Network


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

Hotspot


Dive into the research topics where Steven Pennock is active.

Publication


Featured researches published by Steven Pennock.


American Journal of Pathology | 2011

A Novel Strategy to Develop Therapeutic Approaches to Prevent Proliferative Vitreoretinopathy

Steven Pennock; Marc-Andre Rheaume; Shizuo Mukai; Andrius Kazlauskas

Proliferative vitreoretinopathy (PVR) thwarts the repair of rhegmatogenous retinal detachments. Currently, there is no effective prevention for PVR. Platelet-derived growth factor receptor α (PDGFRα) is associated with PVR in humans and strongly promotes experimental PVR driven by multiple vitreal growth factors outside the PDGF family. We sought to identify vitreal factors required for experimental PVR and to establish a potential approach to prevent PVR. Vitreous was obtained from normal rabbits or those in which PVR was either developing or stabilized. Normal vitreous contained substantial levels of growth factors and cytokines, which changed quantitatively and/or qualitatively as PVR progressed and stabilized. Neutralizing a subset of these agents in rabbit vitreous eliminated their ability to induce PVR-relevant signaling and cellular responses. A single intravitreal injection of neutralizing reagents for this subset prevented experimental PVR. To identify growth factors and cytokines likely driving PVR in humans, we subjected vitreous from patients with or without PVR to a similar series of analyses. This analysis accurately identified those agents required for vitreous-induced contraction of cells from a patient PVR membrane. We conclude that combination therapy encompassing a subset of vitreal growth factors and cytokines is a potential approach to prevent PVR.


Molecular and Cellular Biology | 2012

Vascular Endothelial Growth Factor A Competitively Inhibits Platelet-Derived Growth Factor (PDGF)-Dependent Activation of PDGF Receptor and Subsequent Signaling Events and Cellular Responses

Steven Pennock; Andrius Kazlauskas

ABSTRACT Certain platelet-derived growth factor (PDGF) isoforms are associated with proliferative vitreoretinopathy (PVR), a sight-threatening complication that develops in a subset of patients recovering from retinal reattachment surgery. Although these PDGF isoforms are abundant in the vitreous of patients and experimental animals with PVR, they make only a minor contribution to activating PDGF receptor α (PDGFRα) and driving experimental PVR. Rather, growth factors outside of the PDGF family are the primary (and indirect) agonists of PDGFRα. These observations beg the question of why vitreal PDGFs fail to activate PDGFRα. We report here that vitreous contains an inhibitor of PDGF-dependent activation of PDGFRα and that a major portion of this inhibitory activity is due to vascular endothelial cell growth factor A (VEGF-A). Furthermore, recombinant VEGF-A competitively blocks PDGF-dependent binding and activation of PDGFR, signaling events, and cellular responses. These findings unveil a previously unappreciated relationship between distant members of the PDGF/VEGF family that may contribute to pathogenesis of a blinding eye disease.


American Journal of Pathology | 2013

Ranibizumab Is a Potential Prophylaxis for Proliferative Vitreoretinopathy, a Nonangiogenic Blinding Disease

Steven Pennock; David Y. Kim; Shizuo Mukai; Matthew Kuhnle; Dal Chun; Joanne A. Matsubara; Jing Cui; Patrick Ma; David Maberley; Arif Samad; Rob J. Van Geest; Sarit Y. Lesnik Oberstein; Reinier O. Schlingemann; Andrius Kazlauskas

Proliferative vitreoretinopathy (PVR) exemplifies a disease that is difficult to predict, lacks effective treatment options, and substantially reduces the quality of life of an individual. Surgery to correct a rhegmatogenous retinal detachment fails primarily because of PVR. Likely mediators of PVR are growth factors in vitreous, which stimulate cells within and behind the retina as an inevitable consequence of a breached retina. Three classes of growth factors [vascular endothelial growth factor A (VEGF-A), platelet-derived growth factors (PDGFs), and non-PDGFs (growth factors outside of the PDGF family)] are relevant to PVR pathogenesis because they act on PDGF receptor α, which is required for experimental PVR and is associated with this disease in humans. We discovered that ranibizumab (a clinically approved agent that neutralizes VEGF-A) reduced the bioactivity of vitreous from patients and experimental animals with PVR, and protected rabbits from developing disease. The apparent mechanism of ranibizumab action involved derepressing PDGFs, which, at the concentrations present in PVR vitreous, inhibited non-PDGF-mediated activation of PDGF receptor α. These preclinical findings suggest that available approaches to neutralize VEGF-A are prophylactic for PVR, and that anti-VEGF-based therapies may be effective for managing more than angiogenesis- and edema-driven pathological conditions.


