Network


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

Hotspot


Dive into the research topics where Suja Hiriyanna is active.

Publication


Featured researches published by Suja Hiriyanna.


American Journal of Human Genetics | 2002

A Comprehensive Mutation Analysis of RP2 and RPGR in a North American Cohort of Families with X-Linked Retinitis Pigmentosa

Debra K. Breuer; Beverly M. Yashar; E. Filippova; Suja Hiriyanna; Robert H. Lyons; Alan J. Mears; Bersabell Asaye; Ceren Acar; Raf Vervoort; Alan F. Wright; Maria A. Musarella; Patricia G. Wheeler; Ian M. MacDonald; Alessandro Iannaccone; David G. Birch; Dennis R. Hoffman; Gerald A. Fishman; John R. Heckenlively; Samuel G. Jacobson; Paul A. Sieving; Anand Swaroop

X-linked retinitis pigmentosa (XLRP) is a clinically and genetically heterogeneous degenerative disease of the retina. At least five loci have been mapped for XLRP; of these, RP2 and RP3 account for 10%-20% and 70%-90% of genetically identifiable disease, respectively. However, mutations in the respective genes, RP2 and RPGR, were detected in only 10% and 20% of families with XLRP. Mutations in an alternatively spliced RPGR exon, ORF15, have recently been shown to account for 60% of XLRP in a European cohort of 47 families. We have performed, in a North American cohort of 234 families with RP, a comprehensive screen of the RP2 and RPGR (including ORF15) genes and their 5 upstream regions. Of these families, 91 (39%) show definitive X-linked inheritance, an additional 88 (38%) reveal a pattern consistent with X-linked disease, and the remaining 55 (23%) are simplex male patients with RP who had an early onset and/or severe disease. In agreement with the previous studies, we show that mutations in the RP2 gene and in the original 19 RPGR exons are detected in <10% and approximately 20% of XLRP probands, respectively. Our studies have revealed RPGR-ORF15 mutations in an additional 30% of 91 well-documented families with X-linked recessive inheritance and in 22% of the total 234 probands analyzed. We suggest that mutations in an as-yet-uncharacterized RPGR exon(s), intronic changes, or another gene in the region might be responsible for the disease in the remainder of this North American cohort. We also discuss the implications of our studies for genetic diagnosis, genotype-phenotype correlations, and gene-based therapy.


American Journal of Human Genetics | 1999

Protein-truncation mutations in the RP2 gene in a North American cohort of families with X-linked retinitis pigmentosa.

Alan J. Mears; Linn Gieser; Denise Yan; Cynthia S. Chen; Stacey Fahrner; Suja Hiriyanna; Ricardo Fujita; Samuel G. Jacobson; Paul A. Sieving; Anand Swaroop

We thank Drs. Sten Andreasson, David Birch, Nancy Carson, Bernie Chodirker, Mark Evans, Gerald Fishman, John Heckenlively, Dennis Hoffman, Maria Musarella, and Beth Spriggs and Mr. Eric L. Krivchenia for some of the patient samples that were included in the mutation screening. We acknowledge the assistance of Dr. Wolfgang Berger for providing the RP2 primer sequences. We thank Dr. Monika Buraczynska for organization of the patient registry; Dr. Radha Ayyagari for discussions; Dr. Beverly Yashar for counseling; Ms. Cara Coats for assistance in patient collection; Mr. Jason Cook, Ms. Patricia Forsythe, and Ms. Eve Bingham for technical assistance; and Ms. D. Giebel for secretarial assistance. This research was supported by National Institutes of Health (NIH) grants EY05627, EY06094, and EY07961 and by grants from the Foundation Fighting Blindness, the Chatlos Foundation, the Kirby Foundation, the Mackall Trust, and Research to Prevent Blindness. We also acknowledge NIH grants EY07003 (core) and M01-RR00042 (General Clinical Research Center) and a shared equipment grant from the Office of Vice President for Research (University of Michigan). A.S. is recipient of a Lew R. Wasserman Merit Award, and P.A.S., a Senior Scientific Investigator Award, both from Research to Prevent Blindness.


Nature Communications | 2017

Nrl knockdown by AAV-delivered CRISPR/Cas9 prevents retinal degeneration in mice

Wenhan Yu; Suddhasil Mookherjee; Vijender Chaitankar; Suja Hiriyanna; Jung-Woong Kim; Matthew Brooks; Yasaman Ataeijannati; Xun Sun; Lijin Dong; Tiansen Li; Anand Swaroop; Zhijian Wu

In retinitis pigmentosa, loss of cone photoreceptors leads to blindness, and preservation of cone function is a major therapeutic goal. However, cone loss is thought to occur as a secondary event resulting from degeneration of rod photoreceptors. Here we report a genome editing approach in which adeno-associated virus (AAV)-mediated CRISPR/Cas9 delivery to postmitotic photoreceptors is used to target the Nrl gene, encoding for Neural retina-specific leucine zipper protein, a rod fate determinant during photoreceptor development. Following Nrl disruption, rods gain partial features of cones and present with improved survival in the presence of mutations in rod-specific genes, consequently preventing secondary cone degeneration. In three different mouse models of retinal degeneration, the treatment substantially improves rod survival and preserves cone function. Our data suggest that CRISPR/Cas9-mediated NRL disruption in rods may be a promising treatment option for patients with retinitis pigmentosa.


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.


Ophthalmic Genetics | 2003

Clinical studies of X-linked retinitis pigmentosa in three Swedish families with newly identified mutations in the RP2 and RPGR-ORF15 genes

Sten Andréasson; Debra K. Breuer; Louise Eksandh; Vesna Ponjavic; Christina Frennesson; Suja Hiriyanna; E. Filippova; Beverly M. Yashar; Anand Swaroop

Purpose: To describe new disease-causing RP2 and RPGR-ORF15 mutations and their corresponding clinical phenotypes in Swedish families with X-linked retinitis pigmentosa (XLRP) and to establish genotype-phenotype correlations by studying the clinical spectrum of disease in families with a known molecular defect. Methods: Seventeen unrelated families with RP and an apparent X-linked pattern of disease inheritance were identified from the Swedish RP registry and screened for mutations in the RP2 and RPGR (for the RP3 disease) genes. These families had been previously screened for the RPGR exons 1–19, and disease-causing mutations were identified in four of them. In the remaining 13 families, we sequenced the RP2 gene and the newly discovered RPGR-ORF exon. Detailed clinical evaluations were then obtained from individuals in the three families with identified mutations. Results: Mutations in RP2 and RPGRORF15 were identified in three of the 13 families. Clinical evaluations of affected males and carrier females demonstrated varying degrees of retinal dysfunction and visual handicap, with early onset and severe disease in the families with mutations in the ORF15 exon of the RPGR gene. Conclusions: A total of seven mutations in the RP2 and RPGR genes have been discovered so far in Swedish XLRP families. All affected individuals express a severe form of retinal degeneration with visual handicap early in life, although the degree of retinal dysfunction varies both in hemizygous male patients and in heterozygous carrier females. Retinal disease phenotypes in patients with mutations in the RPGR-ORF15 were more severe than in patients with mutations in RP2 or other regions of the RPGR .


Archive | 2001

X-Linked Retinitis Pigmentosa: Current Status

Debra K. Breuer; Maurizio Affer; Sten Andréasson; David G. Birch; Gerald A. Fishman; John R. Heckenlively; Suja Hiriyanna; Dennis R. Hoffman; Samuel G. Jacobson; Alan J. Mears; Maria A. Musarella; Elena Redolfi; Paul A. Sieving; Alan F. Wright; Beverly M. Yashar; Ileana Zucchi; Anand Swaroop

Retinitis pigmentosa (RP) is a clinically and genetically heterogeneous group of retinal degenerative diseases, characterized by nightblindness, progressive restriction of the visual field and pigmentary retinopathy.1 At least 28 different genetic loci have been mapped for autosomal dominant, autosomal recessive, and X-linked forms of RP. [http://www.sph.uth.tmc.edu/Retnet/home.htm] The X-linked RP (XLRP) subtype is the most severe, with an early age of onset and more rapid progression, accounting for 10 to 20% of RP families.2,3 XLRP is also genetically heterogeneous with at least 5 mapped loci: RP2, RP3, RP6, RP23 and RP24, as schematically depicted in Figure 1. By linkage analysis, RP2 is predicted to account for 10–20% of XLRP and RP3 for 70–90%,4–6 depending on the population. Genes for these two major loci have now been cloned. Our laboratory has been involved in the mutational screening and functional analysis of the two identified XLRP genes (RPGR and RP2), as well as the positional cloning of two other XLRP loci (RP6 and RP24). This report summarizes these efforts as well as the current standing of XLRP research.


Molecular Therapy | 2016

266. In Vivo Rod Photoreceptor Reprogramming Using AAV-Delivered CRISPR/Cas9 Rescues Retinal Degeneration

Wenhan Yu; Suddhasil Mookherjee; Jung-Woong Kim; Suja Hiriyanna; Yasaman Ataeijannati; Xun Sun; Lijin Dong; Tiansen Li; Anand Swaroop; Zhijian Wu

Retinitis pigmentosa (RP) is the most common form of inherited retinal dystrophy and the leading cause of inherited blindness, due to mutations in any of the over 60 genes/loci identified so far. The disease is characterized by an initial loss of rod photoreceptors and secondary cone cell death. Since cone photoreceptors are responsible for day time vision and visual acuity, preserving cone functions in RP patients is a priority when developing treatment strategies. NRL is a transcription factor that determines the rod photoreceptor cell fate during retinal development. Acute gene knockout of Nrl in mice was shown to reprogram adult rods into cone-like cells, rendering them resistant to effects of mutations in rod-specific genes and consequently preventing secondary cone loss (Montana CL, et al. PNAS, 2013; 110: 1732-7). With a goal to develop this approach for treatment of RP, we used adeno-associated virus (AAV)-delivered CRISPR/Cas9 for Nrl-knockdown in rod photoreceptors. AAV vectors were constructed to carry a photoreceptor-specific Cas9 nuclease expression cassette or a single-guided RNA (sgRNA) targeting Nrl or eGFP gene. The Cas9 and the sgRNA vectors were co-delivered into mice by subretinal administration. Potency of the AAV-CRISPR/Cas9 system was validated by EGFP knockdown in a mouse line with eGFP-labeled rods. Nrl knockdown was conducted in wild-type C57/Bl6 or Crxp-Nrl, a mouse line with rod-only photoreceptors. Molecular, histological and functional alterations were examined by next generation sequencing, immunoblot analysis, immunofluorescence, electron microscopy, and electroretinography (ERG). Our results showed that eGFP and Nrl were efficiently knocked down following AAV-CRISPR/Cas9 treatment. For Nrl knockdown, almost all insertions and deletions were detected in the targeted Nrl locus, and very few mutations were identified in ten potential off-target loci. A majority of the transduced rods acquired characteristics of cone photoreceptors following Nrl-CRISPR/Cas9 vector treatment, as demonstrated by reduced expression of rod-specific genes and enhanced expression of cone-specific genes, loss of the unique rod chromatin pattern, and diminished rod ERG response. Rescue of retinal degeneration was assessed in three mouse models harboring either recessive or dominant rod-specific mutations. In all three models, the Nrl-CRISPR/Cas9 vector treated eyes maintained significantly better photoreceptor viability and cone function than control eyes, as revealed by remarkably thicker photoreceptor layer, higher cone cell number, greater cone ERG amplitude and better optomotor behavior. In conclusion, AAV-CRISPR-mediated Nrl gene knockdown can efficiently reprogram rods into cone-like photoreceptors and prevent secondary cone death in retinal degeneration, which could be developed into a viable treatment for RP in humans.


Investigative Ophthalmology & Visual Science | 2002

Microarray Analysis of Gene Expression in the Aging Human Retina

Shigeo Yoshida; Beverly M. Yashar; Suja Hiriyanna; Anand Swaroop


Experimental Eye Research | 2002

Retinal histopathology of an XLRP carrier with a mutation in the RPGR exon ORF15.

Gustavo D. Aguirre; Beverly M. Yashar; Sinoj K John; Julie E. Smith; Debra K. Breuer; Suja Hiriyanna; Anand Swaroop; Ann H. Milam


American Journal of Ophthalmology | 2004

Clinical and immunohistochemical evidence for an X linked retinitis pigmentosa syndrome with recurrent infections and hearing loss in association with an RPGR mutation

Alessandro Iannaccone; Debra K. Breuer; Xinjing Wang; Sharon F. Kuo; E M Normando; E. Filippova; Alfonso Baldi; Suja Hiriyanna; MacDonald Cb; F Baldi; Dominic Cosgrove; Cynthia C. Morton; Anand Swaroop; Monica M. Jablonski

Collaboration


Dive into the Suja Hiriyanna's collaboration.

Top Co-Authors

Avatar

Anand Swaroop

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Zhijian Wu

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Suddhasil Mookherjee

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Tiansen Li

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Paul A. Sieving

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Peter Colosi

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge