Network


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

Hotspot


Dive into the research topics where Yumiko Umino is active.

Publication


Featured researches published by Yumiko Umino.


Nature Medicine | 2007

Restoration of cone vision in a mouse model of achromatopsia

John J. Alexander; Yumiko Umino; Drew Everhart; Bo Chang; Seok Hong Min; Qiuhong Li; Adrian M. Timmers; Norman L Hawes; Ji-jing Pang; Robert B. Barlow; William W. Hauswirth

Loss of cone function in the central retina is a pivotal event in the development of severe vision impairment for many prevalent blinding diseases. Complete achromatopsia is a genetic defect resulting in cone vision loss in 1 in 30,000 individuals. Using adeno-associated virus (AAV) gene therapy, we show that it is possible to target cones and rescue both the cone-mediated electroretinogram response and visual acuity in the Gnat2 cpfl3 mouse model of achromatopsia.


The Journal of Neuroscience | 2008

Speed, Spatial, and Temporal Tuning of Rod and Cone Vision in Mouse

Yumiko Umino; Eduardo Solessio; Robert B. Barlow

Rods and cones subserve mouse vision over a 100 million-fold range of light intensity (−6 to 2 log cd m−2). Rod pathways tune vision to the temporal frequency of stimuli (peak, 0.75 Hz) and cone pathways to their speed (peak, ∼12°/s). Both pathways tune vision to the spatial components of stimuli (0.064–0.128 cycles/°). The specific photoreceptor contributions were determined by two-alternative, forced-choice measures of contrast thresholds for optomotor responses of C57BL/6J mice with normal vision, Gnat2cpfl3 mice without functional cones, and Gnat1−/− mice without functional rods. Gnat2cpfl3 mice (threshold, −6.0 log cd m−2) cannot see rotating gratings above −2.0 log cd m−2 (photopic vision), and Gnat1−/− mice (threshold, −4.0 log cd m−2) are blind below −4.0 log cd m−2 (scotopic vision). Both genotypes can see in the transitional mesopic range (−4.0 to −2.0 log cd m−2). Mouse rod and cone sensitivities are similar to those of human. This parametric study characterizes the functional properties of the mouse visual system, revealing the rod and cone contributions to contrast sensitivity and to the temporal processing of visual stimuli.


Molecular Therapy | 2011

Long-term retinal function and structure rescue using capsid mutant AAV8 vector in the rd10 mouse, a model of recessive retinitis pigmentosa.

Ji-jing Pang; Xufeng Dai; Shannon E. Boye; Ilaria Barone; Sanford L. Boye; Song Mao; Drew Everhart; Astra Dinculescu; Li Liu; Yumiko Umino; Bo Lei; Bo Chang; Robert B. Barlow; Enrica Strettoi; William W. Hauswirth

The retinal degeneration 10 (rd10) mouse is a well-characterized model of autosomal recessive retinitis pigmentosa (RP), which carries a spontaneous mutation in the β subunit of rod cGMP-phosphodiesterase (PDEβ). Rd10 mouse exhibits photoreceptor dysfunction and rapid rod photoreceptor degeneration followed by cone degeneration and remodeling of the inner retina. Here, we evaluate whether gene replacement using the fast-acting tyrosine-capsid mutant AAV8 (Y733F) can provide long-term therapy in this model. AAV8 (Y733F)-smCBA-PDEβ was subretinally delivered to postnatal day 14 (P14) rd10 mice in one eye only. Six months after injection, spectral domain optical coherence tomography (SD-OCT), electroretinogram (ERG), optomotor behavior tests, and immunohistochemistry showed that AAV8 (Y733F)-mediated PDEβ expression restored retinal function and visual behavior and preserved retinal structure in treated rd10 eyes for at least 6 months. This is the first demonstration of long-term phenotypic rescue by gene therapy in an animal model of PDEβ-RP. It is also the first example of tyrosine-capsid mutant AAV8 (Y733F)-mediated correction of a retinal phenotype. These results lay the groundwork for the development of PDEβ-RP gene therapy trial and suggest that tyrosine-capsid mutant AAV vectors may be effective for treating other rapidly degenerating models of retinal degeneration.


