Network


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

Hotspot


Dive into the research topics where Vidyullatha Vasireddy is active.

Publication


Featured researches published by Vidyullatha Vasireddy.


PLOS ONE | 2013

AAV-Mediated Gene Therapy for Choroideremia: Preclinical Studies in Personalized Models

Vidyullatha Vasireddy; Jason A. Mills; Rajashekhar Gaddameedi; Etiena Basner-Tschakarjan; Monika Köhnke; Aaron D. Black; Krill Alexandrov; Shangzhen Zhou; Albert M. Maguire; Daniel C. Chung; Helen Mac; Lisa M. Sullivan; Paul Gadue; Jeannette L. Bennicelli; Deborah L. French; Jean Bennett

Choroideremia (CHM) is an X- linked retinal degeneration that is symptomatic in the 1st or 2nd decade of life causing nyctalopia and loss of peripheral vision. The disease progresses through mid-life, when most patients become blind. CHM is a favorable target for gene augmentation therapy, as the disease is due to loss of function of a protein necessary for retinal cell health, Rab Escort Protein 1 (REP1).The CHM cDNA can be packaged in recombinant adeno-associated virus (rAAV), which has an established track record in human gene therapy studies, and, in addition, there are sensitive and quantitative assays to document REP1 activity. An animal model that accurately reflects the human condition is not available. In this study, we tested the ability to restore REP1 function in personalized in vitro models of CHM: lymphoblasts and induced pluripotent stems cells (iPSCs) from human patients. The initial step of evaluating safety of the treatment was carried out by evaluating for acute retinal histopathologic effects in normal-sighted mice and no obvious toxicity was identified. Delivery of the CHM cDNA to affected cells restores REP1 enzymatic activity and also restores proper protein trafficking. The gene transfer is efficient and the preliminary safety data are encouraging. These studies pave the way for a human clinical trial of gene therapy for CHM.


PLOS ONE | 2011

Rescue of photoreceptor degeneration by curcumin in transgenic rats with P23H rhodopsin mutation.

Vidyullatha Vasireddy; Venkata Ramana Murthy Chavali; Victory Joseph; Rajendra S. Kadam; Jonathan H. Lin; Jeffrey A. Jamison; Uday B. Kompella; Geereddy Bhanuprakash Reddy; Radha Ayyagari

The P23H mutation in the rhodopsin gene causes rhodopsin misfolding, altered trafficking and formation of insoluble aggregates leading to photoreceptor degeneration and autosomal dominant retinitis pigmentosa (RP). There are no effective therapies to treat this condition. Compounds that enhance dissociation of protein aggregates may be of value in developing new treatments for such diseases. Anti-protein aggregating activity of curcumin has been reported earlier. In this study we present that treatment of COS-7 cells expressing mutant rhodopsin with curcumin results in dissociation of mutant protein aggregates and decreases endoplasmic reticulum stress. Furthermore we demonstrate that administration of curcumin to P23H-rhodopsin transgenic rats improves retinal morphology, physiology, gene expression and localization of rhodopsin. Our findings indicate that supplementation of curcumin improves retinal structure and function in P23H-rhodopsin transgenic rats. This data also suggest that curcumin may serve as a potential therapeutic agent in treating RP due to the P23H rhodopsin mutation and perhaps other degenerative diseases caused by protein trafficking defects.


Progress in Retinal and Eye Research | 2010

Genetics and molecular pathology of Stargardt-like macular degeneration

Vidyullatha Vasireddy; Paul Wong; Radha Ayyagari

Stargardt-like macular degeneration (STGD3) is an early onset, autosomal dominant macular degeneration. STGD3 is characterized by a progressive pathology, the loss of central vision, atrophy of the retinal pigment epithelium, and accumulation of lipofuscin, clinical features that are also characteristic of age-related macular degeneration. The onset of clinical symptoms in STGD3, however, is typically observed within the second or third decade of life (i.e., starting in the teenage years). The clinical profile at any given age among STGD3 patients can be variable suggesting that, although STGD3 is a single gene defect, other genetic or environmental factors may play a role in moderating the final disease phenotype. Genetic studies localized the STGD3 disease locus to a small region on the short arm of human chromosome 6, and application of a positional candidate gene approach identified protein truncating mutations in the elongation of very long chain fatty acids-4 gene (ELOVL4) in patients with this disease. The ELOVL4 gene encodes a protein homologous to the ELO group of proteins that participate in fatty acid elongation in yeast. Pathogenic mutations found in the ELOVL4 gene result in altered trafficking of the protein and behave with a dominant negative effect. Mice carrying an Elovl4 mutation developed photoreceptor degeneration and depletion of very long chain fatty acids (VLCFA). ELOVL4 protein participates in the synthesis of fatty acids with chain length longer than 26 carbons. Studies on ELOVL4 indicate that VLCFA may be necessary for normal function of the retina, and the defective protein trafficking and/or altered VLCFA elongation underlies the pathology associated with STGD3. Determining the role of VLCFA in the retina and discerning the implications of abnormal trafficking of mutant ELOVL4 and depleted VLCFA content in the pathology of STGD3 will provide valuable insight in understanding the retinal structure, function, and pathology underlying STGD3 and may lead to a better understanding of the process of macular disease in general.


Experimental Eye Research | 2009

Elovl4 5-bp deletion knock-in mouse model for Stargardt-like macular degeneration demonstrates accumulation of ELOVL4 and lipofuscin

Vidyullatha Vasireddy; Monica M. Jablonski; Naheed W. Khan; Xiao Fei Wang; Priya Sahu; Janet R. Sparrow; Radha Ayyagari

The mechanism underlying photoreceptor degeneration in autosomal dominant Stargardt-like macular degeneration (STGD3) due to mutations in the elongation of very long chain fatty acids-4 (ELOVL4) gene is not fully understood. To evaluate the pathological events associated with STGD3, we used a mouse model that mimics the human STGD3 phenotype and studied the progression of retinal degeneration. Morphological changes in the retina of Elovl4 5-bp deletion knock-in mice (E_mut(+/-)) were evaluated at 22 months of age. The localization of ELOVL4, and the expression pattern of inner retinal tissue marker proteins, and ubiquitin were determined by immunofluorescence labeling of retinal sections. Levels of the retinal pigment epithelium (RPE) lipofuscin fluorophores were measured by quantitative HPLC. Morphological evaluation of the retina revealed an accumulation of RPE debris in the subretinal space. A significant increase in the amount of ELOVL4 was observed in the outer plexiform layer in E_mut(+/-) mice compared to controls. Apart from the accumulation of ELOVL4, E_mut(+/-) mice also exhibited high expression of ubiquitin in the retina. Analysis of lipofuscin fluorophores in the RPE showed a significant elevation of A2E and compounds of the all-trans-retinal dimer series in retinas from four and ten month old E_mut(+/-) mice compared to wild-type littermates. These observations suggest that abnormal accumulation of ELOVL4 protein and lipofuscin may lead to photoreceptor degeneration in E_mut(+/-) mice.


Investigative Ophthalmology & Visual Science | 2009

A Novel Rat Model with Obesity-Associated Retinal Degeneration

Geereddy Bhanuprakash Reddy; Vidyullatha Vasireddy; Nawajes A. Mandal; Mrudula Tiruvalluru; Xiao Fei Wang; Monica M. Jablonski; Giridharan Nappanveettil; Radha Ayyagari

PURPOSE A strong association between retinal degeneration and obesity has been shown in humans. However, the molecular basis of increased risk for retinal degeneration in obesity is unknown. Thus, an animal model with obesity and retinal degeneration would greatly aid the understanding of obesity-associated retinal degeneration. The retinal abnormalities in a novel rat model (WNIN-Ob) with spontaneously developed obesity are described. METHODS Histologic and immunohistochemical examination were performed on retinal sections of 2- to 12-month-old WNIN-Ob rats, and findings were compared with those of lean littermate controls. RNA from retinas of 12-month-old WNIN-Ob and lean littermate rats was used for microarray and qRT-PCR analysis. RESULTS The WNIN-Ob rats developed severe obesity, with an onset at approximately 35 days. Evaluation of retinal morphology in 2- to 12-month-old WNIN-Ob and age-matched lean littermate controls revealed progressive retinal degeneration, with an onset between 4 to 6 months of age. Immunohistochemical analysis with anti-rhodopsin, anti-cone opsin, and PSD-95 antibodies further confirmed retinal degeneration, particularly rod cell loss and thinner outer plexiform layer, in the obese rat retina. Gene expression by microarray analysis and qRT-PCR established activation of stress response, tissue remodeling, impaired phototransduction, and photoreceptor degeneration in WNIN-Ob rat retina. CONCLUSIONS WNIN-Ob rats develop increased stress in retinal tissue and progressive retinal degeneration after the onset of severe obesity. The WNIN-Ob rat is the first rat model to develop retinal degeneration after the onset of obesity. This novel rat model may be a valuable tool for investigating retinal degeneration associated with obesity in humans.


Journal of Gene Medicine | 2014

Adeno-associated virus 8-mediated gene therapy for choroideremia: Preclinical studies in in vitro and in vivo models

Aaron D. Black; Vidyullatha Vasireddy; Daniel C. Chung; Albert M. Maguire; Rajashekhar Gaddameedi; Tania Tolmachova; Miguel C. Seabra; Jean Bennett

Choroideremia (CHM) is a slowly progressive X‐linked retinal degeneration that results in a loss of photoreceptors, retinal pigment epithelium and choroid. CHM, the gene implicated in choroideremia, encodes Rab escort protein‐1 (REP‐1), which is involved in the post‐translational activation via prenylation of Rab proteins.


Scientific Reports | 2015

An Autogenously Regulated Expression System for Gene Therapeutic Ocular Applications

Matthew Sochor; Vidyullatha Vasireddy; Theodore G. Drivas; Adam Wojno; Thu Doung; Ivan Shpylchak; Jeannette L. Bennicelli; Daniel C. Chung; Jean Bennett; Mitchell Lewis

The future of treating inherited and acquired genetic diseases will be defined by our ability to introduce transgenes into cells and restore normal physiology. Here we describe an autogenous transgene regulatory system (ARES), based on the bacterial lac repressor, and demonstrate its utility for controlling the expression of a transgene in bacteria, eukaryotic cells, and in the retina of mice. This ARES system is inducible by the small non-pharmacologic molecule, Isopropyl β-D-1-thiogalactopyranoside (IPTG) that has no off-target effects in mammals. Following subretinal injection of an adeno-associated virus (AAV) vector encoding ARES, luciferase expression can be reversibly controlled in the murine retina by oral delivery of IPTG over three induction-repression cycles. The ability to induce transgene expression repeatedly via administration of an oral inducer in vivo, suggests that this type of regulatory system holds great promise for applications in human gene therapy.


Stem Cell Research | 2018

Use of induced pluripotent stem cell models to probe the pathogenesis of Choroideremia and to develop a potential treatment

Thu T. Duong; Vidyullatha Vasireddy; Pamela S. Herrera; Lanfranco Leo; Carrie Merkel; Jean Bennett; Jason A. Mills

Choroideremia (CHM) is a rare monogenic, X-linked recessive inherited retinal degeneration resulting from mutations in the Rab Escort Protein-1 (REP1) encoding CHM gene. The primary retinal cell type leading to CHM is unknown. In this study, we explored the utility of induced pluripotent stem cell-derived models of retinal pigmented epithelium (iPSC-RPE) to study disease pathogenesis and a potential gene-based intervention in four different genetically distinct forms of CHM. A number of abnormal cell biologic, biochemical, and physiologic functions were identified in the CHM mutant cells. We then identified a recombinant adeno-associated virus (AAV) serotype, AAV7m8, that is optimal for both delivering transgenes to iPSC-RPEs as well as to appropriate target cells (RPE cells and rod photoreceptors) in the primate retina. To establish the proof of concept of AAV7m8 mediated CHM gene therapy, we developed AAV7m8.hCHM, which delivers the human CHM cDNA under control of CMV-enhanced chicken β-actin promoter (CßA). Delivery of AAV7m8.hCHM to CHM iPSC-RPEs restored protein prenylation, trafficking and phagocytosis. The results confirm that AAV-mediated delivery of the REP1-encoding gene can rescue defects in CHM iPSC-RPE regardless of the type of disease-causing mutation. The results also extend our understanding of mechanisms involved in the pathophysiology of choroideremia.


Advances in Experimental Medicine and Biology | 2012

Silencing the Expression of CTRP5/C1QTNF5 and ELOVL4 Genes by Small Interfering RNA

Venkata Ramana Murthy Chavali; Vidyullatha Vasireddy; Radha Ayyagari

Two monogenic macular degenerations, the stargardt-like macular degeneration (STGD3) and the late-onset retinal dystrophy (L-ORD), are caused due to mutations in the genes elongation of very long chain fatty acid-4 (ELOVL4) and C1Q tumor necrosis factor-related protein-5 (CTRP5/C1QTNF5) genes, respectively. It has been shown that disease-causing mutations alter the trafficking of these proteins and exert dominant-negative effect. Selective elimination of these mutant proteins may facilitate restoring the normal function of retinal tissue. In this study, we describe characterization of small interfering RNA (siRNA) probes that selectively and effectively knock down the expression of the wild-type and mutant ELOVL4 and the wild-type CTRP5 genes in Cos-7 cells. These probes will be valuable in studying the function of wild-type ELOVL4 and CTRP5 genes, affect of mutant proteins and the potential use of gene silencing in treating STGD3 and L-ORD.


PLOS ONE | 2015

Correction: AAV-Mediated Gene Therapy for Choroideremia: Preclinical Studies in Personalized Models

Vidyullatha Vasireddy; Jason A. Mills; Rajashekhar Gaddameedi; Etiena Basner-Tschakarjan; Monika Köhnke; Aaron D. Black; Krill Alexandrov; Shangzhen Zhou; Albert M. Maguire; Daniel C. Chung; Helen Mac; Lisa M. Sullivan; Paul Gadue; Jeannette L. Bennicelli; Deborah L. French; Jean Bennett

Choroideremia (CHM) is an X- linked retinal degeneration that is symptomatic in the 1st or 2nd decade of life causing nyctalopia and loss of peripheral vision. The disease progresses through mid-life, when most patients become blind. CHM is a favorable target for gene augmentation therapy, as the disease is due to loss of function of a protein necessary for retinal cell health, Rab Escort Protein 1 (REP1).The CHM cDNA can be packaged in recombinant adeno-associated virus (rAAV), which has an established track record in human gene therapy studies, and, in addition, there are sensitive and quantitative assays to document REP1 activity. An animal model that accurately reflects the human condition is not available. In this study, we tested the ability to restore REP1 function in personalized in vitro models of CHM: lymphoblasts and induced pluripotent stems cells (iPSCs) from human patients. The initial step of evaluating safety of the treatment was carried out by evaluating for acute retinal histopathologic effects in normal-sighted mice and no obvious toxicity was identified. Delivery of the CHM cDNA to affected cells restores REP1 enzymatic activity and also restores proper protein trafficking. The gene transfer is efficient and the preliminary safety data are encouraging. These studies pave the way for a human clinical trial of gene therapy for CHM.

Collaboration


Dive into the Vidyullatha Vasireddy's collaboration.

Top Co-Authors

Avatar

Radha Ayyagari

University of California

View shared research outputs
Top Co-Authors

Avatar

Jean Bennett

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Monica M. Jablonski

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nawajes A. Mandal

University of Oklahoma Health Sciences Center

View shared research outputs
Top Co-Authors

Avatar

Xiao Fei Wang

University of Tennessee Health Science Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jason A. Mills

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar

Paul A. Sieving

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Albert M. Maguire

University of Pennsylvania

View shared research outputs
Researchain Logo
Decentralizing Knowledge