Network


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

Hotspot


Dive into the research topics where Ronald A. Bush is active.

Publication


Featured researches published by Ronald A. Bush.


Nature Genetics | 2001

Nrl is required for rod photoreceptor development.

Alan J. Mears; Mineo Kondo; Prabodha K. Swain; Yuichiro Takada; Ronald A. Bush; Thomas L. Saunders; Paul A. Sieving; Anand Swaroop

The protein neural retina leucine zipper (Nrl) is a basic motif–leucine zipper transcription factor that is preferentially expressed in rod photoreceptors. It acts synergistically with Crx to regulate rhodopsin transcription. Missense mutations in human NRL have been associated with autosomal dominant retinitis pigmentosa. Here we report that deletion of Nrl in mice results in the complete loss of rod function and super-normal cone function, mediated by S cones. The photoreceptors in the Nrl−/− retina have cone-like nuclear morphology and short, sparse outer segments with abnormal disks. Analysis of retinal gene expression confirms the apparent functional transformation of rods into S cones in the Nrl−/− retina. On the basis of these findings, we postulate that Nrl acts as a molecular switch during rod-cell development by directly modulating rod-specific genes while simultaneously inhibiting the S-cone pathway through the activation of Nr2e3.


Gene Therapy | 2009

Intravitreal delivery of AAV8 retinoschisin results in cell type-specific gene expression and retinal rescue in the Rs1-KO mouse.

Tae Kwann Park; Zhijian Wu; Sten Kjellstrom; Yong Zeng; Ronald A. Bush; Paul A. Sieving; Peter Colosi

X-linked juvenile retinoschisis (XLRS) is a neurodevelopmental abnormality caused by retinoschisin gene mutations. XLRS is characterized by splitting through the retinal layers and impaired synaptic transmission of visual signals resulting in impaired acuity and a propensity to retinal detachment. Several groups have treated murine retinoschisis models successfully using adeno-associated virus (AAV) vectors. Owing to the fragile nature of XLRS retina, translating this therapy to the clinic may require an alternative to invasive subretinal vector administration. Here we show that all layers of the retinoschisin knockout (Rs1-KO) mouse retina can be transduced efficiently with AAV vectors administered by simple vitreous injection. Retinoschisin expression was restricted to the neuroretina using a new vector that uses a 3.5-kb human retinoschisin promoter and an AAV type 8 capsid. Intravitreal administration to Rs1-KO mice resulted in robust retinoschisin expression with a retinal distribution similar to that observed in wild-type retina, including the expression by the photoreceptors lying deep in the retina. No off-target expression was observed. Rs1-KO mice treated with this vector showed a decrease in the schisis cavities and had improved retinal signaling evaluated by recording the electroretinogram 11–15 weeks after the application.


Visual Neuroscience | 1999

The electroretinogram of the rhodopsin knockout mouse

Kazushige Toda; Ronald A. Bush; Peter Humphries; Paul A. Sieving

The electroretinogram (ERG) of the rhodopsin knockout (rho-/-) mouse of Humphries et al. (1997) (Humphries et al., 1997) was studied for evidence of light-evoked rod activity and to describe the cone function. The rho-/- retina develops normal numbers of rod and cone nuclei, but the rods have no outer segments, and no rhodopsin is found by immunohistochemistry. The dark-adapted ERG threshold was elevated 4.7 log units above wild-type (WT) control mice, indicating that any residual rod responses were reduced >50,000-fold, consistent with a complete functional knockout. The dark-adapted rho-/- ERG had a cone waveform, and the spectral sensitivity peaked near 510 nm for both dark-adapted and light-adapted conditions, without evidence of a Purkinje shift. The light-adapted ERG b-wave amplitude of young rho-/- mice was the same as WT. The amplitude remained steady up to postnatal day P47, but thereafter it declined to only 1-2% by P80 when no cone outer segments remained. Cone b-wave threshold of dark-adapted rho-/- mice was -1.07 +/- 0.39 log cd-s/m2 (n = 17), which is 1.27 log units more sensitive than light-adapted thresholds against a rod-suppressing Ganzfeld background of 1.61 log scotopic cd/m2. This indicates that dark-adapted WT responses to still dimmer stimuli are exclusively rod driven with minimal cone intrusion. Above this cone threshold intensity, the dark-adapted b-wave of WT will be a summation of rod and cone responses. Threshold versus intensity (TVI) studies gave no evidence of a rod influence on the mouse cone b-wave.