American Journal of Pathology | 2014

Vascular Endothelial Growth Factor Acts Primarily via Platelet-Derived Growth Factor Receptor α to Promote Proliferative Vitreoretinopathy

Steven Pennock; Luis J. Haddock; Shizuo Mukai; Andrius Kazlauskas

Proliferative vitreoretinopathy (PVR) is a nonneovascular blinding disease and the leading cause for failure in surgical repair of rhegmatogenous retinal detachments. Once formed, PVR is difficult to treat. Hence, there is an acute interest in developing approaches to prevent PVR. Of the many growth factors and cytokines that accumulate in vitreous as PVR develops, neutralizing vascular endothelial growth factor (VEGF) A has recently been found to prevent PVR in at least one animal model. The goal of this study was to test if Food and Drug Administration-approved agents could protect the eye from PVR in multiple animal models and to further investigate the underlying mechanisms. Neutralizing VEGF with aflibercept (VEGF Trap-Eye) safely and effectively protected rabbits from PVR in multiple models of disease. Furthermore, aflibercept reduced the bioactivity of both experimental and clinical PVR vitreous. Finally, although VEGF could promote some PVR-associated cellular responses via VEGF receptors expressed on the retinal pigment epithelial cells that drive this disease, VEGFs major contribution to vitreal bioactivity occurred via platelet-derived growth factor receptor α. Thus, VEGF promotes PVR by a noncanonical ability to engage platelet-derived growth factor receptor α. These findings indicate that VEGF contributes to nonangiogenic diseases and that anti-VEGF-based therapies may be effective on a wider spectrum of diseases than previously appreciated.


Molecular and Cellular Biology | 2016

Vascular Endothelial Cell Growth Factor A Acts via Platelet-Derived Growth Factor Receptor α To Promote Viability of Cells Enduring Hypoxia

Steven Pennock; Leo A. Kim; Andrius Kazlauskas

ABSTRACT Vascular endothelial cell growth factor A (VEGF) is a biologically and therapeutically important growth factor because it promotes angiogenesis in response to hypoxia, which underlies a wide variety of both physiological and pathological settings. We report here that both VEGF receptor 2 (VEGFR2)-positive and -negative cells depended on VEGF to endure hypoxia. VEGF enhanced the viability of platelet-derived growth factor receptor α (PDGFRα)-positive and VEGFR2-negative cells by enabling indirect activation of PDGFRα, thereby reducing the level of p53. We conclude that the breadth of VEGFs influence extends beyond VEGFR-positive cells and propose a plausible mechanistic explanation of this phenomenon.


Progress in Retinal and Eye Research | 2014

Is neutralizing vitreal growth factors a viable strategy to prevent proliferative vitreoretinopathy

Steven Pennock; Luis J. Haddock; Dean Eliott; Shizuo Mukai; Andrius Kazlauskas


Investigative Ophthalmology & Visual Science | 2013

Ranibizumab is a potential prophylaxis for proliferative vitreoretinopathy, a non-angiogenic blinding disease

Steven Pennock; David M. Kim; Shizuo Mukai; Matthew Kuhnle; Dal Chun; Joanne A. Matsubara; Jing Cui; Patrick Ma; David Maberley; Andrius Kazlauskas


Investigative Ophthalmology & Visual Science | 2013

Epithelial cells promote fibroblast-mediated contraction of collagen gels by secreting bFGF

Maryada Sharma; Hetian Lei; Steven Pennock; Andrius Kazlauskas


Archive | 2012

and subsequent signaling events and cellular responses

Steven Pennock; Andrius Kazlauskas


Investigative Ophthalmology & Visual Science | 2011

A Novel Strategy To Develop Therapeutic Approaches To Prevent Proliferative Vitreoretinopathy

Steven Pennock; Marc-Andre Rheaume; Shizuo Mukai; Joanne A. Matsubara; Jing Cui; David Maberley; Patrick Ma; Arif Samad; R. J. van Geest; Andrius Kazlauskas

Collaboration


Dive into the Steven Pennock's collaboration.

Top Co-Authors

Avatar

Andrius Kazlauskas

Massachusetts Eye and Ear Infirmary

View shared research outputs
Top Co-Authors

Avatar

Shizuo Mukai

Massachusetts Eye and Ear Infirmary

View shared research outputs
Top Co-Authors

Avatar

Dal Chun

Walter Reed Army Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jing Cui

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Matthew Kuhnle

Walter Reed Army Medical Center

View shared research outputs
Top Co-Authors

Avatar

David Maberley

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Joanne A. Matsubara

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Patrick Ma

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Luis J. Haddock

Massachusetts Eye and Ear Infirmary

View shared research outputs
Top Co-Authors

Avatar

Marc-Andre Rheaume

Massachusetts Eye and Ear Infirmary

View shared research outputs
Researchain Logo
Decentralizing Knowledge