PLOS ONE | 2010

Functional and Behavioral Restoration of Vision by Gene Therapy in the Guanylate Cyclase-1 (GC1) Knockout Mouse

Shannon E. Boye; Sanford L. Boye; Ji-jing Pang; Renee C. Ryals; Drew Everhart; Yumiko Umino; Andy W. Neeley; Joseph C. Besharse; Robert B. Barlow; William W. Hauswirth

Background Recessive mutations in guanylate cyclase-1 (Gucy2d) are associated with severe, early onset Leber congenital amaurosis-1(LCA1). Gucy2d encodes guanylate cyclase (GC1) is expressed in photoreceptor outer segment membranes and produces cGMP in these cells. LCA1 patients present in infancy with severely impaired vision and extinguished electroretinogram (ERG) but retain some photoreceptors in both their macular and peripheral retina for years. Like LCA1 patients, loss of cone function in the GC1 knockout (GC1KO) mouse precedes cone degeneration. The purpose of this study was to test whether delivery of functional GC1 to cone cells of the postnatal GC1KO mouse could restore function to these cells. Methodology/Principal Findings Serotype 5 AAV vectors containing either a photoreceptor-specific, rhodopsin kinase (hGRK1) or ubiquitous (smCBA) promoter driving expression of wild type murine GC1 were subretinally delivered to one eye of P14 GC1KO mice. Visual function (ERG) was analyzed in treated and untreated eyes until 3 months post injection. AAV-treated, isogenic wild type and uninjected control mice were evaluated for restoration of visual behavior using optomotor testing. At 3 months post injection, all animals were sacrificed, and their treated and untreated retinas assayed for expression of GC1 and localization of cone arrestin. Cone-mediated function was restored to treated eyes of GC1KO mice (ERG amplitudes were ∼45% of normal). Treatment effect was stable for at least 3 months. Robust improvements in cone-mediated visual behavior were also observed, with responses of treated mice being similar or identical to that of wild type mice. AAV-vectored GC1 expression was found in photoreceptors and cone cells were preserved in treated retinas. Conclusions/Significance This is the first demonstration of gene-based restoration of both visual function/vision-elicited behavior and cone preservation in a mammalian model of GC1 deficiency. Importantly, results were obtained using a well characterized, clinically relevant AAV vector. These results lay the ground work for the development of an AAV-based gene therapy vector for the treatment of LCA1.


Journal of Vision | 2008

Monitoring mouse retinal degeneration with high-resolution spectral-domain optical coherence tomography

Ki Hean Kim; Mehron Puoris'haag; Gopi N. Maguluri; Yumiko Umino; Karen Cusato; Robert B. Barlow; Johannes F. de Boer

Progression of retinal degeneration in a mouse model was studied in vivo with high-resolution spectral-domain optical coherence tomography (SD-OCT). Imaging in 3D with high depth resolution (<3 mum), SD-OCT resolved all the major layers of the retina of control C57BL/6J mice. Images of transgenic mice having a null mutation of the rhodopsin gene revealed the anatomical consequences of retinal degeneration: thinning of the outer retina, including the outer plexiform layer (OPL), outer nuclear layer (ONL), and inner and outer segments (IS/OS). We monitored the progression of retinal degeneration in rd1 mice (C3H/HeJ) by periodically imaging the same mice from the time the pups opened their eyes on P13 to P34. SD-OCT images showed that the outer retina (OPL, ONL, IS/OS) had already thinned by 73% (100 to 27 mum) at eye opening. The retina continued to degenerate, and by P20 the outer retina was not resolvable. The thickness of entire retina decreased from 228 mum (control) to 152 mum on P13 and to 98 mum by P34, a 57% reduction with the complete loss in the outer retina. In summary, we show that SD-OCT can monitor the progression of retinal degeneration in transgenic mice.


PLOS ONE | 2012

AAV-mediated cone rescue in a naturally occurring mouse model of CNGA3-achromatopsia.

Ji-jing Pang; Wen-Tao Deng; Xufeng Dai; Bo Lei; Drew Everhart; Yumiko Umino; Jie Li; Keqing Zhang; Song Mao; Sanford L. Boye; Li Liu; Vince A. Chiodo; Xuan Liu; Wei Shi; Ye Tao; Bo Chang; William W. Hauswirth

Achromatopsia is a rare autosomal recessive disorder which shows color blindness, severely impaired visual acuity, and extreme sensitivity to bright light. Mutations in the alpha subunits of the cone cyclic nucleotide-gated channels (CNGA3) are responsible for about 1/4 of achromatopsia in the U.S. and Europe. Here, we test whether gene replacement therapy using an AAV5 vector could restore cone-mediated function and arrest cone degeneration in the cpfl5 mouse, a naturally occurring mouse model of achromatopsia with a CNGA3 mutation. We show that gene therapy leads to significant rescue of cone-mediated ERGs, normal visual acuities and contrast sensitivities. Normal expression and outer segment localization of both M- and S-opsins were maintained in treated retinas. The therapeutic effect of treatment lasted for at least 5 months post-injection. This study is the first demonstration of substantial, relatively long-term restoration of cone-mediated light responsiveness and visual behavior in a naturally occurring mouse model of CNGA3 achromatopsia. The results provide the foundation for development of an AAV5-based gene therapy trial for human CNGA3 achromatopsia.


Visual Neuroscience | 2005

Anesthesia can cause sustained hyperglycemia in C57/BL6J mice

E.T. Brown; Yumiko Umino; T. Loi; Eduardo Solessio; Robert B. Barlow

Effects of anesthesia on the blood glucose of C57/BL6J mice were evaluated under conditions commonly used for testing retinal sensitivity with electroretinographic (ERG) recordings. We evaluated the effects of four anesthetics: nembutal (50 mg/kg), pentothal (100 mg/kg), avertin (240 mg/kg), and ketamine/xylazine (100 mg/kg) using saline as control. We measured blood glucose (BG) levels from tail vein blood before and 15 and 60 min following intraperitoneal injections. Fifteen minutes postinjection, all four anesthetics and saline elevated BG with ketamine/xylazine and avertin having substantially greater effects than nembutal, pentothal, and saline. Only the effects of ketamine/xylazine and avertin persisted throughout the test period. Sixty minutes after injecting ketamine/xylazine BG remained elevated at 400 +/- 42 mg/dl, a 167% increase over preinjection levels. Sixty minutes after injecting avertin BG was 288 +/- 10 mg/dl, a 59% increase over preinjection levels. No sustained elevation in BG was detected 60 min following injection of nembutal, pentothal, or saline. Because BG can affect the amplitude of the ERG, caution should be exercised in the use of ketamine/xylazine or avertin. The choice of anesthesia may also be important in diabetes and metabolism research where changes in blood glucose could impact physiological processes.


Gene Therapy | 2010

Self-complementary AAV-mediated gene therapy restores cone function and prevents cone degeneration in two models of Rpe65 deficiency

Ji-jing Pang; Shannon E. Boye; Bo Lei; Sanford L. Boye; Drew Everhart; Renee C. Ryals; Yumiko Umino; Bärbel Rohrer; John J. Alexander; Jie Li; Xufeng Dai; Qiuhong Li; Bo Chang; Robert B. Barlow; William W. Hauswirth

To test whether fast-acting, self-complimentary (sc), adeno-associated virus-mediated RPE65 expression prevents cone degeneration and/or restores cone function, we studied two mouse lines: the Rpe65-deficient rd12 mouse and the Rpe65-deficient, rhodopsin null (‘that is, cone function-only’) Rpe65−/−::Rho−/− mouse. scAAV5 expressing RPE65 was injected subretinally into one eye of rd12 and Rpe65−/−::Rho−/− mice at postnatal day 14 (P14). Contralateral rd12 eyes were injected later, at P35. Rd12 behavioral testing revealed that rod vision loss was prevented with either P14 or P35 treatment, whereas cone vision was only detected after P14 treatment. Consistent with this observation, P35 treatment only restored rod electroretinogram (ERG) signals, a result likely due to reduced cone densities at this time point. For Rpe65−/−::Rho−/− mice in which there is no confounding rod contribution to the ERG signal, cone cells and cone-mediated ERGs were also maintained with treatment at P14. This work establishes that a self-complimentary AAV5 vector can restore substantial visual function in two genetically distinct models of Rpe65 deficiency within 4 days of treatment. In addition, this therapy prevents cone degeneration but only if administered before extensive cone degeneration, thus supporting continuation of current Lebers congenital amaurosis-2 clinical trials with an added emphasis on cone subtype analysis and early intervention.


