Network


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

Hotspot


Dive into the research topics where Melissa A. Goddard is active.

Publication


Featured researches published by Melissa A. Goddard.


Science Translational Medicine | 2014

Gene Therapy Prolongs Survival and Restores Function in Murine and Canine Models of Myotubular Myopathy

Martin K. Childers; Romain Joubert; Karine Poulard; C. Moal; Robert W. Grange; Jonathan Doering; Michael W. Lawlor; Branden E. Rider; T. Jamet; N. Danièle; Samia Martin; Christel Rivière; Thomas Soker; Caroline Hammer; Laetitia van Wittenberghe; Mandy Lockard; Xuan Guan; Melissa A. Goddard; Erin Mitchell; Jane Barber; J. Koudy Williams; David L. Mack; Mark E. Furth; Alban Vignaud; Carole Masurier; Fulvio Mavilio; Philippe Moullier; Alan H. Beggs; Anna Buj-Bello

Intravenous injection of an adeno-associated viral vector expressing the myotubularin (MTM1) gene improves survival and rescues skeletal muscle function in mice and dogs affected by myotubular myopathy. Restoring Skeletal Muscle Function X-linked myotubular myopathy is a fatal disease of skeletal muscle that affects about 1 in 50,000 male births. Patients harbor mutations in the MTM1 gene and are typically born floppy, with severely weak limb and respiratory muscles. Survival requires intensive support, often including tube feeding and mechanical ventilation, but effective therapy is not available for patients. Gene replacement therapy using adeno-associated viral (AAV) vectors has potential for the treatment of inherited diseases like myotubular myopathy. Therefore, Childers et al. tested the effects of a recombinant AAV vector expressing myotubularin in two animal models of myotubularin deficiency: Mtm1 knockout mice and dogs carrying a naturally occurring MTM1 gene mutation. Results in both mice and dogs showed that a single intravascular injection of AAV strengthened severely weak muscles, corrected muscle pathology, and prolonged survival. No toxicity or immune response was observed in dogs. These results demonstrate the efficacy of gene replacement therapy for myotubular myopathy in animal models and pave the way to a clinical trial in patients. Loss-of-function mutations in the myotubularin gene (MTM1) cause X-linked myotubular myopathy (XLMTM), a fatal, congenital pediatric disease that affects the entire skeletal musculature. Systemic administration of a single dose of a recombinant serotype 8 adeno-associated virus (AAV8) vector expressing murine myotubularin to Mtm1-deficient knockout mice at the onset or at late stages of the disease resulted in robust improvement in motor activity and contractile force, corrected muscle pathology, and prolonged survival throughout a 6-month study. Similarly, single-dose intravascular delivery of a canine AAV8-MTM1 vector in XLMTM dogs markedly improved severe muscle weakness and respiratory impairment, and prolonged life span to more than 1 year in the absence of toxicity or a humoral or cell-mediated immune response. These results demonstrate the therapeutic efficacy of AAV-mediated gene therapy for myotubular myopathy in small- and large-animal models, and provide proof of concept for future clinical trials in XLMTM patients.


Muscle & Nerve | 2012

Muscle function in a canine model of X-linked myotubular myopathy.

Robert W. Grange; Jon Doering; Erin Mitchell; Melanie N. Holder; Xuan Guan; Melissa A. Goddard; Christopher Tegeler; Alan H. Beggs; Martin K. Childers

Introduction: We established a colony of dogs that harbor an X‐linked MTM1 missense mutation.Muscle from affected male dogs exhibits reduction and altered localization of the MTM1 gene product, myotubularin, and provides a model analogous to X‐linked myotubular myopathy (XLMTM). Methods: We studied hindlimb muscle function in age‐matched canine XLMTM genotypes between ages 9 and 18 weeks. Results: By the end of the study, affected dogs produce only ∼15% of the torque generated by normals or carriers (0.023 ± 0.005 vs. 0.152 ± 0.007 and 0.154 ± 0.003 N‐m/kg body mass, respectively, P < 0.05) and are too weak to stand unassisted. At this age, XLMTM dogs also demonstrate an abnormally low twitch:tetanus ratio, a right‐shifted torque‐frequency relationship and an increase in torque during repetitive stimulation (P < 0.05). Conclusions: We hypothesize that muscle weakness results from impaired excitation‐contraction (E‐C) coupling. Interventions that improve E‐C coupling might be translated from the XLMTM dog model to patients. Muscle Nerve 46: 588–591, 2012


Physical Medicine and Rehabilitation Clinics of North America | 2012

Establishing Clinical End Points of Respiratory Function in Large Animals for Clinical Translation

Melissa A. Goddard; Erin Mitchell; Barbara K. Smith; Martin K. Childers

Respiratory dysfunction due progressive weakness of the respiratory muscles, particularly the diaphragm, is a major cause of death in the neuromuscular disease (NMD) X-linked myotubular myopathy (XLMTM). Methods of respiratory assessment in patients are often difficult, especially in those who are mechanically ventilated. The naturally occuring XLMTM dog model exhibits a phenotype similar to that in patients and can be used to determine quantitative descriptions of dysfunction as clinical endpoints for treatment and the development of new therapies. In experiments using respiratory impedance plethysmography (RIP), XLMTM dogs challenged with the respiratory stimulant doxapram displayed significant changes indicative of diaphragmatic weakness.


Journal of the Neurological Sciences | 2014

Gait characteristics in a canine model of X-linked myotubular myopathy

Melissa A. Goddard; Emily Burlingame; Alan H. Beggs; Anna Buj-Bello; Martin K. Childers; Anthony P. Marsh; Valerie E. Kelly

X-linked myotubular myopathy (XLMTM) is a fatal pediatric disease where affected boys display profound weakness of the skeletal muscles. Possible therapies are under development but robust outcome measures in animal models are required for effective translation to human patients. We established a naturally-occurring canine model, where XLMTM dogs display clinical symptoms similar to those observed in humans. The aim of this study was to determine potential endpoints for the assessment of future treatments in this model. Video-based gait analysis was selected, as it is a well-established method of assessing limb function in neuromuscular disease and measures have been correlated to the patients quality of life. XLMTM dogs (N = 3) and their true littermate wild type controls (N = 3) were assessed at 4-5 time points, beginning at 10 weeks and continuing through 17 weeks. Motion capture and an instrumented carpet were used separately to evaluate spatiotemporal and kinematic changes over time. XLMTM dogs walk more slowly and with shorter stride lengths than wild type dogs, and these differences became greater over time. However, there was no clear difference in angular measures between affected and unaffected dogs. These data demonstrate that spatiotemporal parameters capture functional changes in gait in an XLMTM canine model and support their utility in future therapeutic trials.


Muscle & Nerve | 2017

Long-term effects of systemic gene therapy in a canine model of myotubular myopathy

Matthew Elverman; Melissa A. Goddard; David L. Mack; Jessica M. Snyder; Michael W. Lawlor; Hui Meng; Alan H. Beggs; Ana Buj-Bello; Karine Poulard; Anthony P. Marsh; Robert W. Grange; Valerie E. Kelly; Martin K. Childers

X‐linked myotubular myopathy (XLMTM), a devastating pediatric disease caused by the absence of the protein myotubularin, results from mutations in the MTM1 gene. While there is no cure for XLMTM, we previously reported effects of MTM1 gene therapy using adeno‐associated virus (AAV) vector on muscle weakness and pathology in MTM1‐mutant dogs. Here, we followed 2 AAV‐infused dogs over 4 years.


Muscle & Nerve | 2014

Respiratory assessment in centronuclear myopathies.

Barbara K. Smith; Melissa A. Goddard; Martin K. Childers

The centronuclear myopathies (CNMs) are a group of inherited neuromuscular disorders classified as congenital myopathies. While several causative genes have been identified, some patients do not harbor any of the currently known mutations. These diverse disorders have common histological features, which include a high proportion of centrally nucleated muscle fibers, and clinical attributes of muscle weakness and respiratory insufficiency. Respiratory problems in CNMs may manifest initially during sleep, but daytime symptoms, ineffective airway clearance, and hypoventilation predominate as more severe respiratory muscle dysfunction evolves. Respiratory muscle capacity can be evaluated using a variety of clinical tests selected with consideration for the age and baseline motor function of the patient. Similar clinical tests of respiratory function can also be incorporated into preclinical CNM canine models to offer insight for clinical trials. Because respiratory problems account for significant morbidity in patients, routine assessments of respiratory muscle function are discussed. Muscle Nerve 50: 315–326, 2014


Methods | 2016

Gene therapy in monogenic congenital myopathies.

Xuan Guan; Melissa A. Goddard; David L. Mack; Martin K. Childers

Current treatment options for patients with monogenetic congenital myopathies (MCM) ameliorate the symptoms of the disorder without resolving the underlying cause. However, gene therapies are being developed where the mutated or deficient gene target is replaced. Preclinical findings in animal models appear promising, as illustrated by gene replacement for X-linked myotubular myopathy (XLMTM) in canine and murine models. Prospective applications and approaches to gene replacement therapy, using these disorders as examples, are discussed in this review.


Human gene therapy. Clinical development | 2015

Validity of a neurological scoring system for canine X-linked myotubular myopathy.

Jessica M. Snyder; Allison Meisner; David L. Mack; Melissa A. Goddard; Ian T. Coulter; Robert W. Grange; Martin K. Childers

A simple clinical neurological test was developed to evaluate response to gene therapy in a preclinical canine model of X-linked myotubular myopathy (XLMTM). This devastating congenital myopathy is caused by mutation in the myotubularin (MTM1) gene. Clinical signs include muscle weakness, early respiratory failure, and ventilator dependence. A spontaneously occurring canine model has a similar clinical picture and histological abnormalities on muscle biopsy compared with patients. We developed a neuromuscular assessment score, graded on a scale from 10 (normal) to 1 (unable to maintain sternal recumbency). We hypothesize that this neurological assessment score correlates with genotype and established measures of disease severity and is reliable when performed by an independent observer. At 17 weeks of age, there was strong correlation between neurological assessment scores and established methods of severity testing. The neurological severity score correctly differentiated between XLMTM and wild-type dogs with good interobserver reliability, on the basis of strong agreement between neurological scores assigned by independent observers. Together, these data indicate that the neurological scoring system developed for this canine congenital neuromuscular disorder is reliable and valid. This scoring system may be helpful in evaluating response to therapy in preclinical testing in this disease model, such as response to gene therapy.


Annals of Translational Medicine | 2015

Muscle pathology, limb strength, walking gait, respiratory function and neurological impairment establish disease progression in the p.N155K canine model of X-linked myotubular myopathy.

Melissa A. Goddard; David L. Mack; Stefan M. Czerniecki; Valerie E. Kelly; Jessica M. Snyder; Robert W. Grange; Michael W. Lawlor; Barbara K. Smith; Alan H. Beggs; Martin K. Childers

BACKGROUND Loss-of-function mutations in the myotubularin (MTM1) gene cause X-linked myotubular myopathy (XLMTM), a fatal, inherited pediatric disease that affects the entire skeletal musculature. Labrador retriever dogs carrying an MTM1 missense mutation exhibit strongly reduced synthesis of myotubularin, the founder member of a lipid phosphatase required for normal skeletal muscle function. The resulting canine phenotype resembles that of human patients with comparably severe mutations, and survival does not normally exceed 4 months. METHODS We studied MTM1 mutant dogs (n=7) and their age-matched control littermates (n=6) between the ages of 10 and 25 weeks. Investigators blinded to the animal identities sequentially measured limb muscle pathology, fore- and hind limb strength, walking gait, respiratory function and neurological impairment. RESULTS MTM1-mutant puppies display centrally-nucleated myofibers of reduced size and disrupted sarcotubular architecture progressing until the end of life, an average of 17 weeks. In-life measures of fore- and hind limb strength establish the rate at which XLMTM muscles weaken, and their corresponding decrease in gait velocity and stride length. Pulmonary function tests in affected dogs reveal a right-shifted relationship between peak inspiratory flow (PIF) and inspiratory time (TI); neurological assessments indicate that affected puppies as young as 10 weeks show early signs of neurological impairment (neurological severity score, NSS =8.6±0.9) with progressive decline (NSS =5.6±1.7 at 17 weeks-of-age). CONCLUSIONS Our findings document the rate of disease progression in a large animal model of XLMTM and lay a foundation for preclinical studies.


Translating Regenerative Medicine to the Clinic | 2016

Chapter 13 – Gene Therapy in Monogenic Congenital Myopathies

Xuan Guan; Melissa A. Goddard; David L. Mack; Martin K. Childers

Current treatment options for patients with monogenic congenital myopathies ameliorate the symptoms of the disorder without resolving the underlying cause. However, therapies are being developed where the mutated or deficient gene target is replaced. Thousands of clinical trials are underway relating to gene therapy, with around 9% focused on monogenic diseases such as Duchenne muscular dystrophy and limb girdle muscular dystrophy. Preclinical findings in animal models have also been promising, as illustrated by studies of a potential treatment for X-linked myotubular myopathy in canine and murine models. We will therefore discuss the prospective applications and approaches of gene replacement therapy, using these disorders as examples.

Collaboration


Dive into the Melissa A. Goddard's collaboration.

Top Co-Authors

Avatar

Martin K. Childers

Wake Forest Institute for Regenerative Medicine

View shared research outputs
Top Co-Authors

Avatar

Alan H. Beggs

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

David L. Mack

University of Washington

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michael W. Lawlor

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Xuan Guan

University of Washington

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hui Meng

Medical College of Wisconsin

View shared research outputs
Researchain Logo
Decentralizing Knowledge