Network


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

Hotspot


Dive into the research topics where Daniel J. Bogan is active.

Publication


Featured researches published by Daniel J. Bogan.


Nature Biotechnology | 2000

In vivo targeted repair of a point mutation in the canine dystrophin gene by a chimeric RNA/DNA oligonucleotide

R. J. Bartlett; Sabine Stockinger; Melvin Denis; William Travis Bartlett; Luca Inverardi; T.T. Le; Nguyen thi Man; Glenn E. Morris; Daniel J. Bogan; Janet Metcalf-Bogan; Joe N. Kornegay

In the canine model of Duchenne muscular dystrophy in golden retrievers (GRMD), a point mutation within the splice acceptor site of intron 6 leads to deletion of exon 7 from the dystrophin mRNA, and the consequent frameshift causes early termination of translation. We have designed a DNA and RNA chimeric oligonucleotide to induce host cell mismatch repair mechanisms and correct the chromosomal mutation to wild type. Direct skeletal muscle injection of the chimeric oligonucleotide into the cranial tibialis compartment of a six-week-old affected male dog, and subsequent analysis of biopsy and necropsy samples, demonstrated in vivo repair of the GRMD mutation that was sustained for 48 weeks. Reverse transcription–polymerase chain reaction (RT-PCR) analysis of exons 5–10 demonstrated increasing levels of exon 7 inclusion with time. An isolated exon 7-specific dystrophin antibody confirmed synthesis of normal-sized dystrophin product and positive localization to the sarcolemma. Chromosomal repair in muscle tissue was confirmed by restriction fragment length polymorphism (RFLP)–PCR and sequencing the PCR product. This work provides evidence for the long-term repair of a specific dystrophin point mutation in muscle of a live animal using a chimeric oligonucleotide.


Molecular Therapy | 2010

Widespread Muscle Expression of an AAV9 Human Mini-dystrophin Vector After Intravenous Injection in Neonatal Dystrophin-deficient Dogs

Joe N. Kornegay; Juan Li; Janet R. Bogan; Daniel J. Bogan; Chunlian Chen; Hui Zheng; Bing Wang; Chunping Qiao; James F. Howard; Xiao Xiao

Duchenne (DMD) and golden retriever (GRMD) muscular dystrophy are caused by genetic mutations in the dystrophin gene and afflict striated muscles. We investigated systemic gene delivery in 4-day-old GRMD dogs given a single intravenous injection of an AAV9 vector (1.5 x 10(14) vector genomes/kg) carrying a human codon-optimized human mini-dystrophin gene under control of the cytomegalovirus (CMV) promoter. One of the three treated dogs was euthanized 9 days later due to pre-existing conditions. Scattered mini-dystrophin-positive myofibers were seen by immunofluorescent (IF) staining in numerous muscles. At the end of the 16-week study, the other two dogs showed generalized muscle expression of mini-dystrophin in ~15% to nearly 100% of myofibers. Western blot and vector DNA quantitative PCR results agreed with the IF data. Delayed growth and pelvic limb muscle atrophy and contractures were seen several weeks after vector delivery. T-2 weighted magnetic resonance imaging (MRI) at 8 weeks showed increased signal intensity compatible with inflammation in several pelvic limb muscles. This marked early inflammatory response raised concerns regarding methodology. Use of the ubiquitous CMV promoter, extra-high vector dose, and marked expression of a human protein in canine muscles may have contributed to the pathologic changes seen in the pelvic limbs.


Mammalian Genome | 2012

Canine Models of Duchenne Muscular Dystrophy and Their Use in Therapeutic Strategies

Joe N. Kornegay; Janet R. Bogan; Daniel J. Bogan; Martin K. Childers; Juan Li; Peter P. Nghiem; David A. Detwiler; C. Aaron Larsen; Robert W. Grange; Ratna K. Bhavaraju-Sanka; Sandra P. Tou; Bruce P. Keene; James F. Howard; Jiahui Wang; Zheng Fan; Scott J. Schatzberg; Martin Styner; Kevin M. Flanigan; Xiao Xiao; Eric P. Hoffman

Duchenne muscular dystrophy (DMD) is an X-linked recessive disorder in which the loss of dystrophin causes progressive degeneration of skeletal and cardiac muscle. Potential therapies that carry substantial risk, such as gene- and cell-based approaches, must first be tested in animal models, notably the mdx mouse and several dystrophin-deficient breeds of dogs, including golden retriever muscular dystrophy (GRMD). Affected dogs have a more severe phenotype, in keeping with that of DMD, so may better predict disease pathogenesis and treatment efficacy. Various phenotypic tests have been developed to characterize disease progression in the GRMD model. These biomarkers range from measures of strength and joint contractures to magnetic resonance imaging. Some of these tests are routinely used in clinical veterinary practice, while others require specialized equipment and expertise. By comparing serial measurements from treated and untreated groups, one can document improvement or delayed progression of disease. Potential treatments for DMD may be broadly categorized as molecular, cellular, or pharmacologic. The GRMD model has increasingly been used to assess efficacy of a range of these therapies. A number of these studies have provided largely general proof-of-concept for the treatment under study. Others have demonstrated efficacy using the biomarkers discussed. Importantly, just as symptoms in DMD vary among patients, GRMD dogs display remarkable phenotypic variation. Though confounding statistical analysis in preclinical trials, this variation offers insight regarding the role that modifier genes play in disease pathogenesis. By correlating functional and mRNA profiling results, gene targets for therapy development can be identified.


Neuromuscular Disorders | 2003

The cranial sartorius muscle undergoes true hypertrophy in dogs with golden retriever muscular dystrophy

Joe N. Kornegay; Diane D Cundiff; Daniel J. Bogan; Janet R. Bogan; Carol S. Okamura

The degree of atrophy or hypertrophy of selected pelvic limb muscles was determined in the canine homologue of Duchenne muscular dystrophy. While most muscles were atrophied, the caudal and cranial sartorius were hypertrophied. Cranial sartorius weights were corrected for body weight and endomysial space to determine true muscle weights (g/kg; mean+/-SD) in three golden retriever muscular dystrophy age groups, 4-10 (Group 1; n=15), 13-26 (Group 2; n=4), and 33-66 (Group 3; n=4) months and grouped normal dogs (6-20 months; n=12). Group 1 golden retriever muscular dystrophy weights (2.2063+/-0.6884) were greater than those of normal dogs (1.2699+/-0.1966), indicating that young golden retriever muscular dystrophy dogs have true cranial sartorius muscle hypertrophy. Values of Group 2 (1.3758+/-0.5078) and Group 3 (0.5720+/-0.2423) golden retriever muscular dystrophy dogs were less than those of Group 1, suggesting that the cranial sartorius muscle atrophies over time. Given that cranial sartorius muscle weight correlated with tarsal joint angle in affected dogs (r=-0.817), the hypertrophied muscle may play a role analogous to iliotibial band tightness in Duchenne muscular dystrophy.


Muscle & Nerve | 1998

Evaluating motor end-plate-targeted injections of botulinum toxin type A in a canine model.

Martin K. Childers; Joe N. Kornegay; Roger Aoki; Laura Otaviani; Daniel J. Bogan; Greg Petroski

Tarsal joint forces were measured in dogs over 70 days following botulinum toxin type A (BTX‐A) injections. Three dogs were injected at motor end‐plates located by electromyography (EMG), while 3 dogs were similarly injected, but without EMG guidance. Extension forces were significantly (P < 0.05) smaller in limbs injected at motor end‐plates than in corresponding limbs on days 14 and 35. There were no significant differences at other times. Using these techniques, EMG end‐plate targeting potentiates effects of BTX‐A.


Journal of the Neurological Sciences | 1999

Contraction force generated by tarsal joint flexion and extension in dogs with golden retriever muscular dystrophy

Joe N. Kornegay; Daniel J. Bogan; Janet R. Bogan; Martin K. Childers; Diane D Cundiff; Gregory F. Petroski; Ronald O. Schueler

Force generated due to torque caused by tarsal joint flexion and extension was measured noninvasively at 3, 4.5, 6, and 12 months of age in dogs with the Duchenne homologue, golden retriever muscular dystrophy (GRMD). Absolute and body-weight-corrected GRMD twitch and tetanic force values were lower than normal at all ages (P<0.01 for most). Tarsal flexion and extension were differentially affected. Flexion values were especially low at 3 months, whereas extension was affected more at later ages. Several other GRMD findings differed from normal. The twitch/tetany ratio was generally lower; post-tetanic potentiation for flexion values was less marked; and extension relaxation and contraction times were longer. The consistency of GRMD values was studied to determine which measurements will be most useful in evaluating treatment outcome. Standard deviation was proportionally greater for GRMD versus normal recordings. More consistent values were seen for tetany versus twitch and for flexion versus extension. Left and right limb tetanic flexion values did not differ in GRMD; extension values were more variable. These results suggest that measurement of tarsal tetanic force should be most useful to document therapeutic benefit in GRMD dogs.


Muscle & Nerve | 2004

Effects of prednisone in canine muscular dystrophy.

Jocelyn M. K. Liu; Carol S. Okamura; Daniel J. Bogan; Janet R. Bogan; Martin K. Childers; Joe N. Kornegay

Glucocorticoid use may provide short‐term functional improvement in boys with Duchenne muscular dystrophy (DMD). We report functional and histopathologic changes following a 4‐month course of daily oral prednisone in a canine model of DMD, termed golden retriever muscular dystrophy (GRMD). Muscle extension forces in GRMD dogs treated daily with 1 and 2 mg/kg prednisone measured 2.349 ± 0.92 and 3.486 ± 0.67 N/kg, respectively, compared to 1.927 ± 0.63 N/kg in untreated GRMD controls (p < 0.05 for 2mg/kg group); GRMD muscle flexion forces measured 0.435 ± 0.13 and 0.303 ± 0.08 N/kg, respectively, compared to 0.527 ± 0.01 N/kg in untreated GRMD controls (p < 0.05 for both groups). Although cranial sartorius hypertrophy and tibiotarsal joint angles also tended to improve, myofiber calcification increased and fetal myosin expression decreased following prednisone. Thus, functional data indicate benefit but histopathologic changes following prednisone treatment in GRMD suggest possible deleterious consequences. Muscle Nerve, 2004


Physical Medicine and Rehabilitation Clinics of North America | 2012

The Paradox of Muscle Hypertrophy in Muscular Dystrophy

Joe N. Kornegay; Martin K. Childers; Daniel J. Bogan; Janet R. Bogan; Peter P. Nghiem; Jiahui Wang; Zheng Fan; James F. Howard; Scott J. Schatzberg; Jennifer L. Dow; Robert W. Grange; Martin Styner; Eric P. Hoffman; Kathryn R. Wagner

Mutations in the dystrophin gene cause Duchenne and Becker muscular dystrophy in humans and syndromes in mice, dogs, and cats. Affected humans and dogs have progressive disease that leads primarily to muscle atrophy. Mdx mice progress through an initial phase of muscle hypertrophy followed by atrophy. Cats have persistent muscle hypertrophy. Hypertrophy in humans has been attributed to deposition of fat and connective tissue (pseudohypertrophy). Increased muscle mass (true hypertrophy) has been documented in animal models. Muscle hypertrophy can exaggerate postural instability and joint contractures. Deleterious consequences of muscle hypertrophy should be considered when developing treatments for muscular dystrophy.


Methods of Molecular Biology | 2011

Golden retriever muscular dystrophy (GRMD): Developing and maintaining a colony and physiological functional measurements.

Joe N. Kornegay; Janet R. Bogan; Daniel J. Bogan; Martin K. Childers; Robert W. Grange

Studies of canine models of Duchenne muscular dystrophy (DMD) provide insight regarding disease pathogenesis and treatment efficacy. To take maximal advantage, colonies of affected dogs must be maintained and outcome parameters developed. In this chapter, we review our 25 years of experience with the golden retriever muscular dystrophy (GRMD) model. Key challenges in colony development (breeding, neonatal death, and the risk of inbreeding) and representative functional measurements (tibiotarsal joint angle and torque force; and eccentric contraction decrement) are discussed.


American Journal of Physical Medicine & Rehabilitation | 2001

Myofiber injury and regeneration in a canine homologue of Duchenne muscular dystrophy

Martin K. Childers; Carol S. Okamura; Daniel J. Bogan; Janet R. Bogan; Margaret J. Sullivan; Joe N. Kornegay

Childers MK, Okamura CS, Bogan DJ, Bogan JR, Sullivan MJ, Kornegay JN: Myofiber injury and regeneration in a canine homologue of Duchenne muscular dystrophy. Am J Phys Med Rehabil 2001;80:175–181. ObjectiveTo test the hypothesis that differential skeletal muscle involvement, previously observed in dogs with a homologue of Duchenne muscular dystrophy, correlates with the histochemical markers of myofiber injury and regeneration. DesignEvidence of injury (cellular penetration by Evans blue dye, immunoglobulin G expression, hematoxylin and eosin staining of necrotic figures), myofiber regeneration (fetal myosin heavy chain isoform expression), and morphologic indices in the cranial sartorius (CS), long digital extensor, and vastus lateralis muscles were examined in five dogs with dystrophy and five normal dogs. ResultsOnly the CS muscle, at 1 mo, demonstrated significant differences in injury when compared with age-matched controls. By 6 mo, the long digital extensor and vastus lateralis also suffered greater than normal injury. Only the dystrophic CS tissue expressed a notable increase in mean myofiber diameter when compared with other muscles at 6 mo. Normal CS muscles revealed a distinct population of small myofibers at this age. ConclusionThe CS seems unique in its selective pathologic involvement. These differences may contribute to the marked regenerative response of this muscle in the dystrophic state. An improved understanding of mechanisms by which some dystrophin-deficient canine muscles remain spared from injury may provide clues to investigate and prevent the degenerative processes in humans.

Collaboration


Dive into the Daniel J. Bogan's collaboration.

Top Co-Authors

Avatar

Janet R. Bogan

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jennifer L. Dow

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Jiahui Wang

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Martin Styner

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Zheng Fan

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hongtu Zhu

University of Texas Health Science Center at Houston

View shared research outputs
Researchain Logo
Decentralizing Knowledge