Visual Neuroscience | 2001

Characterization of the rod photoresponse isolated from the dark-adapted primate ERG.

Jamison Ja; Ronald A. Bush; Lei B; Paul A. Sieving

The a-wave of the human dark-adapted ERG is thought to derive from activity of rod photoreceptors. However, other sources within the retina could potentially perturb this simple equation. We investigated the extent to which the short-latency dark-adapted rod a-wave of the primate ERG is dominated by the rod photoresponse and the applicability of the phototransduction model to fit the rod a-wave. Dark-adapted Ganzfeld ERGs were elicited over a 5-log-unit intensity range using short bright xenon flashes, and the light-adapted cone responses were subtracted to isolate the rod ERG a-wave. Intravitreal 4-phosphono-butyric acid (APB) and cis-2,3-piperidine-dicarboxylic acid (PDA) were applied to isolate the photoreceptor response. The Hood and Birch version of the phototransduction model, Rmax[1 - e(-I x S x (t-t(eff)))2], was fitted to the a-wave data while allowing Rmax and S to vary. Three principle observations were made: (1) At flash intensities > or =0.77 log sc-td-s the leading edge of the normalized rod ERG a-wave tracks the isolated photoreceptor response across the first 20 ms or up to the point of b-wave intrusion. The rod ERG a-wave was essentially identical to the isolated receptor response for all intensities that produce peak responses within 14 ms after the flash. (2) The best fit of sensitivity (S) was not affected by APB and/or PDA, suggesting that the inner retina contributes very little to the dark-adapted a-wave. (3) APB always reduced the maximum dark-adapted a-wave amplitude (by 15-30%), and PDA always increased it (by 7-15%). Using the phototransduction model, both events can be interpreted as a scaling of the photoreceptor dark current. This suggests that activity of postreceptor cells somehow influences the rod dark current, possibly by feedback through horizontal cells (although currently not demonstrated for the rod system), or by altering the ionic concentrations near the photoreceptors, or by neuromodulator effects mediated by dopamine or melatonin.


The Journal of Neuroscience | 2007

Constitutive Excitation by Gly90Asp Rhodopsin Rescues Rods from Degeneration Caused by Elevated Production of cGMP in the Dark

Michael L. Woodruff; Elena V. Olshevskaya; Andrey Savchenko; Igor V. Peshenko; Ronald Barrett; Ronald A. Bush; Paul A. Sieving; Gordon L. Fain; Alexander M. Dizhoor

Previous experiments indicate that congenital human retinal degeneration caused by genetic mutations that change the Ca2+ sensitivity of retinal guanylyl cyclase (retGC) can result from an increase in concentration of free intracellular cGMP and Ca2+ in the photoreceptors. To rescue degeneration in transgenic mouse models having either the Y99C or E155G mutations of the retGC modulator guanylyl cyclase-activating protein 1 (GCAP-1), which produce elevated cGMP synthesis in the dark, we used the G90D rhodopsin mutation, which produces constitutive stimulation of cGMP hydrolysis. The effects of the G90D transgene were evaluated by measuring retGC activity biochemically, by recording single rod and electroretinogram (ERG) responses, by intracellular free Ca2+ measurement, and by retinal morphological analysis. Although the G90D rhodopsin did not alter the abnormal Ca2+ sensitivity of retGC in the double-mutant animals, the intracellular free cGMP and Ca2+ concentrations returned close to normal levels, consistent with constitutive activation of the phosphodiesterase PDE6 cascade in darkness. G90D decreased the light sensitivity of rods but spared them from severe retinal degeneration in Y99C and E155G GCAP-1 mice. More than half of the photoreceptors remained alive, appeared morphologically normal, and produced electrical responses, at the time when their siblings lacking the G90D rhodopsin transgene lost the entire retinal outer nuclear layer and no longer responded to illumination. These experiments indicate that mutations that lead to increases in cGMP and Ca2+ can trigger photoreceptor degeneration but that constitutive activation of the transduction cascade in these animals can greatly enhance cell survival.


