Network


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

Hotspot


Dive into the research topics where Erin R. Burnight is active.

Publication


Featured researches published by Erin R. Burnight.


Journal of Virology | 2005

Persistent Gene Expression in Mouse Nasal Epithelia following Feline Immunodeficiency Virus-Based Vector Gene Transfer

Patrick L. Sinn; Erin R. Burnight; Melissa A. Hickey; Gary W. Blissard; Paul B. McCray

ABSTRACT Gene transfer development for treatment or prevention of cystic fibrosis lung disease has been limited by the inability of vectors to efficiently and persistently transduce airway epithelia. Influenza A is an enveloped virus with natural lung tropism; however, pseudotyping feline immunodeficiency virus (FIV)-based lentiviral vector with the hemagglutinin envelope protein proved unsuccessful. Conversely, pseudotyping FIV with the envelope protein from influenza D (Thogoto virus GP75) resulted in titers of 106 transducing units (TU)/ml and conferred apical entry into well-differentiated human airway epithelial cells. Baculovirus GP64 envelope glycoproteins share sequence identity with influenza D GP75 envelope glycoproteins. Pseudotyping FIV with GP64 from three species of baculovirus resulted in titers of 107 to 109 TU/ml. Of note, GP64 from Autographa californica multicapsid nucleopolyhedrovirus resulted in high-titer FIV preparations (∼109 TU/ml) and conferred apical entry into polarized primary cultures of human airway epithelia. Using a luciferase reporter gene and bioluminescence imaging, we observed persistent gene expression from in vivo gene transfer in the mouse nose with A. californica GP64-pseudotyped FIV (AcGP64-FIV). Longitudinal bioluminescence analysis documented persistent expression in nasal epithelia for ∼1 year without significant decline. According to histological analysis using a LacZ reporter gene, olfactory and respiratory epithelial cells were transduced. In addition, methylcellulose-formulated AcGP64-FIV transduced mouse nasal epithelia with much greater efficiency than similarly formulated vesicular stomatitis virus glycoprotein-pseudotyped FIV. These data suggest that AcGP64-FIV efficiently transduces and persistently expresses a transgene in nasal epithelia in the absence of agents that disrupt the cellular tight junction integrity.


Proceedings of the National Academy of Sciences of the United States of America | 2013

piggyBac transposase tools for genome engineering

Xianghong Li; Erin R. Burnight; Ashley L. Cooney; Nirav Malani; Troy Brady; Jeffry D. Sander; Janice M. Staber; Sarah J. Wheelan; J. Keith Joung; Paul B. McCray; Frederic D. Bushman; Patrick L. Sinn; Nancy L. Craig

Significance DNA transposons that translocate by excision from a donor site and insertion into a target site are often used for genome engineering by insertional mutagenesis and transgenesis. The piggyBac element is especially useful because it can excise precisely from an insertion site, restoring the site to its pretransposon state. Precise excision is particularly useful when transient transgenesis is needed, for example, in the transient introduction of transcription factors for induced pluripotent stem cell production. We have used mutagenesis to generate an Excision+ Integration− transposase that allows piggyBac excision without potentially harmful reintegration. These mutations likely lie in a target DNA-binding domain. The transposon piggyBac is being used increasingly for genetic studies. Here, we describe modified versions of piggyBac transposase that have potentially wide-ranging applications, such as reversible transgenesis and modified targeting of insertions. piggyBac is distinguished by its ability to excise precisely, restoring the donor site to its pretransposon state. This characteristic makes piggyBac useful for reversible transgenesis, a potentially valuable feature when generating induced pluripotent stem cells without permanent alterations to genomic sequence. To avoid further genome modification following piggyBac excision by reintegration, we generated an excision competent/integration defective (Exc+Int−) transposase. Our findings also suggest the position of a target DNA–transposase interaction. Another goal of genome engineering is to develop reagents that can guide transgenes to preferred genomic regions. Others have shown that piggyBac transposase can be active when fused to a heterologous DNA-binding domain. An Exc+Int− transposase, the intrinsic targeting of which is defective, might also be a useful intermediate in generating a transposase whose integration activity could be rescued and redirected by fusion to a site-specific DNA-binding domain. We show that fusion to two designed zinc finger proteins rescued the Int− phenotype. Successful guided transgene integration into genomic DNA would have broad applications to gene therapy and molecular genetics. Thus, an Exc+Int− transposase is a potentially useful reagent for genome engineering and provides insight into the mechanism of transposase–target DNA interaction.


Progress in Retinal and Eye Research | 2015

Patient-specific induced pluripotent stem cells (iPSCs) for the study and treatment of retinal degenerative diseases.

