in ADmouse model for their therapeutic effect against AD pathology. Our in vitro to in vivo studies have successfully identified candidate therapeutic molecules to test in future clinical studies. Conclusions:Our findings demonstrated the BBB as a therapeutic target to prevent and/or slow the progression of the amyloid pathogenesis disorders CAA and AD.


PLOS ONE | 2017

Effects of combination treatment with alendronate and raloxifene on skeletal properties in a beagle dog model.

Matthew R. Allen; Erin McNerny; Mohammad W. Aref; Jason M. Organ; Christopher L. Newman; Brian McGowan; Tim Jang; David B. Burr; Drew M. Brown; Max A. Hammond; Paul R. Territo; Chen Lin; Scott Persohn; Lei Jiang; Amanda A. Riley; Brian P. McCarthy; Gary D. Hutchins; Joseph M. Wallace

A growing number of studies have investigated combination treatment as an approach to treat bone disease. The goal of this study was to investigate the combination of alendronate and raloxifene with a particular focus on mechanical properties. To achieve this goal we utilized a large animal model, the beagle dog, used previously by our laboratory to study both alendronate and raloxifene monotherapies. Forty-eight skeletally mature female beagles (1–2 years old) received daily oral treatment: saline vehicle (VEH), alendronate (ALN), raloxifene (RAL) or both ALN and RAL. After 6 and 12 months of treatment, all animals underwent assessment of bone material properties using in vivo reference point indentation (RPI) and skeletal hydration using ultra-short echo magnetic resonance imaging (UTE-MRI). End point measures include imaging, histomorphometry, and mechanical properties. Bone formation rate was significantly lower in iliac crest trabecular bone of animals treated with ALN (-71%) and ALN+RAL (-81%) compared to VEH. In vivo assessment of properties by RPI yielded minimal differences between groups while UTE-MRI showed a RAL and RAL+ALN treatment regimens resulted in significantly higher bound water compared to VEH (+23 and +18%, respectively). There was no significant difference among groups for DXA- or CT-based measures lumbar vertebra, or femoral diaphysis. Ribs of RAL-treated animals were smaller and less dense compared to VEH and although mechanical properties were lower the material-level properties were equivalent to normal. In conclusion, we present a suite of data in a beagle dog model treated for one year with clinically-relevant doses of alendronate and raloxifene monotherapies or combination treatment with both agents. Despite the expected effects on bone remodeling, our study did not find the expected benefit of ALN to BMD or structural mechanical properties, and thus the viability of the combination therapy remains unclear.


Cancer Research | 2014

Abstract 4961: Longitudinal bioluminescence imaging of primary versus abdominal metastatic tumor growth in orthotopic pancreatic tumor models in NOD/SCIDγ(-/-) mice

Harlan E. Shannon; Melissa L. Fishel; Jingwu Xie; Dongsheng Gu; Brian P. McCarthy; Amanda A. Riley; Anthony L. Sinn; Jayne M. Silver; Mark R. Kelley; Helmut Hanenberg; Murray Korc; Karen E. Pollok; Paul R. Territo

Pancreatic ductal adenocarcinoma (PDAC) has a very poor prognosis and is currently the fourth leading cause of cancer death in the United States. The lethal nature of PDAC is strongly associated with metastases to distant organs. The purpose of the present study was to develop and validate noninvasive bioluminescence imaging methods for differentially monitoring the kinetics of primary and abdominal metastatic tumor growth in mouse orthotopic models of pancreatic cancer. A semiautomated maximum entropy segmentation method was implemented for the primary tumor region-of-interest (ROI), and a rule-based method for manually drawing an ROI for the abdominal metastatic region also was developed. The two ROI methods were first validated by having two observers independently construct ROIs for the tumors of animals implanted orthotopically with Panc-1 cells, and the results compared with the number of mesenteric lymph node metastatic nodules counted upon necropsy. The findings were extended to orthotopic tumors of the more metastatic MIA PaCa-2 and AsPC-1 cells where different groups of animals were implanted with different numbers of cells. When the data were expressed as the total photon flux (Ph/sec) in the ROIs for the primary tumor and metastases, the total flux within the metastasis ROI was larger in magnitude than the total flux from the primary tumor ROI, at times by as much as several orders of magnitude. However, when the data were expressed as the average flux density (Ph/sec*mm2) within the ROIs, the density of the flux within the smaller primary tumor ROI was larger in magnitude than the density of the flux from the larger metastasis ROI, by as much as several orders of magnitude. Interobserver assessments for total flux and flux density from ROIs for both the primary tumors and metastatic region were highly concordant, with correlation coefficients of r > 0.98, coefficients of variation of ≤ 0.02, and limits of agreement within Citation Format: Harlan E. Shannon, Melissa L. Fishel, Jingwu Xie, Dongsheng Gu, Brian P. McCarthy, Amanda A. Riley, Anthony L. Sinn, Jayne M. Silver, Mark R. Kelley, Helmut Hanenberg, Murray Korc, Karen E. Pollok, Paul R. Territo. Longitudinal bioluminescence imaging of primary versus abdominal metastatic tumor growth in orthotopic pancreatic tumor models in NOD/SCIDγ(-/-) mice. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 4961. doi:10.1158/1538-7445.AM2014-4961

Collaboration


Dive into the Amanda A. Riley's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge