Network


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

Hotspot


Dive into the research topics where Yoshikazu Imanishi is active.

Publication


Featured researches published by Yoshikazu Imanishi.


Nature Neuroscience | 2004

Essential role of Ca2+-binding protein 4, a Cav1.4 channel regulator, in photoreceptor synaptic function

Françoise Haeseleer; Yoshikazu Imanishi; Tadao Maeda; Daniel E. Possin; Akiko Maeda; Amy Lee; Fred Rieke; Krzysztof Palczewski

CaBP1–8 are neuronal Ca2+-binding proteins with similarity to calmodulin (CaM). Here we show that CaBP4 is specifically expressed in photoreceptors, where it is localized to synaptic terminals. The outer plexiform layer, which contains the photoreceptor synapses with secondary neurons, was thinner in the Cabp4−/− mice than in control mice. Cabp4−/− retinas also had ectopic synapses originating from rod bipolar and horizontal cells tha HJt extended into the outer nuclear layer. Responses of Cabp4−/− rod bipolars were reduced in sensitivity about 100-fold. Electroretinograms (ERGs) indicated a reduction in cone and rod synaptic function. The phenotype of Cabp4−/− mice shares similarities with that of incomplete congenital stationary night blindness (CSNB2) patients. CaBP4 directly associated with the C-terminal domain of the Cav1.4 α1-subunit and shifted the activation of Cav1.4 to hyperpolarized voltages in transfected cells. These observations indicate that CaBP4 is important for normal synaptic function, probably through regulation of Ca2+ influx and neurotransmitter release in photoreceptor synaptic terminals.


Journal of Biological Chemistry | 2003

Pharmacological Chaperone-mediated in Vivo Folding and Stabilization of the P23H-opsin Mutant Associated with Autosomal Dominant Retinitis Pigmentosa

Syed Mohammed Noorwez; Vladimir Kuksa; Yoshikazu Imanishi; Li Zhu; Slawomir Filipek; Krzysztof Palczewski; Shalesh Kaushal

Protein conformational disorders, which include certain types of retinitis pigmentosa, are a set of inherited human diseases in which mutant proteins are misfolded and often aggregated. Many opsin mutants associated with retinitis pigmentosa, the most common being P23H, are misfolded and retained within the cell. Here, we describe a pharmacological chaperone, 11-cis-7-ring retinal, that quantitatively induces the in vivo folding of P23H-opsin. The rescued protein forms pigment, acquires mature glycosylation, and is transported to the cell surface. Additionally, we determined the temperature stability of the rescued protein as well as the reactivity of the retinal-opsin Schiff base to hydroxylamine. Our study unveils novel properties of P23H-opsin and its interaction with the chromophore. These properties suggest that 11-cis-7-ring retinal may be a useful therapeutic agent for the rescue of P23H-opsin and the prevention of retinal degeneration.


Journal of Biological Chemistry | 2002

Dual-substrate Specificity Short Chain Retinol Dehydrogenases from the Vertebrate Retina

Françoise Haeseleer; Geeng Fu Jang; Yoshikazu Imanishi; C.A.G.G. Driessen; Masazumi Matsumura; Peter S. Nelson; Krzysztof Palczewski

Retinoids are chromophores involved in vision, transcriptional regulation, and cellular differentiation. Members of the short chain alcohol dehydrogenase/reductase superfamily catalyze the transformation of retinol to retinal. Here, we describe the identification and properties of three enzymes from a novel subfamily of four retinol dehydrogenases (RDH11–14) that display dual-substrate specificity, uniquely metabolizing all-trans- andcis-retinols with C15 pro-Rspecificity. RDH11–14 could be involved in the first step of all-trans- and 9-cis-retinoic acid production in many tissues. RDH11–14 fill the gap in our understanding of 11-cis-retinal and all-trans-retinal transformations in photoreceptor (RDH12) and retinal pigment epithelial cells (RDH11). The dual-substrate specificity of RDH11 explains the minor phenotype associated with mutations in 11-cis-retinol dehydrogenase (RDH5) causing fundus albipunctatus in humans and engineered mice lacking RDH5. Furthermore, photoreceptor RDH12 could be involved in the production of 11-cis-retinal from 11-cis-retinol during regeneration of the cone visual pigments. These newly identified enzymes add new elements to important retinoid metabolic pathways that have not been explained by previous genetic and biochemical studies.


Journal of Biological Chemistry | 2005

Role of Photoreceptor-specific Retinol Dehydrogenase in the Retinoid Cycle in Vivo

Akiko Maeda; Tadao Maeda; Yoshikazu Imanishi; Vladimir Kuksa; Andrei Alekseev; J. Darin Bronson; Houbin Zhang; Li Zhu; Wenyu Sun; David A. Saperstein; Fred Rieke; Wolfgang Baehr; Krzysztof Palczewski

The retinoid cycle is a recycling system that replenishes the 11-cis-retinal chromophore of rhodopsin and cone pigments. Photoreceptor-specific retinol dehydrogenase (prRDH) catalyzes reduction of all-trans-retinal to all-trans-retinol and is thought to be a key enzyme in the retinoid cycle. We disrupted mouse prRDH (human gene symbol RDH8) gene expression by targeted recombination and generated a homozygous prRDH knock-out (prRDH–/–) mouse. Histological analysis and electron microscopy of retinas from 6- to 8-week-old prRDH–/– mice revealed no structural differences of the photoreceptors or inner retina. For brief light exposure, absence of prRDH did not affect the rate of 11-cis-retinal regeneration or the decay of Meta II, the activated form of rhodopsin. Absence of prRDH, however, caused significant accumulation of all-trans-retinal following exposure to bright lights and delayed recovery of rod function as measured by electroretinograms and single cell recordings. Retention of all-trans-retinal resulted in slight overproduction of A2E, a condensation product of all-trans-retinal and phosphatidylethanolamine. We conclude that prRDH is an enzyme that catalyzes reduction of all-trans-retinal in the rod outer segment, most noticeably at higher light intensities and prolonged illumination, but is not an essential enzyme of the retinoid cycle.


Progress in Retinal and Eye Research | 2003

Rhodopsin phosphorylation: 30 years later

Tadao Maeda; Yoshikazu Imanishi; Krzysztof Palczewski

Phototransduction in vertebrate photoreceptor cells mediated by rhodopsin is one of the most comprehensively examined G protein-coupled receptor (GPCR) signaling pathways. The signal transduction pathway can be mapped from the initial absorption of light to conformational changes within rhodopsin, through activation of the G protein transducin, and to the ultimate closure of the cation cGMP-gated channels in the plasma membrane. Furthermore, phototransduction has become an intensely studied model system for understanding the desensitizing processes that allow reduced non-linear responses of photoreceptor cells to increasing levels of illumination. Although some general themes appear to occur in GPCR systems, the details of these desensitizing processes are likely to be specific to each of the receptors. These differences are attributed to the fact that each receptor has unique kinetic constraints, amplification levels, tolerance to basal constitutive activity, intracellular internalization and recycling, redundancy of isoforms, and morphologies of the cell of their expression. One of the biochemical processes that are believed to be a common part of this desensitization of the GPCR-mediated cascade is receptor phosphorylation catalyzed by members of a small family of the GPCR kinases. The enzymatic, physiological and genetic aspects of rhodopsin phosphorylation and rhodopsin kinase have been characterized extensively over the last 30 yr. However, new structurally based approaches to examining rhodopsin kinase and rhodopsin phosphorylation are still awaiting further investigations. We present here a summary of the current understanding of rhodopsin phosphorylation and the properties of rhodopsin kinase, along with some expectations of future investigations into these topics.


Cell Metabolism | 2008

RBP4 Disrupts Vitamin A Uptake Homeostasis in a STRA6-Deficient Animal Model for Matthew-Wood Syndrome

Andrea Isken; Marcin Golczak; Vitus Oberhauser; Silke Hunzelmann; Wolfgang Driever; Yoshikazu Imanishi; Krzysztof Palczewski; Johannes von Lintig

The cellular uptake of vitamin A from its RBP4-bound circulating form (holo-RBP4) is a homeostatic process that evidently depends on the multidomain membrane protein STRA6. In humans, mutations in STRA6 are associated with Matthew-Wood syndrome, manifested by multisystem developmental malformations. Here we addressed the metabolic basis of this inherited disease. STRA6-dependent transfer of retinol from RBP4 into cultured NIH 3T3 fibroblasts was enhanced by lecithin:retinol acyltransferase (LRAT). The retinol transfer was bidirectional, strongly suggesting that STRA6 acts as a retinol channel/transporter. Loss-of-function analysis in zebrafish embryos revealed that Stra6 deficiency caused vitamin A deprivation of the developing eyes. We provide evidence that, in the absence of Stra6, holo-Rbp4 provokes nonspecific vitamin A excess in several embryonic tissues, impairing retinoic acid receptor signaling and gene regulation. These fatal consequences of Stra6 deficiency, including craniofacial and cardiac defects and microphthalmia, were largely alleviated by reducing embryonic Rbp4 levels by morpholino oligonucleotide or pharmacological treatments.


FEBS Letters | 1998

A novel subtype of G-protein-coupled receptor kinase, GRK7, in teleost cone photoreceptors

Osamu Hisatomi; Shinji Matsuda; Takunori Satoh; Shuichi Kotaka; Yoshikazu Imanishi; Fumio Tokunaga

Two kinds of retinal cDNA fragments (OlGRK‐R and ‐C) encoding the putative G‐protein‐coupled receptor kinases (GRKs) were isolated from medaka, Oryzias latipes. OlGRK‐R appears to be closely related to the rhodopsin kinase (RK) found in the outer segments of mammalian photoreceptors, but the deduced amino acid sequence of OlGRK‐C shows less than 50% identity to those of GRKs known to date, suggesting that OlGRK‐C is a novel GRK subtype (GRK7). The mRNA of OlGRK‐R is detectable in rods, and that of OlGRK‐C is found in all four types of cone photoreceptor. The C‐terminal of OlGRK‐R has a consensus sequence for farnesylation, whereas, surprisingly, OlGRK‐C has a consensus sequence for geranylgeranylation. Our result are consistent with the concept that lower vertebrates have rod‐ and cone‐specific opsin kinases.


European Journal of Neuroscience | 2002

Characterization of retinal guanylate cyclase-activating protein 3 (GCAP3) from zebrafish to man

Yoshikazu Imanishi; Ning Li; Izabela Sokal; Mathew E. Sowa; Olivier Lichtarge; Theodore G. Wensel; David A. Saperstein; Wolfgang Baehr; Krzysztof Palczewski

Calmodulin‐like neuronal Ca2+‐binding proteins (NCBPs) are expressed primarily in neurons and contain a combination of four functional and nonfunctional EF‐hand Ca2+‐binding motifs. The guanylate cyclase‐activating proteins 1–3 (GCAP1–3), the best characterized subgroup of NCBPs, function in the regulation of transmembrane guanylate cyclases 1–2 (GC1–2). The pairing of GCAPs and GCs in vivo depends on cell expression. Therefore, we investigated the expression of these genes in retina using in situ hybridization and immunocytochemistry. Our results demonstrate that GCAP1, GCAP2, GC1 and GC2 are expressed in human rod and cone photoreceptors, while GCAP3 is expressed exclusively in cones. As a consequence of extensive modification, the GCAP3 gene is not expressed in mouse retina. However, this lack of evolutionary conservation appears to be restricted to only some species as we cloned all three GCAPs from teleost (zebrafish) retina and localized them to rod cells, short single cones (GCAP1–2), and all subtypes of cones (GCAP3). Furthermore, sequence comparisons and evolutionary trace analysis coupled with functional testing of the different GCAPs allowed us to identify the key conserved residues that are critical for GCAP structure and function, and to define class‐specific residues for the NCBP subfamilies.


Molecular Pharmacology | 2006

Effects of Potent Inhibitors of the Retinoid Cycle on Visual Function and Photoreceptor Protection from Light Damage in Mice

Akiko Maeda; Tadao Maeda; Marcin Golczak; Yoshikazu Imanishi; Patrick Leahy; Ryo Kubota; Krzysztof Palczewski

Regeneration of the chromophore 11-cis-retinal is essential for the generation of light-sensitive visual pigments in the vertebrate retina. A deficiency in 11-cis-retinal production leads to congenital blindness in humans; however, a buildup of the photoisomerized chromophore can also be detrimental. Such is the case when the photoisomerized all-trans-retinal is produced but cannot be efficiently cleared from the internal membrane of the outer segment discs. Sustained increase of all-trans-retinal can lead to the formation of toxic condensation products in the eye. Thus, there is a need for potent, selective inhibitors that can regulate the flux of retinoids through the metabolism pathway termed the visual (retinoid) cycle. Here we systematically study the effects of the most potent inhibitor of this cycle, retinylamine (Ret-NH2), on visual function in mice. Prolonged, sustainable, but reversible suppression of the visual function was observed by Ret-NH2 as a result of its storage in a prodrug form, N-retinylamides. Direct comparison of other inhibitors such as fenretinide and 13-cis-retinoic acid showed multiple advantages of Ret-NH2 and its amides, including a higher potency, specificity, and lower transcription activation. Our results also revealed that mice treated with Ret-NH2 were completely resistant to the light-induced retina damage. As an experimental tool, Ret-NH2 allows the replacement of the native chromophore with synthetic analogs in wild-type mice to better understand the function of the chromophore in the activation of rhodopsin and its metabolism through the retinoid cycle.


Journal of Biological Chemistry | 2006

Retinol Dehydrogenase (RDH12) Protects Photoreceptors from Light-induced Degeneration in Mice

Akiko Maeda; Tadao Maeda; Yoshikazu Imanishi; Wenyu Sun; Beata Jastrzebska; Denise A. Hatala; H.J. Winkens; Klaus Peter Hofmann; J.J.M. Janssen; Wolfgang Baehr; C.A.G.G. Driessen; Krzysztof Palczewski

RDH12 has been suggested to be one of the retinol dehydrogenases (RDH) involved in the vitamin A recycling system (visual cycle) in the eye. Loss of function mutations in the RDH12 gene were recently reported to be associated with autosomal recessive childhood-onset severe retinal dystrophy. Here we show that RDH12 localizes to the photoreceptor inner segments and that deletion of this gene in mice slows the kinetics of all-trans-retinal reduction, delaying dark adaptation. However, accelerated 11-cis-retinal production and increased susceptibility to light-induced photoreceptor apoptosis were also observed in Rdh12-/- mice, suggesting that RDH12 plays a unique, nonredundant role in the photoreceptor inner segments to regulate the flow of retinoids in the eye. Thus, severe visual impairments of individuals with null mutations in RDH12 may likely be caused by light damage1.

Collaboration


Dive into the Yoshikazu Imanishi's collaboration.

Top Co-Authors

Avatar

Krzysztof Palczewski

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tadao Maeda

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Akiko Maeda

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Guilian Tian

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Ina Nemet

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Philip Ropelewski

Case Western Reserve University

View shared research outputs
Researchain Logo
Decentralizing Knowledge