Luke A. Wiley; Erin R. Burnight; Allison E. Songstad; Arlene V. Drack; Robert F. Mullins; Edwin M. Stone; Budd A. Tucker

Vision is the sense that we use to navigate the world around us. Thus it is not surprising that blindness is one of peoples most feared maladies. Heritable diseases of the retina, such as age-related macular degeneration and retinitis pigmentosa, are the leading cause of blindness in the developed world, collectively affecting as many as one-third of all people over the age of 75, to some degree. For decades, scientists have dreamed of preventing vision loss or of restoring the vision of patients affected with retinal degeneration through drug therapy, gene augmentation or a cell-based transplantation approach. In this review we will discuss the use of the induced pluripotent stem cell technology to model and develop various treatment modalities for the treatment of inherited retinal degenerative disease. We will focus on the use of iPSCs for interrogation of disease pathophysiology, analysis of drug and gene therapeutics and as a source of autologous cells for cell transplantation and replacement.


Scientific Reports | 2016

cGMP production of patient-specific iPSCs and photoreceptor precursor cells to treat retinal degenerative blindness

Luke A. Wiley; Erin R. Burnight; Adam P. DeLuca; Kristin R. Anfinson; Cathryn M. Cranston; Emily E. Kaalberg; Jessica A. Penticoff; Louisa M. Affatigato; Robert F. Mullins; Edwin M. Stone; Budd A. Tucker

Immunologically-matched, induced pluripotent stem cell (iPSC)-derived photoreceptor precursor cells have the potential to restore vision to patients with retinal degenerative diseases like retinitis pigmentosa. The purpose of this study was to develop clinically-compatible methods for manufacturing photoreceptor precursor cells from adult skin in a non-profit cGMP environment. Biopsies were obtained from 35 adult patients with inherited retinal degeneration and fibroblast lines were established under ISO class 5 cGMP conditions. Patient-specific iPSCs were then generated, clonally expanded and validated. Post-mitotic photoreceptor precursor cells were generated using a stepwise cGMP-compliant 3D differentiation protocol. The recapitulation of the enhanced S-cone phenotype in retinal organoids generated from a patient with NR2E3 mutations demonstrated the fidelity of these protocols. Transplantation into immune compromised animals revealed no evidence of abnormal proliferation or tumor formation. These studies will enable clinical trials to test the safety and efficiency of patient-specific photoreceptor cell replacement in humans.


Molecular therapy. Nucleic acids | 2012

A Hyperactive Transposase Promotes Persistent Gene Transfer of a piggyBac DNA Transposon

Erin R. Burnight; Janice M. Staber; Pavel Korsakov; Xianghong Li; Benjamin T. Brett; Todd E. Scheetz; Nancy L. Craig; Paul B. McCray

Nonviral vector systems are used increasingly in gene targeting and gene transfer applications. The piggyBac transposon represents an alternative integrating vector for in vivo gene transfer. We hypothesized that this system could achieve persistent gene transfer to the liver when administered systemically. We report that a novel hyperactive transposase generated higher transposition efficiency than a codon-optimized transposase in a human liver cell line. Hyperactive transposase-mediated reporter gene expression persisted at levels twice that of codon-optimized transposase in the livers of mice for the 6-month study. Of note, expression persisted in mice following partial hepatectomy, consistent with expression from an integrated transgene. We also used the hyperactive transposase to deliver the human α1-antitrypsin gene and achieved stable expression in serum. To determine the integration pattern of insertions, we performed large-scale mapping in human cells and recovered 60,685 unique hyperactive transposase-mediated insertions. We found that a hyperactive piggyBac transposase conferred an altered pattern of integration from that of insect piggyBac transposase, with a decreased frequency of integration near transcription start sites than previously reported. Our results support that the piggyBac transposon combined with the hyperactive transposase is an efficient integrating vector system for in vitro and in vivo applications.


Gene Therapy | 2009

Progress and prospects: prospects of repeated pulmonary administration of viral vectors.

Patrick L. Sinn; Erin R. Burnight; Paul B. McCray

Pulmonary gene therapy may ultimately cure diseases such as cystic fibrosis, α1-antitrypsin deficiency, lung cancer and pulmonary hypertension. Efficient expression of delivered genes in target cell types is essential for the achievement of this goal. To this end, re-administration of viral vectors may be required (1) to increase the percentage of transduced airway epithelial cells, (2) to direct gene transfer to individual lobes during successive delivery sessions or (3) to boost attenuated expression over time. Immune responses to viral proteins or viral-encoded proteins are the greatest barrier to repeated vector administration.


Stem Cells Translational Medicine | 2016

Concise Review: Patient-Specific Stem Cells to Interrogate Inherited Eye Disease

Joseph C. Giacalone; Luke A. Wiley; Erin R. Burnight; Allison E. Songstad; Robert F. Mullins; Edwin M. Stone; Budd A. Tucker

Whether we are driving to work or spending time with loved ones, we depend on our sense of vision to interact with the world around us. Therefore, it is understandable why blindness for many is feared above death itself. Heritable diseases of the retina, such as glaucoma, age‐related macular degeneration, and retinitis pigmentosa, are major causes of blindness worldwide. The recent success of gene augmentation trials for the treatment of RPE65‐associated Leber congenital amaurosis has underscored the need for model systems that accurately recapitulate disease. With the advent of patient‐specific induced pluripotent stem cells (iPSCs), researchers are now able to obtain disease‐specific cell types that would otherwise be unavailable for molecular analysis. In the present review, we discuss how the iPSC technology is being used to confirm the pathogenesis of novel genetic variants, interrogate the pathophysiology of disease, and accelerate the development of patient‐centered treatments.


Molecular Therapy | 2015

687. Therapeutic Correction of an LCA-Causing Splice Defect in the CEP290 Gene by CRISPR/Cas-Mediated Genome Editing

Morgan L. Maeder; Shen Shen; Erin R. Burnight; Sebastian Gloskowski; Rina Mepani; Ari E. Friedland; Hari Jayaram; Greg Gotta; Budd A. Tucker; David Bumcrot

Leber congenital amaurosis (LCA) comprises a genetically heterogeneous group of early-onset retinal disorders characterized by severe loss of vision in the first years of life. Approximately 30% of LCA patients harbor mutations in the CEP290 gene, which codes for a centrosomal protein involved in ciliogenesis and ciliary trafficking in the photoreceptors. While AAV-mediated gene therapy is showing promise in clinical trials for RPE65-associated LCA, the size of CEP290 exceeds the limit for efficient packaging in AAV. Thus, alternative approaches to treat CEP290 associated LCA are needed. Here, we report a gene editing approach in which the CRISPR/Cas9 system is used to modify the endogenous CEP290 locus and restore normal function of the gene.The most common mutation in CEP290 is the IVS26 c. 2991+1655 A>G mutation. This point mutation in intron 26 of the gene generates a novel splice donor, resulting in aberrant splicing and the inclusion of an extra 128bp cryptic exon into the coding sequence of the transcript. Using the S. aureus CRISPR/Cas9 system, we employed a dual-cut approach in which two gRNAs are used to induce a pair of double strand breaks and excise the region of DNA containing the mutation. We screened gRNA pairs in primary fibroblasts derived from LCA patients harboring homozygous IVS26 mutations and identified pairs capable of generating targeted deletions with high efficiency. Using qRT-PCR, we showed that deletion of the mutation results in increased expression of wildtype CEP290 and concomitant decrease in expression of the aberrantly spliced mutant RNA species. Genome-wide specificity profiling allows for assessment of potential off-target modifications induced by the gRNAs and informs selection of optimal gRNA pairs for further study. Assays to assess ciliogenesis, as well as molecular characterization of gene editing in patient-derived photoreceptor precursor cells, demonstrate phenotypic correction in the relevant cell population. The use of the S. aureus CRISPR/Cas9 system enables efficient packaging of the Cas9 gene, as well as two gRNA genes, into a single AAV vector and provides a method for delivery of this system into patient photoreceptors.


Cold Spring Harbor Perspectives in Medicine | 2015

Stem Cells as Tools for Studying the Genetics of Inherited Retinal Degenerations

Luke A. Wiley; Erin R. Burnight; Robert F. Mullins; Edwin M. Stone; Budd A. Tucker

The ability to provide early clinical intervention for inherited disorders is heavily dependent on knowledge of a patients disease-causing mutations and the resultant pathophysiologic mechanism(s). Without knowing a patients disease-causing gene, and how gene mutations alter the health and functionality of affected cells, it would be difficult to develop and deliver patient-specific molecular or small molecule therapies. Many believe that the field of stem cell biology holds the keys to the future development of disease-, patient-, and cell-specific therapies. In the case of the eye, which is susceptible to an extremely common late-onset degenerative disease known as age-related macular degeneration, stem cell-based therapies could increase the quality of life for millions of patients worldwide. Furthermore, autologous, patient-specific induced pluripotent stem cells could be a viable source to treat rare Mendelian retinal degenerative diseases such as retinitis pigmentosa, Stargardt disease, and Best disease, to name a few.


Cold Spring Harbor Perspectives in Medicine | 2015

Gene Therapy Using Stem Cells

Erin R. Burnight; Luke A. Wiley; Robert F. Mullins; Edwin M. Stone; Budd A. Tucker

Viral-mediated gene augmentation therapy has recently shown success in restoring vision to patients with retinal degenerative disorders. Key to this success was the availability of animal models that accurately presented the human phenotype to test preclinical efficacy and safety. These exciting studies support the use of gene therapy in the treatment of devastating retinal degenerative diseases. In some cases, however, in vivo gene therapy for retinal degeneration would not be effective because the cell types targeted are no longer present. The development of somatic cell reprogramming methods provides an attractive source of autologous cells for transplantation and treatment of retinal degenerative disease. This article explores the development of gene therapy and patient-derived stem cells for the purpose of restoring vision to individuals suffering from inherited retinal degenerations.

Collaboration


Dive into the Erin R. Burnight's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Joseph C. Giacalone

Roy J. and Lucille A. Carver College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Patrick L. Sinn

Roy J. and Lucille A. Carver College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Emily E. Kaalberg

Roy J. and Lucille A. Carver College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Kristin R. Anfinson

Roy J. and Lucille A. Carver College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge