Network


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

Hotspot


Dive into the research topics where Brian P. McCarthy is active.

Publication


Featured researches published by Brian P. McCarthy.


Journal of Immunology | 2005

Enhanced Production of IL-10 by Dendritic Cells Deficient in CIITA

Christina S.K. Yee; Yongxue Yao; Qi Xu; Brian P. McCarthy; Deqin Sun-Lin; Masahide Tone; Herman Waldmann; Cheong Hee Chang

Dendritic cells (DC) are professional APCs that play a critical role in regulating immunity. In DC, maturation-induced changes in MHC class II expression and Ag presentation require transcriptional regulation by CIITA. To study the role of CIITA in DC, we evaluated key cell functions in DC from CIITA-deficient (CIITA−/−) mice. The ability to take up Ag, measured by fluid phase endocytosis, was comparable between CIITA−/− and control DC. Although CIITA−/− DC lack MHC class II, they maintained normal expression of costimulatory molecules CD80, CD86, and CD40. In contrast, CIITA−/− DC activated with LPS or CpG expressed increased IL-10 levels, but normal levels of TNF-α and IL-12 relative to control. Enhanced IL-10 was due to greater IL-10 mRNA in CIITA−/− DC. Aβ−/− DC, which lack MHC class II but express CIITA normally, had exhibited no difference in IL-10 compared with control. When CIITA was cotransfected with an IL-10 promoter-reporter into a mouse monocyte cell line, RAW 264.7, IL-10 promoter activity was decreased. In addition, reintroducing CIITA into CIITA−/− DC reduced production of IL-10. In all, these data suggest that CIITA negatively regulates expression of IL-10, and that CIITA may direct DC function in ways that extend beyond control of MHC class II.


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 Leukocyte Biology | 2007

IL-4-stimulated NF-κB activity is required for Stat6 DNA binding

Vivian T. Thieu; Evelyn T. Nguyen; Brian P. McCarthy; Heather A. Bruns; Reuben Kapur; Cheong Hee Chang; Mark H. Kaplan

IL‐4 is a critical cytokine in the regulation of immune responses. In B lymphocytes, IL‐4 signaling promotes the Stat6‐dependent cell surface expression of several proteins including MHC Class II and CD86. However, the requirement for other transcription factors in IL‐4‐induced B cell gene expression has not been studied extensively. Here, we show that IL‐4 induces NF‐κB p100 processing to NF‐κB p52 in B cells but not in T cells or macrophages. IL‐4 induced NF‐κB p52 production requires PI‐3K activity and correlates with IκB kinase phosphorylation and TNF receptor‐associated factor 3 degradation. Blocking NF‐κB activity eliminates IL‐4‐stimulated gene expression in B cells by reducing IL‐4‐induced DNA binding but not phosphorylation or nuclear localization of Stat6. These results describe a novel role for NF‐κB in IL‐4‐induced signaling and gene expression.


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.


JCI insight | 2017

SOCS1 is a negative regulator of metabolic reprogramming during sepsis

Annie Rocio Piñeros Alvarez; Nicole L. Glosson-Byers; Stephanie L. Brandt; Soujuan Wang; Hector R. Wong; Sarah Sturgeon; Brian P. McCarthy; Paul R. Territo; José C. Alves-Filho; C. Henrique Serezani

Sepsis can induce an overwhelming systemic inflammatory response, resulting in organ damage and death. Suppressor of cytokine signaling 1 (SOCS1) negatively regulates signaling by cytokine receptors and Toll-like receptors (TLRs). However, the cellular targets and molecular mechanisms for SOCS1 activity during polymicrobial sepsis are unknown. To address this, we utilized a cecal ligation and puncture (CLP) model for sepsis; C57BL/6 mice subjected to CLP were then treated with a peptide (iKIR) that binds the SOCS1 kinase inhibitory region (KIR) and blocks its activity. Treatment with iKIR increased CLP-induced mortality, bacterial burden, and inflammatory cytokine production. Myeloid cell-specific SOCS1 deletion (Socs1Δmyel) mice were also more susceptible to sepsis, demonstrating increased mortality, higher bacterial loads, and elevated inflammatory cytokines, compared with Socs1fl littermate controls. These effects were accompanied by macrophage metabolic reprograming, as evidenced by increased lactic acid production and elevated expression of the glycolytic enzymes hexokinase, lactate dehydrogenase A, and glucose transporter 1 in septic Socs1Δmyel mice. Upregulation was dependent on the STAT3/HIF-1α/glycolysis axis, and blocking glycolysis ameliorated increased susceptibility to sepsis in iKIR-treated CLP mice. These results reveal a role of SOCS1 as a regulator of metabolic reprograming that prevents overwhelming inflammatory response and organ damage during sepsis.


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.


Cancer Research | 2017

EZH2 Modifies Sunitinib Resistance in Renal Cell Carcinoma by Kinome Reprogramming

Remi Adelaiye-Ogala; Justin Budka; Nur P. Damayanti; Justine V. Arrington; Mary W. Ferris; Chuan-Chih Hsu; Sreenivasulu Chintala; Ashley Orillion; Kiersten Marie Miles; Li Shen; May Elbanna; Eric Ciamporcero; Sreevani Arisa; Piergiorgio Pettazzoni; Giulio Draetta; Mukund Seshadri; Bradley A. Hancock; Milan Radovich; Janaiah Kota; Michael J. Buck; Heike Keilhack; Brian P. McCarthy; Scott Persohn; Paul R. Territo; Yong Zang; Joseph Irudayaraj; Andy Tao; Peter C. Hollenhorst; Roberto Pili

Acquired and intrinsic resistance to receptor tyrosine kinase inhibitors (RTKi) represents a major hurdle in improving the management of clear cell renal cell carcinoma (ccRCC). Recent reports suggest that drug resistance is driven by tumor adaptation via epigenetic mechanisms that activate alternative survival pathways. The histone methyl transferase EZH2 is frequently altered in many cancers, including ccRCC. To evaluate its role in ccRCC resistance to RTKi, we established and characterized a spontaneously metastatic, patient-derived xenograft model that is intrinsically resistant to the RTKi sunitinib, but not to the VEGF therapeutic antibody bevacizumab. Sunitinib maintained its antiangiogenic and antimetastatic activity but lost its direct antitumor effects due to kinome reprogramming, which resulted in suppression of proapoptotic and cell-cycle-regulatory target genes. Modulating EZH2 expression or activity suppressed phosphorylation of certain RTKs, restoring the antitumor effects of sunitinib in models of acquired or intrinsically resistant ccRCC. Overall, our results highlight EZH2 as a rational target for therapeutic intervention in sunitinib-resistant ccRCC as well as a predictive marker for RTKi response in this disease. Cancer Res; 77(23); 6651-66. ©2017 AACR.

Collaboration


Dive into the Brian P. McCarthy'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