Network


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

Hotspot


Dive into the research topics where Catherine Bowes Rickman is active.

Publication


Featured researches published by Catherine Bowes Rickman.


Progress in Retinal and Eye Research | 2010

The Pivotal Role of the Complement System in Aging and Age-related Macular Degeneration: Hypothesis Re-visited

Don H. Anderson; Monte J. Radeke; Natasha Gallo; Ethan A. Chapin; P.T. Johnson; Christy R. Curletti; Lisa S. Hancox; Jane Hu; J.N. Ebright; Goldis Malek; Michael A. Hauser; Catherine Bowes Rickman; Dean Bok; Gregory S. Hageman; Lincoln V. Johnson

During the past ten years, dramatic advances have been made in unraveling the biological bases of age-related macular degeneration (AMD), the most common cause of irreversible blindness in western populations. In that timeframe, two distinct lines of evidence emerged which implicated chronic local inflammation and activation of the complement cascade in AMD pathogenesis. First, a number of complement system proteins, complement activators, and complement regulatory proteins were identified as molecular constituents of drusen, the hallmark extracellular deposits associated with early AMD. Subsequently, genetic studies revealed highly significant statistical associations between AMD and variants of several complement pathway-associated genes including: Complement factor H (CFH), complement factor H-related 1 and 3 (CFHR1 and CFHR3), complement factor B (CFB), complement component 2 (C2), and complement component 3 (C3). In this article, we revisit our original hypothesis that chronic local inflammatory and immune-mediated events at the level of Bruchs membrane play critical roles in drusen biogenesis and, by extension, in the pathobiology of AMD. Secondly, we report the results of a new screening for additional AMD-associated polymorphisms in a battery of 63 complement-related genes. Third, we identify and characterize the local complement system in the RPE-choroid complex - thus adding a new dimension of biological complexity to the role of the complement system in ocular aging and AMD. Finally, we evaluate the most salient, recent evidence that bears directly on the role of complement in AMD pathogenesis and progression. Collectively, these recent findings strongly re-affirm the importance of the complement system in AMD. They lay the groundwork for further studies that may lead to the identification of a transcriptional disease signature of AMD, and hasten the development of new therapeutic approaches that will restore the complement-modulating activity that appears to be compromised in genetically susceptible individuals.


Annual Review of Genomics and Human Genetics | 2009

Unraveling a multifactorial late-onset disease: from genetic susceptibility to disease mechanisms for age-related macular degeneration.

Anand Swaroop; Emily Y. Chew; Catherine Bowes Rickman; Gonçalo R. Abecasis

Aging-associated neurodegenerative diseases significantly influence the quality of life of affected individuals. Genetic approaches, combined with genomic technology, have provided powerful insights into common late-onset diseases, such as age-related macular degeneration (AMD). Here, we discuss current findings on the genetics of AMD to highlight areas of rapid progress and new challenges. We also attempt to integrate available genetic and biochemical data with cellular pathways involved in aging to formulate an integrated model of AMD pathogenesis.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Anti-amyloid therapy protects against retinal pigmented epithelium damage and vision loss in a model of age-related macular degeneration

Jindong Ding; Lincoln V. Johnson; Rolf Herrmann; Sina Farsiu; Stephanie Smith; Marybeth Groelle; Brian E. Mace; Patrick M. Sullivan; Jeffrey A. Jamison; Una Kelly; Ons Harrabi; Sangeetha Subbarao Bollini; Jeanette Dilley; Dione Kobayashi; Bing Kuang; Wenlin Li; Jaume Pons; John C. Lin; Catherine Bowes Rickman

Age-related macular degeneration (AMD) is a leading cause of visual dysfunction worldwide. Amyloid β (Aβ) peptides, Aβ1–40 (Aβ40) and Aβ1–42 (Aβ42), have been implicated previously in the AMD disease process. Consistent with a pathogenic role for Aβ, we show here that a mouse model of AMD that invokes multiple factors that are known to modify AMD risk (aged human apolipoprotein E 4 targeted replacement mice on a high-fat, cholesterol-enriched diet) presents with Aβ-containing deposits basal to the retinal pigmented epithelium (RPE), histopathologic changes in the RPE, and a deficit in scotopic electroretinographic response, which is reflective of impaired visual function. Strikingly, these electroretinographic deficits are abrogated in a dose-dependent manner by systemic administration of an antibody targeting the C termini of Aβ40 and Aβ42. Concomitant reduction in the levels of Aβ and activated complement components in sub-RPE deposits and structural preservation of the RPE are associated with anti-Aβ40/42 antibody immunotherapy and visual protection. These observations are consistent with the reduction in amyloid plaques and improvement of cognitive function in mouse models of Alzheimers disease treated with anti-Aβ antibodies. They also implicate Aβ in the pathogenesis of AMD and identify Aβ as a viable therapeutic target for its treatment.


Vision Research | 2008

Targeting age-related macular degeneration with Alzheimer’s disease based immunotherapies: Anti-amyloid-β antibody attenuates pathologies in an age-related macular degeneration mouse model

Jindong Ding; John C. Lin; Brian E. Mace; Rolf Herrmann; Patrick M. Sullivan; Catherine Bowes Rickman

Age-related macular degeneration (AMD) is a late-onset, neurodegenerative retinal disease that shares several clinical and pathological features with Alzheimers disease (AD) including extracellular deposits containing amyloid-beta (Abeta) peptides. Immunotherapy targeting the Abeta protein has been investigated as a potential treatment for AD. Here, we present the rationale for extending this approach to treat AMD. We tested an anti-Abeta antibody administered systemically in a mouse model of AMD. Histological and functional measurements in treated animals compared to controls showed that following immunotherapy, the amounts of Abeta in the retina and brain were decreased and the ERG deficits in the retina were attenuated. These data support the hypothesis that Abeta is a therapeutic target for AMD.


Autophagy | 2014

Dysregulated autophagy in the RPE is associated with increased susceptibility to oxidative stress and AMD

Sayak K. Mitter; Chunjuan Song; Xiaoping Qi; Haoyu Mao; Haripriya Vittal Rao; Debra Akin; Alfred S. Lewin; Maria B. Grant; William A. Dunn; Jindong Ding; Catherine Bowes Rickman; Michael E. Boulton

Autophagic dysregulation has been suggested in a broad range of neurodegenerative diseases including age-related macular degeneration (AMD). To test whether the autophagy pathway plays a critical role to protect retinal pigmented epithelial (RPE) cells against oxidative stress, we exposed ARPE-19 and primary cultured human RPE cells to both acute (3 and 24 h) and chronic (14 d) oxidative stress and monitored autophagy by western blot, PCR, and autophagosome counts in the presence or absence of autophagy modulators. Acute oxidative stress led to a marked increase in autophagy in the RPE, whereas autophagy was reduced under chronic oxidative stress. Upregulation of autophagy by rapamycin decreased oxidative stress-induced generation of reactive oxygen species (ROS), whereas inhibition of autophagy by 3-methyladenine (3-MA) or by knockdown of ATG7 or BECN1 increased ROS generation, exacerbated oxidative stress-induced reduction of mitochondrial activity, reduced cell viability, and increased lipofuscin. Examination of control human donor specimens and mice demonstrated an age-related increase in autophagosome numbers and expression of autophagy proteins. However, autophagy proteins, autophagosomes, and autophagy flux were significantly reduced in tissue from human donor AMD eyes and 2 animal models of AMD. In conclusion, our data confirm that autophagy plays an important role in protection of the RPE against oxidative stress and lipofuscin accumulation and that impairment of autophagy is likely to exacerbate oxidative stress and contribute to the pathogenesis of AMD.


Proceedings of the National Academy of Sciences of the United States of America | 2015

Regulation of age-related macular degeneration-like pathology by complement factor H

Christopher B. Toomey; Una Kelly; Daniel R. Saban; Catherine Bowes Rickman

Significance Age-related macular degeneration (AMD) affects approximately one-third of Americans over 70 and is characterized by lipoprotein-rich sub-retinal pigmented epithelium (sub-RPE) deposits. Substantial evidence has emerged that implicates complement factor H (CFH) in the pathogenesis of AMD. Here, we conduct an in vivo analysis to elucidate the role of CFH in AMD pathology. We show that (i) CFH and lipoproteins compete for binding in the sub-RPE extracellular matrix such that decreasing CFH leads to lipoprotein accumulation and sub-RPE deposit formation; and (ii) detrimental complement activation within sub-RPE deposits leads to RPE damage and vision loss. This new understanding of the complicated interactions of CFH in development of AMD-like pathology paves the way for identifying more targeted therapeutic strategies for AMD. Complement factor H (CFH) is a major susceptibility gene for age-related macular degeneration (AMD); however, its impact on AMD pathobiology is unresolved. Here, the role of CFH in the development of AMD pathology in vivo was interrogated by analyzing aged Cfh+/− and Cfh−/− mice fed a high-fat, cholesterol-enriched diet. Strikingly, decreased levels of CFH led to increased sub-retinal pigmented epithelium (sub-RPE) deposit formation, specifically basal laminar deposits, following high-fat diet. Mechanistically, our data show that deposits are due to CFH competition for lipoprotein binding sites in Bruch’s membrane. Interestingly and despite sub-RPE deposit formation occurring in both Cfh+/− and Cfh−/− mice, RPE damage accompanied by loss of vision occurred only in old Cfh+/− mice. We demonstrate that such pathology is a function of excess complement activation in Cfh+/− mice versus complement deficiency in Cfh−/− animals. Due to the CFH-dependent increase in sub-RPE deposit height, we interrogated the potential of CFH as a previously unidentified regulator of Bruch’s membrane lipoprotein binding and show, using human Bruch’s membrane explants, that CFH removes endogenous human lipoproteins in aged donors. Thus, advanced age, high-fat diet, and decreased CFH induce sub-RPE deposit formation leading to complement activation, which contributes to RPE damage and visual function impairment. This new understanding of the complicated interactions of CFH in AMD-like pathology provides an improved foundation for the development of targeted therapies for AMD.


Biomedical Optics Express | 2012

Automatic segmentation of closed-contour features in ophthalmic images using graph theory and dynamic programming.

Stephanie J. Chiu; Cynthia A. Toth; Catherine Bowes Rickman; Joseph A. Izatt; Sina Farsiu

This paper presents a generalized framework for segmenting closed-contour anatomical and pathological features using graph theory and dynamic programming (GTDP). More specifically, the GTDP method previously developed for quantifying retinal and corneal layer thicknesses is extended to segment objects such as cells and cysts. The presented technique relies on a transform that maps closed-contour features in the Cartesian domain into lines in the quasi-polar domain. The features of interest are then segmented as layers via GTDP. Application of this method to segment closed-contour features in several ophthalmic image types is shown. Quantitative validation experiments for retinal pigmented epithelium cell segmentation in confocal fluorescence microscopy images attests to the accuracy of the presented technique.


Journal of Immunology | 2010

Heparan Sulfate, Including That in Bruch’s Membrane, Inhibits the Complement Alternative Pathway: Implications for Age-Related Macular Degeneration

Una Kelly; L. Yu; Pallavi Kumar; Jin Dong Ding; Haixiang Jiang; Gregory S. Hageman; Vadim Y. Arshavsky; Michael M. Frank; Michael A. Hauser; Catherine Bowes Rickman

An imbalance between activation and inhibition of the complement system has been implicated in the etiologies of numerous common diseases. Allotypic variants of a key complement fluid-phase regulatory protein, complement factor H (CFH), are strongly associated with age-related macular degeneration (AMD), a leading cause of worldwide visual dysfunction, although its specific role in AMD pathogenesis is still not clear. CFH was isolated from individuals carrying combinations of two of the nonsynonymous coding variants most strongly associated with AMD risk, V62/H402 (risk haplotype variants), I62/Y402 (nonrisk haplotype variants), and V62/Y402. These proteins were used in two functional assays (cell surface- and fluid-phase–based) measuring cofactor activity of CFH in the factor I-mediated cleavage of C3b. Although no variant-specific differences in the cofactor activity were detected, when heparan sulfate (HS) was added to these assays, it accelerated the rate of C3b cleavage, and this effect could be modulated by degree of HS sulfation. Bruch’s membrane/choroid, a site of tissue damage in AMD, contains high concentrations of glycosaminoglycans, including HS. Addition of human Bruch’s membrane/choroid to the fluid-phase assay accelerated the C3b cleavage, and this effect was lost posttreatment of the tissue with heparinase III. Binding of CFH variants to Bruch’s membrane/choroid isolated from elderly, non-AMD donor eyes, was similar, as was the functional activity of bound CFH. These findings refine our understanding of interactions of HS and complement and support the hypothesis that these interactions play a role in the transition between normal aging and AMD in Bruch’s membrane/choroid.


The Journal of Neuroscience | 2011

ApoER2 Function in the Establishment and Maintenance of Retinal Synaptic Connectivity

Justin Trotter; Martin Klein; Umesh K. Jinwal; Jose F. Abisambra; Chad A. Dickey; Jeremy Tharkur; Irene Masiulis; Jindong Ding; Kirsten G. Locke; Catherine Bowes Rickman; David G. Birch; Edwin J. Weeber; Joachim Herz

The cellular and molecular mechanisms responsible for the development of inner retinal circuitry are poorly understood. Reelin and apolipoprotein E (apoE), ligands of apoE receptor 2 (ApoER2), are involved in retinal development and degeneration, respectively. Here we describe the function of ApoER2 in the developing and adult retina. ApoER2 expression was highest during postnatal inner retinal synaptic development and was considerably lower in the mature retina. Both during development and in the adult, ApoER2 was expressed by A-II amacrine cells. ApoER2 knock-out (KO) mice had rod bipolar morphogenic defects, altered A-II amacrine dendritic development, and impaired rod-driven retinal responses. The presence of an intact ApoER2 NPxY motif, necessary for binding Disabled-1 and transducing the Reelin signal, was also necessary for development of the rod bipolar pathway, while the alternatively spliced exon 19 was not. Mice deficient in another Reelin receptor, very low-density lipoprotein receptor (VLDLR), had normal rod bipolar morphology but altered A-II amacrine dendritic development. VLDLR KO mice also had reductions in oscillatory potentials and delayed synaptic response intervals. Interestingly, age-related reductions in rod and cone function were observed in both ApoER2 and VLDLR KOs. These results support a pivotal role for ApoER2 in the establishment and maintenance of normal retinal synaptic connectivity.


Developmental Brain Research | 1999

Characterization of the cell death promoter, Bad, in the developing rat retina and forebrain

Dennis W. Rickman; Randall E Nacke; Catherine Bowes Rickman

Neuronal programmed cell death, or apoptosis, occurs during development, following injury or in certain disease processes, and is regulated by members of the B-cell leukemia-2 (Bcl-2) protein family. These molecules include both positive and negative regulators of cell death and act by selective dimerization that results in permissive or inhibitory effects on a cascade of cellular events, including mitochondrial release of cytochrome c, stimulation of cysteine protease activity and subsequent cellular deterioration. Here, we have characterized the expression of the cell death agonist, Bad, in the postnatal rat retina and forebrain. Isolation, subsequent amplification by RT-PCR and DNA sequence analysis revealed that retinal Bad was identical to Bad expressed in the developing and adult rat brain. Using a polyclonal antibody to Bad, we determined that, in the retina, on the day of birth (postnatal day-0, PND-0) Bad immunoreactivity was expressed primarily by retinal ganglion cells, some cells in the inner neuroblastic layer (NBL) and an indistinct plexus of processes in the inner plexiform layer (IPL). On PND-7, Bad immunoreactivity was observed in most cells in the ganglion cell layer (GCL), numerous cells scattered throughout the inner nuclear layer (INL), a lightly stained IPL and in a distinct band of immunostained fibers in the forming outer plexiform layer (OPL). By PND-15, Bad immunoreactivity was present in cells in the GCL, in some cells in the proximal INL and in horizontal cell processes in the OPL. The IPL was only faintly labeled. In the adult retina, specific Bad immunostaining was confined to large cells in the ganglion cell layer (presumed ganglion cells), occasional lightly stained horizontal cells and their processes in the OPL and to occasional small, lightly stained cells in the proximal INL (presumed amacrine cells) and GCL (presumed displaced amacrine cells). Again, the interposed IPL was faintly labeled. In the brain, Bad immunoreactive cells were scattered throughout the forebrain parenchyma but were particularly concentrated in neurons of the cerebral cortex, hippocampus and amygdala. Bad immunoreactivity was heaviest in these cells at PND-7, distinctly weaker at PND-10 and absent by PND-24. At all time points examined, Bad immunoreactivity was present in epithelial cells of the choroid plexus, as previously reported in the adult rat brain. These data suggest that Bad is transiently expressed by various cell types in the perinatal retina, particularly ganglion cells, and in discrete forebrain regions. In the context of corroborative observations, Bad expression may be regulated in response to acute ischemia and may act as a control point for retinal neuronal apoptosis.

Collaboration


Dive into the Catherine Bowes Rickman'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