Network


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

Hotspot


Dive into the research topics where Peter D Westenskow is active.

Publication


Featured researches published by Peter D Westenskow.


Journal of Clinical Investigation | 2012

Targeted deletion of Vegfa in adult mice induces vision loss

Toshihide Kurihara; Peter D Westenskow; Stephen Bravo; Edith Aguilar; Martin Friedlander

Current therapies directed at controlling vascular abnormalities in cancers and neovascular eye diseases target VEGF and can slow the progression of these diseases. While the critical role of VEGF in development has been well described, the function of locally synthesized VEGF in the adult eye is incompletely understood. Here, we show that conditionally knocking out Vegfa in adult mouse retinal pigmented epithelial (RPE) cells, which regulate retinal homeostasis, rapidly leads to vision loss and ablation of the choriocapillaris, the major blood supply for the outer retina and photoreceptor cells. This deletion also caused rapid dysfunction of cone photoreceptors, the cells responsible for fine visual acuity and color vision. Furthermore, Vegfa deletion showed significant downregulation of multiple angiogenic genes in both physiological and pathological states, whereas the deletion of the upstream regulatory transcriptional factors HIFs did not affect the physiological expressions of angiogenic genes. These results suggest that endogenous VEGF provides critical trophic support necessary for retinal function. Targeting factors upstream of VEGF, such as HIFs, may be therapeutically advantageous compared with more potent and selective VEGF antagonists, which may have more off-target inhibitory trophic effects.


Analytical Chemistry | 2014

Interactive XCMS Online: Simplifying Advanced Metabolomic Data Processing and Subsequent Statistical Analyses

Harsha Gowda; Julijana Ivanisevic; Caroline H. Johnson; Michael E. Kurczy; H. Paul Benton; Duane Rinehart; Thomas Nguyen; Jayashree Ray; Jennifer V. Kuehl; Bernardo Arevalo; Peter D Westenskow; Junhua Wang; Adam P. Arkin; Adam M. Deutschbauer; Gary J. Patti; Gary Siuzdak

XCMS Online (xcmsonline.scripps.edu) is a cloud-based informatic platform designed to process and visualize mass-spectrometry-based, untargeted metabolomic data. Initially, the platform was developed for two-group comparisons to match the independent, “control” versus “disease” experimental design. Here, we introduce an enhanced XCMS Online interface that enables users to perform dependent (paired) two-group comparisons, meta-analysis, and multigroup comparisons, with comprehensive statistical output and interactive visualization tools. Newly incorporated statistical tests cover a wide array of univariate analyses. Multigroup comparison allows for the identification of differentially expressed metabolite features across multiple classes of data while higher order meta-analysis facilitates the identification of shared metabolic patterns across multiple two-group comparisons. Given the complexity of these data sets, we have developed an interactive platform where users can monitor the statistical output of univariate (cloud plots) and multivariate (PCA plots) data analysis in real time by adjusting the threshold and range of various parameters. On the interactive cloud plot, metabolite features can be filtered out by their significance level (p-value), fold change, mass-to-charge ratio, retention time, and intensity. The variation pattern of each feature can be visualized on both extracted-ion chromatograms and box plots. The interactive principal component analysis includes scores, loadings, and scree plots that can be adjusted depending on scaling criteria. The utility of XCMS functionalities is demonstrated through the metabolomic analysis of bacterial stress response and the comparison of lymphoblastic leukemia cell lines.


Stem Cells Translational Medicine | 2012

Generation of Retinal Pigment Epithelial Cells from Small Molecules and OCT4 Reprogrammed Human Induced Pluripotent Stem Cells

Tim U. Krohne; Peter D Westenskow; Toshihide Kurihara; David F. Friedlander; Mandy Lehmann; Alison L. Dorsey; Wenlin Li; Saiyong Zhu; Andrew Schultz; Junhua Wang; Gary Siuzdak; Sheng Ding; Martin Friedlander

Autologous retinal pigment epithelium (RPE) grafts derived from induced pluripotent stem cells (iPSCs) may be used to cure blinding diseases in which RPE dysfunction results in photoreceptor degeneration. Four‐, two‐, and one‐factor‐derived iPSCs (4F‐, 2F‐, and 1F‐iPSCs, respectively) were differentiated into fully functional cuboidal pigmented cells in polarized monolayers that express RPE‐specific markers. 1F‐iPSC‐RPE (1F‐iPS‐RPE) strongly resembles primary human fetal RPE (hfRPE) based on proteomic and untargeted metabolomic analyses, and using novel in vivo imaging technology coupled with electroretinography, we demonstrated that 1F‐iPS‐RPE mediate anatomical and functional rescue of photoreceptors after transplantation in an animal model of RPE‐mediated retinal degeneration. 1F‐iPS‐RPE cells were injected subretinally as a suspension and formed a monolayer dispersed between host RPE cells. Furthermore, 1F‐iPS‐RPE do not simply provide trophic support to rescue photoreceptors as previously speculated but actually phagocytose photoreceptor outer segments in vivo and maintain visual cycling. Thus, 1F‐iPS‐RPE grafts may be superior to conventional iPS‐RPE for clinical use because 1F‐iPS‐RPE closely resemble hfRPE, mediate anatomical and functional photoreceptor rescue in vivo, and are generated using a reduced number of potentially oncogenic reprogramming factors.


Journal of Clinical Investigation | 2015

Neurovascular crosstalk between interneurons and capillaries is required for vision

Yoshihiko Usui; Peter D Westenskow; Toshihide Kurihara; Edith Aguilar; Susumu Sakimoto; Liliana P Paris; Carli M Wittgrove; Daniel Feitelberg; M. Friedlander; Stacey K. Moreno; Michael I. Dorrell; Martin Friedlander

Functional interactions between neurons, vasculature, and glia within neurovascular units are critical for maintenance of the retina and other CNS tissues. For example, the architecture of the neurosensory retina is a highly organized structure with alternating layers of neurons and blood vessels that match the metabolic demand of neuronal activity with an appropriate supply of oxygen within perfused blood. Here, using murine genetic models and cell ablation strategies, we have demonstrated that a subset of retinal interneurons, the amacrine and horizontal cells, form neurovascular units with capillaries in 2 of the 3 retinal vascular plexuses. Moreover, we determined that these cells are required for generating and maintaining the intraretinal vasculature through precise regulation of hypoxia-inducible and proangiogenic factors, and that amacrine and horizontal cell dysfunction induces alterations to the intraretinal vasculature and substantial visual deficits. These findings demonstrate that specific retinal interneurons and the intraretinal vasculature are highly interdependent, and loss of either or both elicits profound effects on photoreceptor survival and function.


Journal of Clinical Investigation | 2013

Ras pathway inhibition prevents neovascularization by repressing endothelial cell sprouting

Peter D Westenskow; Toshihide Kurihara; Edith Aguilar; Elizabeth L. Scheppke; Stacey K. Moreno; Carli M Wittgrove; Valentina Marchetti; Iacovos P. Michael; Sudarshan Anand; Andras Nagy; David A. Cheresh; Martin Friedlander

Vascular networks develop from a growing vascular front that responds to VEGF and other guidance cues. Angiogenesis is required for normal tissue function, but, under conditions of stress, inappropriate vascularization can lead to disease. Therefore, inhibition of angiogenic sprouting may prevent neovascularization in patients with blinding neovascular eye diseases, including macular degeneration. VEGF antagonists have therapeutic benefits but also can elicit off-target effects. Here, we found that the Ras pathway, which functions downstream of a wide range of cytokines including VEGF, is active in the growing vascular front of developing and pathological vascular networks. The endogenous Ras inhibitor p120RasGAP was expressed predominately in quiescent VEGF-insensitive endothelial cells and was ectopically downregulated in multiple neovascular models. MicroRNA-132 negatively regulated p120RasGAP expression. Experimental delivery of α-miR-132 to developing mouse eyes disrupted tip cell Ras activity and prevented angiogenic sprouting. This strategy prevented ocular neovascularization in multiple rodent models even more potently than the VEGF antagonist, VEGF-trap. Targeting microRNA-132 as a therapeutic strategy may prove useful for treating multiple neovascular diseases of the eye and for preventing vision loss regardless of the neovascular stimulus.


eLife | 2016

Hypoxia-induced metabolic stress in retinal pigment epithelial cells is sufficient to induce photoreceptor degeneration

Toshihide Kurihara; Peter D Westenskow; Marin L. Gantner; Yoshihiko Usui; Andrew Schultz; Stephen Bravo; Edith Aguilar; Carli M Wittgrove; M. Friedlander; Liliana P Paris; Emily Y. Chew; Gary Siuzdak; Martin Friedlander

Photoreceptors are the most numerous and metabolically demanding cells in the retina. Their primary nutrient source is the choriocapillaris, and both the choriocapillaris and photoreceptors require trophic and functional support from retinal pigment epithelium (RPE) cells. Defects in RPE, photoreceptors, and the choriocapillaris are characteristic of age-related macular degeneration (AMD), a common vision-threatening disease. RPE dysfunction or death is a primary event in AMD, but the combination(s) of cellular stresses that affect the function and survival of RPE are incompletely understood. Here, using mouse models in which hypoxia can be genetically triggered in RPE, we show that hypoxia-induced metabolic stress alone leads to photoreceptor atrophy. Glucose and lipid metabolism are radically altered in hypoxic RPE cells; these changes impact nutrient availability for the sensory retina and promote progressive photoreceptor degeneration. Understanding the molecular pathways that control these responses may provide important clues about AMD pathogenesis and inform future therapies. DOI: http://dx.doi.org/10.7554/eLife.14319.001


Investigative Ophthalmology & Visual Science | 2012

Using Flow Cytometry to Compare the Dynamics of Photoreceptor Outer Segment Phagocytosis in iPS-Derived RPE Cells

Peter D Westenskow; Stacey K. Moreno; Tim U. Krohne; Toshihide Kurihara; Saiyong Zhu; Zhen Ning Zhang; Tongbiao Zhao; Yang Xu; Sheng Ding; Martin Friedlander

PURPOSE Retinal pigment epithelium (RPE) autologous grafts can be readily derived from induced pluripotent stem (iPS) cells. It is critical to stringently characterize iPS-RPE using standardized and quantifiable methods to be confident that they are safe and adequate replacements for diseased RPE before utilizing them in clinical settings. One important and required function is that the iPS-RPE phagocytose photoreceptor outer segments (POS). METHODS We developed a flow cytometry-based assay to monitor binding and internalization of FITC labeled POS by ARPE-19, human fetal RPE (hfRPE), and two types of iPS-RPE. Expression and density of α(v)β₅ integrin, CD36, and MerTK receptors, which are required for phagocytosis, were compared. RESULTS Trypsinization of treated RPE cells results in the release of bound POS. The number of freed POS, the percentage of cells that internalized POS, the brightness of the FITC signal from the cells, and the surface density of the phagocytosis receptors on single RPE cells were measured using flow cytometry. These assays reveal that receptor density is dynamic during differentiation and this can affect the binding and internalization dynamics of the RPE cells. Highly differentiated iPS-RPE phagocytose POS more efficiently than hfRPE. CONCLUSIONS Caution should be exercised to not use RPE grafts until demonstrating that they are fully functional. The density of the phagocytosis receptors is dynamic and may be used as a predictor for how well the iPS-RPE cells will function in vivo. The phagocytosis dynamics observed between iPS-RPE and primary RPE is very encouraging and adds to mounting evidence that iPS-RPE may be a viable replacement for dysfunctional or dying RPE in human patients.


Advances in Experimental Medicine and Biology | 2014

Hypoxia-Inducible Factor (HIF)/Vascular Endothelial Growth Factor (VEGF) Signaling in the Retina

Toshihide Kurihara; Peter D Westenskow; Martin Friedlander

Over a span of two decades, it has become increasingly clear that vascular endothelial growth factor (VEGF) plays an important role in the pathogenesis of retinal diseases including age-related macular degeneration (AMD) and diabetic retinopathy (DR). Based on these observations, anti-VEGF therapies are being developed and approved for clinical use in the treatment of neovascular eye diseases. Hypoxia-inducible factors (HIFs) are transcriptional factors that are stabilized and activated under hypoxic conditions and induce expression of gene products, including VEGF, that are required for cell survival under hypoxia. Here we discuss recent findings from our lab and others that define roles of the HIF-VEGF axis in the retina.


Journal of Clinical Investigation | 2017

VEGF regulates local inhibitory complement proteins in the eye and kidney

Lindsay S. Keir; Rachel Firth; Lyndsey Aponik; Daniel Feitelberg; Susumu Sakimoto; Edith Aguilar; Gavin I. Welsh; Anna Richards; Yoshihiko Usui; Simon C. Satchell; Valeryia Kuzmuk; Richard J M Coward; Jonathan Goult; Katherine R. Bull; Ruchi Sharma; Kapil Bharti; Peter D Westenskow; Iacovos P. Michael; Moin A. Saleem; Martin Friedlander

Outer retinal and renal glomerular functions rely on specialized vasculature maintained by VEGF that is produced by neighboring epithelial cells, the retinal pigment epithelium (RPE) and podocytes, respectively. Dysregulation of RPE- and podocyte-derived VEGF is associated with neovascularization in wet age-related macular degeneration (ARMD), choriocapillaris degeneration, and glomerular thrombotic microangiopathy (TMA). Since complement activation and genetic variants in inhibitory complement factor H (CFH) are also features of both ARMD and TMA, we hypothesized that VEGF and CFH interact. Here, we demonstrated that VEGF inhibition decreases local CFH and other complement regulators in the eye and kidney through reduced VEGFR2/PKC-&agr;/CREB signaling. Patient podocytes and RPE cells carrying disease-associated CFH genetic variants had more alternative complement pathway deposits than controls. These deposits were increased by VEGF antagonism, a common wet ARMD treatment, suggesting that VEGF inhibition could reduce cellular complement regulatory capacity. VEGF antagonism also increased markers of endothelial cell activation, which was partially reduced by genetic complement inhibition. Together, these results suggest that VEGF protects the retinal and glomerular microvasculature, not only through VEGFR2-mediated vasculotrophism, but also through modulation of local complement proteins that could protect against complement-mediated damage. Though further study is warranted, these findings could be relevant for patients receiving VEGF antagonists.


Journal of Cell Biology | 2011

Astrocyte pVHL and HIF-α isoforms are required for embryonic-to-adult vascular transition in the eye

Toshihide Kurihara; Peter D Westenskow; Tim U. Krohne; Edith Aguilar; Randall S. Johnson; Martin Friedlander

The von Hippel–Lindau tumor suppressor and the hypoxia-inducible factor-αs are essential for the transition from embryonic hyaloidal vascular system to adult retinal vasculature in the mouse eye.

Collaboration


Dive into the Peter D Westenskow's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Edith Aguilar

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yoshihiko Usui

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Daniel Feitelberg

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Carli M Wittgrove

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Liliana P Paris

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Stephen Bravo

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Felicitas Bucher

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Gary Siuzdak

Scripps Research Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge