Network


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

Hotspot


Dive into the research topics where Anthony R. Prisco is active.

Publication


Featured researches published by Anthony R. Prisco.


Physiological Genomics | 2013

Vascular endothelial growth factor-A signaling in bone marrow-derived endothelial progenitor cells exposed to hypoxic stress.

Brian R. Hoffmann; Jordan R. Wagner; Anthony R. Prisco; Agnieszka Janiak; Andrew S. Greene

Bone marrow-derived endothelial progenitor cells (BM-EPCs) are stimulated by vascular endothelial growth factor-A (VEGF-A) and other potent proangiogenic factors. During angiogenesis, an increase in VEGF-A expression stimulates BM-EPCs to enhance endothelial tube formation and contribute to an increase in microvessel density. Hypoxia is known to produce an enhanced angiogenic response and heightened levels of VEGF-A have been seen in oxygen deprived epithelial and endothelial cells, yet the pathways for VEGF-A signaling in BM-EPCs have not been described. This study explores the influence of hypoxia on VEGF-A signaling in rat BM-EPCs utilizing a novel proteomic strategy to directly identify interacting downstream components of the combined VEGF receptor(s) signaling pathways, gene expression analysis, and functional phenotyping. VEGF-A signaling network analysis following liquid chromatographic separation and tandem mass spectrometry revealed proteins related to inositol/calcium signaling, nitric oxide signaling, cell survival, cell migration, and inflammatory responses. Alterations in BM-EPC expression of common angiogenic genes and tube formation in response to VEGF-A during hypoxia were measured and combined with the proteomic analysis to enhance and support the signaling pathways detected. BM-EPC tube formation assays in response to VEGF-A exhibited little tube formation; however, a cell projection/migratory phenotype supported the signaling data. Additionally, a novel assay measuring BM-EPC incorporation into preformed endothelial cell tubes indicated a significant increase of incorporated BM-EPCs after pretreatment with VEGF-A during hypoxia. This study verifies known VEGF-A pathway components and reveals several unidentified mechanisms of VEGF-A signaling in BM-EPCs during hypoxia that may be important for migration to sites of vascular regeneration.


American Journal of Physiology-heart and Circulatory Physiology | 2015

TNF-α increases endothelial progenitor cell adhesion to the endothelium by increasing bond expression and affinity

Anthony R. Prisco; Michael R Prisco; Brian E. Carlson; Andrew S. Greene

Endothelial progenitor cells (EPCs) are a rare population of cells that participate in angiogenesis. To effectively use EPCs for regenerative therapy, the mechanisms by which they participate in tissue repair must be elucidated. This study focused on the process by which activated EPCs bind to a target tissue. It has been demonstrated that EPCs can bind to endothelial cells (ECs) through the tumore necrosis factor-α (TNF-α)-regulated vascular cell adhesion molecule 1/very-late antigen 4 (VLA4) interaction. VLA4 can bind in a high or low affinity state, a process that is difficult to experimentally isolate from bond expression upregulation. To separate these processes, a new parallel plate flow chamber was built, a detachment assay was developed, and a mathematical model was created that was designed to analyze the detachment assay results. The mathematical model was developed to predict the relative expression of EPC/EC bonds made for a given bond affinity distribution. EPCs treated with TNF-α/vehicle were allowed to bind to TNF-α/vehicle-treated ECs in vitro. Bound cells were subjected to laminar flow, and the cellular adherence was quantified as a function of shear stress. Experimental data were fit to the mathematical model using changes in bond expression or affinity as the only free parameter. It was found that TNF-α treatment of ECs increased adhesion through bond upregulation, whereas TNF-α treatment of EPCs increased adhesion by increasing bond affinity. These data suggest that injured tissue could potentially increase recruitment of EPCs for tissue regeneration via the secretion of TNF-α.


Stem Cells | 2016

Tumor Necrosis Factor α Regulates Endothelial Progenitor Cell Migration via CADM1 and NF‐kB

Anthony R. Prisco; Brian R. Hoffmann; Catherine C. Kaczorowski; Chris McDermott-Roe; Timothy J. Stodola; Eric C. Exner; Andrew S. Greene

Shortly after the discovery of endothelial progenitor cells (EPCs) in 1997, many clinical trials were conducted using EPCs as a cellular based therapy with the goal of restoring damaged organ function by inducing growth of new blood vessels (angiogenesis). Results were disappointing, largely because the cellular and molecular mechanisms of EPC‐induced angiogenesis were not clearly understood. Following injection, EPCs must migrate to the target tissue and engraft prior to induction of angiogenesis. In this study EPC migration was investigated in response to tumor necrosis factor α (TNFα), a pro‐inflammatory cytokine, to test the hypothesis that organ damage observed in ischemic diseases induces an inflammatory signal that is important for EPC homing. In this study, EPC migration and incorporation were modeled in vitro using a coculture assay where TNFα treated EPCs were tracked while migrating toward vessel‐like structures. It was found that TNFα treatment of EPCs increased migration and incorporation into vessel‐like structures. Using a combination of genomic and proteomic approaches, NF‐kB mediated upregulation of CADM1 was identified as a mechanism of TNFα induced migration. Inhibition of NF‐kB or CADM1 significantly decreased migration of EPCs in vitro suggesting a role for TNFα signaling in EPC homing during tissue repair. Stem Cells 2016;34:1922–1933


American Journal of Physiology-heart and Circulatory Physiology | 2014

Vascular dysfunction precedes hypertension associated with a blood pressure locus on rat chromosome 12

Sasha Z. Prisco; Jessica R. C. Priestley; Brian D. Weinberg; Anthony R. Prisco; Matthew J. Hoffman; Howard J. Jacob; Michael J. Flister; Julian H. Lombard; Jozef Lazar

We previously isolated a 6.1-Mb region of SS/Mcwi (Dahl salt-sensitive) rat chromosome 12 (13.4-19.5 Mb) that significantly elevated blood pressure (BP) (Δ+34 mmHg, P < 0.001) compared with the SS-12(BN) consomic control. In the present study, we examined the role of vascular dysfunction and remodeling in hypertension risk associated with the 6.1-Mb (13.4-19.5 Mb) locus on rat chromosome 12 by reducing dietary salt, which lowered BP levels so that there were no substantial differences in BP between strains. Consequently, any observed differences in the vasculature were considered BP-independent. We also reduced the candidate region from 6.1 Mb with 133 genes to 2 Mb with 23 genes by congenic mapping. Both the 2 Mb and 6.1 Mb congenic intervals were associated with hypercontractility and decreased elasticity of resistance vasculature prior to elevations of BP, suggesting that the vascular remodeling and dysfunction likely contribute to the pathogenesis of hypertension in these congenic models. Of the 23 genes within the narrowed congenic interval, 12 were differentially expressed between the resistance vasculature of the 2 Mb congenic and SS-12(BN) consomic strains. Among these, Grifin was consistently upregulated 2.7 ± 0.6-fold (P < 0.05) and 2.0 ± 0.3-fold (P < 0.01), and Chst12 was consistently downregulated -2.8 ± 0.3-fold (P < 0.01) and -4.4 ± 0.4-fold (P < 0.00001) in the 2 Mb congenic compared with SS-12(BN) consomic under normotensive and hypertensive conditions, respectively. A syntenic region on human chromosome 7 has also been associated with BP regulation, suggesting that identification of the genetic mechanism(s) underlying cardiovascular phenotypes in this congenic strain will likely be translated to a better understanding of human hypertension.


Stem Cells | 2016

TNFα Regulates Endothelial Progenitor Cell Migration via CADM1 and NF‐kB

Anthony R. Prisco; Brian R. Hoffmann; Catherine C. Kaczorowski; Chris McDermott-Roe; Timothy J. Stodola; Eric C. Exner; Andrew S. Greene

Shortly after the discovery of endothelial progenitor cells (EPCs) in 1997, many clinical trials were conducted using EPCs as a cellular based therapy with the goal of restoring damaged organ function by inducing growth of new blood vessels (angiogenesis). Results were disappointing, largely because the cellular and molecular mechanisms of EPC‐induced angiogenesis were not clearly understood. Following injection, EPCs must migrate to the target tissue and engraft prior to induction of angiogenesis. In this study EPC migration was investigated in response to tumor necrosis factor α (TNFα), a pro‐inflammatory cytokine, to test the hypothesis that organ damage observed in ischemic diseases induces an inflammatory signal that is important for EPC homing. In this study, EPC migration and incorporation were modeled in vitro using a coculture assay where TNFα treated EPCs were tracked while migrating toward vessel‐like structures. It was found that TNFα treatment of EPCs increased migration and incorporation into vessel‐like structures. Using a combination of genomic and proteomic approaches, NF‐kB mediated upregulation of CADM1 was identified as a mechanism of TNFα induced migration. Inhibition of NF‐kB or CADM1 significantly decreased migration of EPCs in vitro suggesting a role for TNFα signaling in EPC homing during tissue repair. Stem Cells 2016;34:1922–1933


Stem Cells | 2016

Tumor Necrosis Factor α Regulates Endothelial Progenitor Cell Migration via CADM1 and NF-kB: TNFα Induced EPC Migration

Anthony R. Prisco; Brian R. Hoffmann; Catherine C. Kaczorowski; Chris McDermott-Roe; Timothy J. Stodola; Eric C. Exner; Andrew S. Greene

Shortly after the discovery of endothelial progenitor cells (EPCs) in 1997, many clinical trials were conducted using EPCs as a cellular based therapy with the goal of restoring damaged organ function by inducing growth of new blood vessels (angiogenesis). Results were disappointing, largely because the cellular and molecular mechanisms of EPC‐induced angiogenesis were not clearly understood. Following injection, EPCs must migrate to the target tissue and engraft prior to induction of angiogenesis. In this study EPC migration was investigated in response to tumor necrosis factor α (TNFα), a pro‐inflammatory cytokine, to test the hypothesis that organ damage observed in ischemic diseases induces an inflammatory signal that is important for EPC homing. In this study, EPC migration and incorporation were modeled in vitro using a coculture assay where TNFα treated EPCs were tracked while migrating toward vessel‐like structures. It was found that TNFα treatment of EPCs increased migration and incorporation into vessel‐like structures. Using a combination of genomic and proteomic approaches, NF‐kB mediated upregulation of CADM1 was identified as a mechanism of TNFα induced migration. Inhibition of NF‐kB or CADM1 significantly decreased migration of EPCs in vitro suggesting a role for TNFα signaling in EPC homing during tissue repair. Stem Cells 2016;34:1922–1933


Physiological Genomics | 2013

Targeting the endothelial progenitor cell surface proteome to identify novel mechanisms that mediate angiogenic efficacy in a rodent model of vascular disease

Catherine C. Kaczorowski; Timothy J. Stodola; Brian R. Hoffmann; Anthony R. Prisco; Pengyuan Liu; Daniela N. Didier; Jamie R. Karcher; Mingyu Liang; Howard J. Jacob; Andrew S. Greene


Breast Cancer Research and Treatment | 2017

Host genetic modifiers of nonproductive angiogenesis inhibit breast cancer

Michael J. Flister; Shirng-Wern Tsaih; Alexander Stoddard; Cody Plasterer; Jaidip Jagtap; Abdul K. Parchur; Gayatri Sharma; Anthony R. Prisco; Angela Lemke; Dana Murphy; Mona M. Al-Gizawiy; Michael W. Straza; Sophia Ran; Aron M. Geurts; Melinda R. Dwinell; Andrew S. Greene; Carmen Bergom; Peter S. LaViolette; Amit Joshi


心臓 | 2015

Broken Bones/Broken Heart : The Cardiovascular-Bone Marrow Axis

Andrew S. Greene; Sarah Parker; Anthony R. Prisco


The FASEB Journal | 2015

Hyperglycemia-induced alterations of the vascular endothelium in type 2 diabetes mellitus

Brian R. Hoffmann; Anthony R. Prisco; Michael E. Widlansky; Andrew S. Greene

Collaboration


Dive into the Anthony R. Prisco's collaboration.

Top Co-Authors

Avatar

Andrew S. Greene

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Brian R. Hoffmann

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Catherine C. Kaczorowski

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar

Howard J. Jacob

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Timothy J. Stodola

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Jamie R. Karcher

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Daniela N. Didier

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Michael J. Flister

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Mingyu Liang

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Chris McDermott-Roe

Medical College of Wisconsin

View shared research outputs
Researchain Logo
Decentralizing Knowledge