Proceedings of the National Academy of Sciences of the United States of America | 2009

Depleting Rac1 in mouse rod photoreceptors protects them from photo-oxidative stress without affecting their structure or function

Masatoshi Haruta; Ronald A. Bush; Sten Kjellström; Camasamudram Vijayasarathy; Yong Zeng; Yun-Zheng Le; Paul A. Sieving

In nonphagocytic cells, Rac1 is a component of NADPH oxidase that produces reactive oxygen species [Ushio-Fukai M (2006) Sci STKE 2006:re8]. Rac1 is expressed abundantly in mammalian retinal photoreceptors, where it is activated in response to light stimuli [Balasubramanian N, Slepak VZ (2003) Curr Biol 13:1306–1310]. We used Cre-LoxP conditional gene targeting to knock down Rac1 expression in mouse rod photoreceptors and found protection against light-induced photoreceptor death compared with WT litter-mates. We also found a similar protective effect on rods using apocynin, which inhibits NADPH oxidase activity. These results implicate both neuronal Rac1 and NADPH oxidase in cell death in this model of CNS degeneration. Studies in which dominant-mutants of Rac1 were expressed in transgenic Drosophila species demonstrated that Rac1 is a key regulator of photoreceptor morphogenesis and polarity [Chang HY, Ready DF (2000) Science 290:1978–1980]. However, we found that diminished Rac1 expression in mouse rods had no effect on retinal structure or function examined by light microscopy, electron microscopy, rhodopsin measurement, electroretinogram activity, and visual acuity, indicating rod outer segment morphogenesis proceeded normally in Rac1 conditional knockout mice. The lack of structural or functional effect of Rac1 depletion on photoreceptors, but protection under conditions of stress, indicate that the Rac1 pathway warrants exploration as a target for therapy in retinal neurodegenerative diseases.


Visual Neuroscience | 2001

Comparative structural and functional analysis of photoreceptor neurons of Rho-/- mice reveal increased survival on C57BL/6J in comparison to 129Sv genetic background

Marian M. Humphries; Sophie Kiang; Niamh McNally; Mary Anne Donovan; Paul A. Sieving; Ronald A. Bush; Shigeki Machida; Thomas G. Cotter; Audrey Hobson; Jane Farrar; Peter Humphries; Paul F. Kenna

To explore the possible influence of defined genetic backgrounds on photoreceptor viability and function in mice carrying a targeted disruption of the rhodopsin gene, the severities of retinopathies in Rho-/- mice on C57BL/6J and 129Sv congenic backgrounds were compared by light microscopy and electroretinography and qualitatively by in situ end labeling of DNA in apoptotic photoreceptor nuclei of retinal sections. Cone photoreceptor viability and function were shown to deteriorate more slowly on the C57BL/6J background in comparison to that of the 129Sv, with significantly greater numbers of outer nuclear layer nuclei in the retinas of C57BL/6J mice at 3 and 4 months of age. Both amplitude and waveform features of the ERG were shown to be remarkably different in the two strains, indicating an approximately 6-fold difference in C57BL/6J Rho-/- mice compared to 129Sv Rho-/- mice at 80 days. Thus, in comparison with the 129Sv strain, genetic modifiers appear to constitute a component of the C57BL/6J background, the expression of which significantly protects cone photoreceptors from apoptotic death in a mutation-induced murine retinopathy. The differences in phenotype revealed in this study are sufficient in principle to provide a basis for comparisons to be made between QTLs in light-induced and mutation-induced systems.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Probing potassium channel function in vivo by intracellular delivery of antibodies in a rat model of retinal neurodegeneration.

Dorit Raz-Prag; William N. Grimes; Robert N. Fariss; Camasamudram Vijayasarathy; Maria M. Campos; Ronald A. Bush; Jeffrey S. Diamond; Paul A. Sieving

Inward rectifying potassium (Kir) channels participate in regulating potassium concentration (K+) in the central nervous system (CNS), including in the retina. We explored the contribution of Kir channels to retinal function by delivering Kir antibodies (Kir-Abs) into the rat eye in vivo to interrupt channel activity. Kir-Abs were coupled to a peptide carrier to reach intracellular epitopes. Functional effects were evaluated by recording the scotopic threshold response (STR) and photopic negative response (PhNR) of the electroretinogram (ERG) noninvasively with an electrode on the cornea to determine activity of the rod and cone pathways, respectively. Intravitreal delivery of Kir2.1-Ab coupled to the peptide carrier diminished these ERG responses equivalent to dimming the stimulus 10- to 100-fold. Immunohistochemistry (IHC) showed Kir2.1 immunostaining of retinal bipolar cells (BCs) matching the labeling pattern obtained with conventional IHC of applying Kir2.1-Ab to fixed retinal sections postmortem. Whole-cell voltage-clamp BC recordings in rat acute retinal slices showed suppression of barium-sensitive Kir2.1 currents upon inclusion of Kir2.1-Ab in the patch pipette. The in vivo functional and structural results implicate a contribution of Kir2.1 channel activity in these electronegative ERG potentials. Studies with Kir4.1-Ab administered in vivo also suppressed the ERG components and showed immunostaining of Müller cells. The strategy of administering Kir antibodies in vivo, coupled to a peptide carrier to facilitate intracellular delivery, identifies roles for Kir2.1 and Kir4.1 in ERG components arising in the proximal retina and suggests this approach could be of further value in research.


Vision Research | 1997

Increased phase lag of the fundamental harmonic component of the 30 Hz flicker ERG in Schubert-Bornschein complete type CSNB

Steve H. Kim; Ronald A. Bush; Paul A. Sieving

The 30 Hz flicker electroretinogram (ERG) response was studied in seven patients with Schubert-Bornschein complete-type congenital stationary night blindness (SB-CSNB) by measuring conventional peak implicit times and by harmonic analysis. Responses were elicited with xenon photostrobe flashes. The fundamental flicker component showed a significant increased phase lag (P = 0.002), even though the peak implicit times were not delayed (P = 0.22). Although others have noted a squared-off flicker waveform in SB-CSNB, this is the first quantitative demonstration of a temporal abnormality in the flicker ERG and is the first report that a retinal disease can delay the fundamental flicker harmonic while the peak implicit time remains normal. Similarly, the phase of the flicker fundamental harmonic in monkey was also delayed after intravitreal 2-amino-4-phosphonobutyric acid (APB) that blocks the activity of depolarizing bipolar cells (DBC), whereas peak implicit time remained virtually unchanged. These observations, coupled with the normal amplitude and slope of the photopic a-wave (which was recorded under conditions that elicit a substantial contribution from hyperpolarizing bipolar cells) supports the hypothesis that the abnormality in the DBC pathway in SB-CSNB lies post-synaptic to the cones and not in the presynaptic glutamate release by cone photoreceptors.


Human gene therapy. Clinical development | 2014

Preclinical Safety Evaluation of a Recombinant AAV8 Vector for X-Linked Retinoschisis After Intravitreal Administration in Rabbits

Dario Marangoni; Zhijian Wu; Henry E. Wiley; Caroline J. Zeiss; Camasamudram Vijayasarathy; Yong Zeng; Suja Hiriyanna; Ronald A. Bush; Lisa L. Wei; Peter Colosi; Paul A. Sieving

X-linked retinoschisis (XLRS) is a retinal disease caused by mutations in the gene encoding the protein retinoschisin (RS1) and one of the most common causes of macular degeneration in young men. Currently, no FDA-approved treatments are available for XLRS and a replacement gene therapy could provide a promising strategy. We have developed a novel gene therapy approach for XLRS, based on the administration of AAV8-scRS/IRBPhRS, an adeno-associated viral vector coding the human RS1 protein, via the intravitreal route. On the basis of our prior study in an Rs1-KO mouse, this construct transduces efficiently all the retinal layers, resulting in an RS1 expression similar to that observed in the wild-type and improving retinal structure and function. In support of a clinical trial, we carried out a study to evaluate the ocular safety of intravitreal administration of AAV8-scRS/IRBPhRS into 39 New Zealand White rabbits. Two dose levels of vector, 2e(10) and 2e(11) vector genomes per eye (vg/eye), were tested and ocular inflammation was monitored over a 12-week period by serial ophthalmological and histopathological analysis. A mild ocular inflammatory reaction, consisting mainly of vitreous infiltrates, was observed within 4 weeks from injection, in both 2e(10) and 2e(11) vg/eye groups and was likely driven by the AAV8 capsid. At 12-week follow-up, ophthalmological examination revealed no clinical signs of vitreitis in either of the dose groups. However, while vitreous inflammatory infiltrate was significantly reduced in the 2e(10) vg/eye group at 12 weeks, some rabbits in the higher dose group still showed persistence of inflammatory cells, histologically. In conclusion, intravitreal administration of AAV8-scRS/IRBPhRS into the rabbit eye produces a mild and transient intraocular inflammation that resolves, at a 2e(10) vg/eye dose, within 3 months, and does not cause irreversible tissue damages. These data support the initiation of a clinical trial of intravitreal administration of AAV8-scRS/IRBPhRS in XLRS patients.

Collaboration


Dive into the Ronald A. Bush's collaboration.

Top Co-Authors

Avatar

Paul A. Sieving

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yong Zeng

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Peter Colosi

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Bo Lei

University of Michigan

View shared research outputs
Top Co-Authors

Avatar

Dorit Raz-Prag

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge