Network


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

Hotspot


Dive into the research topics where Michael W. Stuck is active.

Publication


Featured researches published by Michael W. Stuck.


Human Molecular Genetics | 2010

Differences in RDS trafficking, assembly and function in cones versus rods: insights from studies of C150S-RDS

Dibyendu Chakraborty; Shannon M. Conley; Michael W. Stuck; Muna I. Naash

Cysteine 150 of retinal degeneration slow protein (RDS) mediates the intermolecular disulfide bonding necessary for large RDS complex assembly and morphogenesis of the rim region of photoreceptor outer segments. Previously, we showed that cones have a different requirement for RDS than rods, but the nature of that difference was unclear. Here, we express oligomerization-incompetent RDS (C150S-RDS) in the cone-dominant nrl(-/-) mouse. Expression of C150S-RDS leads to dominant functional abnormalities, ultrastructural changes, biochemical anomalies and protein mislocalization in cones. These data suggest that RDS complexes in cones are more susceptible to disruption than those in rods, possibly due to structural or microenvironmental differences in the two cell types. Furthermore, our results suggest that RDS intermolecular disulfide bonding may be part of RDS inner-segment assembly in cones but not in rods. These data highlight significant differences in assembly, trafficking and function of RDS in rods versus cones.


PLOS ONE | 2012

Defects in the Outer Limiting Membrane Are Associated with Rosette Development in the Nrl−/− Retina

Michael W. Stuck; Shannon M. Conley; Muna I. Naash

The neural retinal leucine zipper (Nrl) knockout mouse is a widely used model to study cone photoreceptor development, physiology, and molecular biology in the absence of rods. In the Nrl−/− retina, rods are converted into functional cone-like cells. The Nrl−/− retina is characterized by large undulations of the outer nuclear layer (ONL) commonly known as rosettes. Here we explore the mechanism of rosette development in the Nrl−/− retina. We report that rosettes first appear at postnatal day (P)8, and that the structure of nascent rosettes is morphologically distinct from what is seen in the adult retina. The lumen of these nascent rosettes contains a population of aberrant cells protruding into the subretinal space that induce infolding of the ONL. Morphologically adult rosettes do not contain any cell bodies and are first detected at P15. The cells found in nascent rosettes are photoreceptors in origin but lack inner and outer segments. We show that the adherens junctions between photoreceptors and Müller glia which comprise the retinal outer limiting membrane (OLM) are not uniformly formed in the Nrl−/− retina and thus allow protrusion of a population of developing photoreceptors into the subretinal space where their maturation becomes delayed. These data suggest that the rosettes of the Nrl−/− retina arise due to defects in the OLM and delayed maturation of a subset of photoreceptors, and that rods may play an important role in the proper formation of the OLM.


Human Molecular Genetics | 2014

Insights into the mechanisms of macular degeneration associated with the R172W mutation in RDS.

Shannon M. Conley; Michael W. Stuck; Justin L. Burnett; Dibyendu Chakraborty; Seifollah Azadi; Steven J. Fliesler; Muna I. Naash

Mutations in the photoreceptor tetraspanin gene peripherin-2/retinal degeneration slow (PRPH2/RDS) cause both rod- and cone-dominant diseases. While rod-dominant diseases, such as autosomal dominant retinitis pigmentosa, are thought to arise due to haploinsufficiency caused by loss-of-function mutations, the mechanisms underlying PRPH2-associated cone-dominant diseases are unclear. Here we took advantage of a transgenic mouse line expressing an RDS mutant (R172W) known to cause macular degeneration (MD) in humans. To facilitate the study of cones in the heavily rod-dominant mouse retina, R172W mice were bred onto an Nrl(-/-) background (in which developing rods adopt a cone-like fate). In this model the R172W protein and the key RDS-binding partner, rod outer segment (OS) membrane protein 1 (ROM-1), were properly expressed and trafficked to cone OSs. However, the expression of R172W led to dominant defects in cone structure and function with equal effects on S- and M-cones. Furthermore, the expression of R172W in cones induced subtle alterations in RDS/ROM-1 complex assembly, specifically resulting in the formation of abnormal, large molecular weight ROM-1 complexes. Fundus imaging demonstrated that R172W mice developed severe clinical signs of disease nearly identical to those seen in human MD patients, including retinal degeneration, retinal pigment epithlium (RPE) defects and loss of the choriocapillaris. Collectively, these data identify a primary disease-causing molecular defect in cone cells and suggest that RDS-associated disease in patients may be a result of this defect coupled with secondary sequellae involving RPE and choriocapillaris cell loss.


Progress in Retinal and Eye Research | 2016

PRPH2/RDS and ROM-1: Historical context, current views and future considerations

Michael W. Stuck; Shannon M. Conley; Muna I. Naash

Peripherin 2 (PRPH2), also known as RDS (retinal degeneration slow) is a photoreceptor specific glycoprotein which is essential for normal photoreceptor health and vision. PRPH2/RDS is necessary for the proper formation of both rod and cone photoreceptor outer segments, the organelle specialized for visual transduction. When PRPH2/RDS is defective or absent, outer segments become disorganized or fail to form entirely and the photoreceptors subsequently degenerate. Multiple PRPH2/RDS disease-causing mutations have been found in humans, and they are associated with various blinding diseases of the retina such as macular degeneration and retinitis pigmentosa, the vast majority of which are inherited dominantly, though recessive LCA and digenic RP have also been associated with RDS mutations. Since its initial discovery, the scientific community has dedicated a considerable amount of effort to understanding the molecular function and disease mechanisms of PRPH2/RDS. This work has led to an understanding of how the PRPH2/RDS molecule assembles into complexes and functions as a necessary part of the machinery that forms new outer segment discs, as well as leading to fundamental discoveries about the mechanisms that underlie OS biogenesis. Here we discuss PRPH2/RDS-associated research and how experimental results have driven the understanding of the PRPH2/RDS protein and its role in human disease.


Human Molecular Genetics | 2014

The Y141C knockin mutation in RDS leads to complex phenotypes in the mouse

Michael W. Stuck; Shannon M. Conley; Muna I. Naash

Mutations in the photoreceptor-specific gene peripherin-2 (PRPH-2, also known as retinal degeneration slow/RDS) cause incurable retinal degeneration with a high degree of phenotypic variability. Patient phenotypes range from retinitis pigmentosa to various forms of macular and pattern dystrophy. Macular and pattern dystrophy in particular are associated with complex, poorly understood disease mechanisms, as severe vision loss is often associated both with defects in the photoreceptors, as well as the choroid and retinal pigment epithelium (RPE). Since there is currently no satisfactory model to study pattern dystrophy disease mechanisms, we generated a knockin mouse model expressing an RDS pattern dystrophy mutation, Y141C. Y141C mice exhibited clinical signs similar to those in patients including late-onset fundus abnormalities characteristic of RPE and choroidal defects and electroretinogram defects. Ultrastructural examination indicated that disc formation was initiated by the Y141C protein, but proper sizing and alignment of discs required wild-type RDS. The biochemical mechanism underlying these abnormalities was tied to defects in the normal process of RDS oligomerization which is required for proper RDS function. Y141C-RDS formed strikingly abnormal disulfide-linked complexes which were localized to the outer segment (OS) where they impaired the formation of proper OS structure. These data support a model of pattern dystrophy wherein a primary molecular defect occurring in all photoreceptors leads to secondary sequellae in adjacent tissues, an outcome which leads to macular vision loss. An understanding of the role of RDS in the interplay between these tissues significantly enhances our understanding of RDS-associated pathobiology and our ability to design rational treatment strategies.


PLOS ONE | 2015

SNAREs Interact with Retinal Degeneration Slow and Rod Outer Segment Membrane Protein-1 during Conventional and Unconventional Outer Segment Targeting

Rahel Zulliger; Shannon M. Conley; Maggie Mwoyosvi; Michael W. Stuck; Seifollah Azadi; Muna I. Naash

Mutations in the photoreceptor protein peripherin-2 (also known as RDS) cause severe retinal degeneration. RDS and its homolog ROM-1 (rod outer segment protein 1) are synthesized in the inner segment and then trafficked into the outer segment where they function in tetramers and covalently linked larger complexes. Our goal is to identify binding partners of RDS and ROM-1 that may be involved in their biosynthetic pathway or in their function in the photoreceptor outer segment (OS). Here we utilize several methods including mass spectrometry after affinity purification, in vitro co-expression followed by pull-down, in vivo pull-down from mouse retinas, and proximity ligation assay to identify and confirm the SNARE proteins Syntaxin 3B and SNAP-25 as novel binding partners of RDS and ROM-1. We show that both covalently linked and non-covalently linked RDS complexes interact with Syntaxin 3B. RDS in the mouse is trafficked from the inner segment to the outer segment by both conventional (i.e., Golgi dependent) and unconventional secretory pathways, and RDS from both pathways interacts with Syntaxin3B. Syntaxin 3B and SNAP-25 are enriched in the inner segment (compared to the outer segment) suggesting that the interaction with RDS/ROM-1 occurs in the inner segment. Syntaxin 3B and SNAP-25 are involved in mediating fusion of vesicles carrying other outer segment proteins during outer segment targeting, so could be involved in the trafficking of RDS/ROM-1.


Journal of Biological Chemistry | 2015

Retinal Degeneration Slow (RDS) Glycosylation Plays a Role in Cone Function and in the Regulation of RDS·ROM-1 Protein Complex Formation

Michael W. Stuck; Shannon M. Conley; Muna I. Naash

Background: Mutations in retinal degeneration slow (RDS) lead to rod and cone-dominant retinal degeneration by mechanisms that remain unknown. Results: Knockin mice carrying unglycosylated RDS have reduced RDS levels and functional defects in cones. Conclusion: RDS glycosylation is important for cone but not rod function. Significance: Differential reliance on glycosylation may help explain why some RDS disease mutations target cones and others target rods. The photoreceptor-specific glycoprotein retinal degeneration slow (RDS, also called PRPH2) is necessary for the formation of rod and cone outer segments. Mutations in RDS cause rod and cone-dominant retinal disease, and it is well established that both cell types have different requirements for RDS. However, the molecular mechanisms for this difference remain unclear. Although RDS glycosylation is highly conserved, previous studies have revealed no apparent function for the glycan in rods. In light of the highly conserved nature of RDS glycosylation, we hypothesized that it is important for RDS function in cones and could underlie part of the differential requirement for RDS in the two photoreceptor subtypes. We generated a knockin mouse expressing RDS without the N-glycosylation site (N229S). Normal levels of RDS and the unglycosylated RDS binding partner rod outer segment membrane protein 1 (ROM-1) were found in N229S retinas. However, cone electroretinogram responses were decreased by 40% at 6 months of age. Because cones make up only 3–5% of photoreceptors in the wild-type background, N229S mice were crossed into the nrl−/− background (in which all rods are converted to cone-like cells) for biochemical analysis. In N229S/nrl−/− retinas, RDS and ROM-1 levels were decreased by ∼60% each. These data suggest that glycosylation of RDS is required for RDS function or stability in cones, a difference that may be due to extracellular versus intradiscal localization of the RDS glycan in cones versus rods.


Human Molecular Genetics | 2017

Rom1 converts Y141C-Prph2-associated pattern dystrophy to retinitis pigmentosa

Shannon M. Conley; Michael W. Stuck; Jamie N. Watson; Muna I. Naash

&NA; Mutations in peripherin 2 (PRPH2), also known as retinal degeneration slow/RDS, lead to various retinal degenerations including retinitis pigmentosa (RP) and macular/pattern dystrophy (MD/PD). PRPH2‐associated disease is often characterized by a phenotypic variability even within families carrying the same mutation, raising interest in potential modifiers. PRPH2 oligomerizes with its homologue rod outer segment (OS) membrane protein 1 (ROM1), and non‐pathogenic PRPH2/ROM1 mutations, when present together, lead to digenic RP. We asked whether ROM1 could modify the phenotype of a PRPH2 mutation associated with a high degree of intrafamilial phenotypic heterogeneity: Y141C. In vitro, Y141C‐Prph2 showed signs of retention in the endoplasmic reticulum (ER), however co‐expression with Rom1 rescued this phenotype. In the heterozygous Y141C knockin mouse model (Prph2Y/+), Y141C‐Prph2 and Rom1 formed abnormal complexes but were present at normal levels. Abnormal complexes were eliminated in the absence of Rom1 (Prph2Y/+/Rom1‐/‐) and total Prph2 levels were reduced to those found in the haploinsufficient Prph2+/‐ RP model. The biochemical changes had functional and structural consequences; while Prph2Y/+ animals exhibited a cone‐rod electroretinogram defect, Prph2Y/+/Rom1‐/‐ animals displayed a rod‐dominant phenotype and OSs similar to those seen in the Prph2+/‐. These data show that ablation of Rom1 results in the conversion of an MD/PD phenotype characterized by cone functional defects and the formation of abnormal Prph2/Rom1 complexes to an RP phenotype characterized by rod‐dominant functional defects and reductions in total Prph2 protein. Thus one method by which ROM1 may act as a disease modifier is by contributing to the large variability in PRPH2‐associated disease phenotypes.


Advances in Experimental Medicine and Biology | 2016

RDS Functional Domains and Dysfunction in Disease

Michael W. Stuck; Shannon M. Conley; Muna I. Naash

The photoreceptor specific tetraspanin protein retina degeneration slow (RDS) is a critical component of the machinery necessary for the formation of rod and cone outer segments. Over 80 individual pathogenic mutations in RDS have been identified in human patients that lead to a wide variety of retinal degenerative diseases including retinitis pigmentosa, cone-rod dystrophy, and various forms of macular dystrophy. RDS-associated disease is characterized by a high degree of variability in phenotype and penetrance, making analysis of the underlying molecular mechanisms of interest difficult. Here we summarize our modern understanding of RDS functional domains and oligomerization and how disruption of these domains and complexes could contribute to the variety of disease pathologies seen in human patients with RDS mutations.


Human Molecular Genetics | 2018

Prph2 initiates outer segment morphogenesis but maturation requires Prph2/Rom1 oligomerization

Shannon M. Conley; Michael W. Stuck; Jamie N. Watson; Rahel Zulliger; Justin L. Burnett; Muna I. Naash

Abstract The retinal disease gene peripherin 2 (PRPH2) is essential for the formation of photoreceptor outer segments (OSs), where it functions in oligomers with and without its homologue ROM1. However, the precise role of these proteins in OS morphogenesis is not understood. By utilizing a knock‐in mouse expressing a chimeric protein comprised of the body of Rom1 and the C‐terminus of Prph2 (termed RRCT), we find that the Prph2 C‐terminus is necessary and sufficient for the initiation of OSs, while OS maturation requires the body of Prph2 and associated large oligomers. Importantly, dominant‐negative physiological and biochemical defects in RRCT heterozygous rods are rescued by removing Rom1, suggesting Rom1 is a regulator for OS formation. Our experiments evaluating Prph2 trafficking show that Rom1 is a key determinant of whether Prph2 complexes utilize conventional versus unconventional (Golgi bypass) secretory pathways to reach the OS. These findings significantly advance our understanding of the molecular underpinnings of OS morphogenesis and particularly the role of Rom1.

Collaboration


Dive into the Michael W. Stuck's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Shannon M. Conley

University of Oklahoma Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar

Dibyendu Chakraborty

University of Oklahoma Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jamie N. Watson

University of Oklahoma Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar

Justin L. Burnett

University of Oklahoma Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar

Seifollah Azadi

University of Oklahoma Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

David M. Sherry

University of Oklahoma Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar

Hemant Khanna

University of Massachusetts Medical School

View shared research outputs
Researchain Logo
Decentralizing Knowledge