Human Molecular Genetics | 2009

Impaired cone function and cone degeneration resulting from CNGB3 deficiency: down-regulation of CNGA3 biosynthesis as a potential mechanism

Xi-Qin Ding; Cynthia S. Harry; Yumiko Umino; Alexander V. Matveev; Steven J. Fliesler; Robert B. Barlow

The cone cyclic nucleotide-gated (CNG) channel is essential for central and color vision and visual acuity. This channel is composed of two structurally related subunits, CNGA3 and CNGB3; CNGA3 is the ion-conducting subunit, whereas CNGB3 is a modulatory subunit. Mutations in both subunits are associated with achromatopsia and progressive cone dystrophy, with mutations in CNGB3 alone accounting for 50% of all known cases of achromatopsia. However, the molecular mechanisms underlying cone diseases that result from CNGB3 deficiency are unknown. This study investigated the role of CNGB3 in cones, using CNGB3(-/-) mice. Cone dysfunction was apparent at the earliest time point examined (post-natal day 30) in CNGB3(-/-) mice. When compared with wild-type (WT) controls: photopic electroretingraphic (ERG) responses were decreased by approximately 75%, whereas scotopic ERG responses were unchanged; visual acuity was decreased by approximately 20%, whereas contrast sensitivity was unchanged; cone density was reduced by approximately 40%; photoreceptor apoptosis was detected; and outer segment disorganization was observed in some cones. Notably, CNGA3 protein and mRNA levels were significantly decreased in CNGB3(-/-) mice; in contrast, mRNA levels of S-opsin, Gnat2 and Pde6c were unchanged, relative to WT mice. Hence, we show that loss of CNGB3 reduces biosynthesis of CNGA3 and impairs cone CNG channel function. We suggest that down-regulation of CNGA3 contributes to the pathogenic mechanism by which CNGB3 mutations lead to human cone disease.


Proceedings of the National Academy of Sciences of the United States of America | 2006

Hypoglycemia leads to age-related loss of vision

Yumiko Umino; D. Everhart; Eduardo Solessio; K. Cusato; J. C. Pan; T. H. Nguyen; E. T. Brown; R. Hafler; B. A. Frio; Barry E. Knox; G. A. Engbretson; M. Haeri; L. Cui; A. S. Glenn; Maureen J. Charron; Robert B. Barlow

The retina is among the most metabolically active tissues in the body, requiring a constant supply of blood glucose to sustain function. We assessed the impact of low blood glucose on the vision of C57BL/6J mice rendered hypoglycemic by a null mutation of the glucagon receptor gene, Gcgr. Metabolic stress from moderate hypoglycemia led to late-onset loss of retinal function in Gcgr−/− mice, loss of visual acuity, and eventual death of retinal cells. Retinal function measured by the electroretinogram b-wave threshold declined >100-fold from age 9 to 13 months, whereas decreases in photoreceptor function measured by the ERG a-wave were delayed by 3 months. At 10 months of age Gcgr−/− mice began to lose visual acuity and exhibit changes in retinal anatomy, including an increase in cell death that was initially more pronounced in the inner retina. Decreases in retinal function and visual acuity correlated directly with the degree of hypoglycemia. This work demonstrates a metabolic-stress-induced loss of vision in mammals, which has not been described previously. Linkage between low blood glucose and loss of vision in mice may highlight the importance for glycemic control in diabetics and retinal diseases related to metabolic stress as macular degeneration.

Collaboration


Dive into the Yumiko Umino's collaboration.

Top Co-Authors

Avatar

Robert B. Barlow

State University of New York Upstate Medical University

View shared research outputs
Top Co-Authors

Avatar

Eduardo Solessio

State University of New York Upstate Medical University

View shared research outputs
Top Co-Authors

Avatar

Drew Everhart

State University of New York Upstate Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bo Chang

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bo Lei

Chongqing Medical University

View shared research outputs
Top Co-Authors

Avatar

Xufeng Dai

Wenzhou Medical College

View shared research outputs
Top Co-Authors

Avatar

Barry E. Knox

State University of New York Upstate Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge