Network


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

Hotspot


Dive into the research topics where Stephen J. Kolb is active.

Publication


Featured researches published by Stephen J. Kolb.


Molecular Therapy | 2011

Systemic Gene Delivery in Large Species for Targeting Spinal Cord, Brain, and Peripheral Tissues for Pediatric Disorders

Adam K. Bevan; S. Duque; Kevin D. Foust; Pablo R Morales; Lyndsey Braun; Leah Schmelzer; Curtis M. Chan; Mary McCrate; Louis G. Chicoine; Brian D. Coley; Paul Porensky; Stephen J. Kolb; Arthur H.M. Burghes; Brian K. Kaspar

Adeno-associated virus type 9 (AAV9) is a powerful tool for delivering genes throughout the central nervous system (CNS) following intravenous injection. Preclinical results in pediatric models of spinal muscular atrophy (SMA) and lysosomal storage disorders provide a compelling case for advancing AAV9 to the clinic. An important translational step is to demonstrate efficient CNS targeting in large animals at various ages. In the present study, we tested systemically injected AAV9 in cynomolgus macaques, administered at birth through 3 years of age for targeting CNS and peripheral tissues. We show that AAV9 was efficient at crossing the blood–brain barrier (BBB) at all time points investigated. Transgene expression was detected primarily in glial cells throughout the brain, dorsal root ganglia neurons and motor neurons within the spinal cord, providing confidence for translation to SMA patients. Systemic injection also efficiently targeted skeletal muscle and peripheral organs. To specifically target the CNS, we explored AAV9 delivery to cerebrospinal fluid (CSF). CSF injection efficiently targeted motor neurons, and restricted gene expression to the CNS, providing an alternate delivery route and potentially lower manufacturing requirements for older, larger patients. Our findings support the use of AAV9 for gene transfer to the CNS for disorders in pediatric populations.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Direct conversion of patient fibroblasts demonstrates non-cell autonomous toxicity of astrocytes to motor neurons in familial and sporadic ALS

Kathrin Meyer; Laura Ferraiuolo; Carlos J. Miranda; Shibi Likhite; Sohyun McElroy; Samantha R. Renusch; Dara Ditsworth; Clotilde Lagier-Tourenne; Richard Smith; John Ravits; Arthur H.M. Burghes; Pamela J. Shaw; Don W. Cleveland; Stephen J. Kolb; Brian K. Kaspar

Significance Direct conversion is a recently established method to generate neuronal progenitor cells (NPCs) from skin fibroblasts in a fast and efficient manner. In this study, we show that this method can be used to model neurodegenerative diseases such as amyotrophic lateral sclerosis (ALS). Because the origin of ALS is mainly sporadic with unknown cause, methods to model the disease are urgently needed. The produced NPCs are differentiated into astrocytes, which are involved in motor neuron death in ALS. Strikingly, skin-derived astrocytes show similar toxicity toward motor neurons as astrocytes from autopsies of patients. This tool now allows studying ALS while the patient is still alive and can help in testing potential therapeutics for individual patients. Amyotrophic lateral sclerosis (ALS) causes motor neuron degeneration, paralysis, and death. Accurate disease modeling, identifying disease mechanisms, and developing therapeutics is urgently needed. We previously reported motor neuron toxicity through postmortem ALS spinal cord-derived astrocytes. However, these cells can only be harvested after death, and their expansion is limited. We now report a rapid, highly reproducible method to convert adult human fibroblasts from living ALS patients to induced neuronal progenitor cells and subsequent differentiation into astrocytes (i-astrocytes). Non-cell autonomous toxicity to motor neurons is found following coculture of i-astrocytes from familial ALS patients with mutation in superoxide dismutase or hexanucleotide expansion in C9orf72 (ORF 72 on chromosome 9) the two most frequent causes of ALS. Remarkably, i-astrocytes from sporadic ALS patients are as toxic as those with causative mutations, suggesting a common mechanism. Easy production and expansion of i-astrocytes now enables rapid disease modeling and high-throughput drug screening to alleviate astrocyte-derived toxicity.


JAMA Neurology | 2011

Spinal muscular atrophy: a timely review.

Stephen J. Kolb; John T. Kissel

Spinal muscular atrophy (SMA) is a neurodegenerative disease characterized by loss of motor neurons in the anterior horn of the spinal cord and resultant weakness. The most common form of SMA, accounting for 95% of cases, is autosomal recessive proximal SMA associated with mutations in the survival of motor neurons (SMN1) gene. Relentless progress during the past 15 years in the understanding of the molecular genetics and pathophysiology of SMA has resulted in a unique opportunity for rational, effective therapeutic trials. The goal of SMA therapy is to increase the expression levels of the SMN protein in the correct cells at the right time. With this target in sight, investigators can now effectively screen potential therapies in vitro, test them in accurate, reliable animal models, move promising agents forward to clinical trials, and accurately diagnose patients at an early or presymptomatic stage of disease. A major challenge for the SMA community will be to prioritize and develop the most promising therapies in an efficient, timely, and safe manner with the guidance of the appropriate regulatory agencies. This review will take a historical perspective to highlight important milestones on the road to developing effective therapies for SMA.


Neurology | 2010

Mutant small heat shock protein B3 causes motor neuropathy: Utility of a candidate gene approach

Stephen J. Kolb; P. J. Snyder; E. J. Poi; E. A. Renard; A. Bartlett; S. Gu; S. Sutton; W. D. Arnold; M. L. Freimer; V. H. Lawson; John T. Kissel; T. W. Prior

Objective: Idiopathic peripheral neuropathy is common and likely due to genetic factors that are not detectable using standard linkage analysis. We initiated a candidate gene approach to study the genetic influence of the small heat shock protein (sHSP) gene family on an axonal motor and motor/sensory neuropathy patient population. Methods: The promoter region and all exonic and intronic sequences of the 10 sHSP genes (HSPB1-HSPB10) were screened in a cohort of presumed nonacquired, axonal motor and motor/sensory neuropathy patients seen at the Ohio State University Neuromuscular Clinic. Results: A missense mutation in the gene encoding small heat shock protein B3 (HSPB3, also called HSP27, protein 3) was discovered in 2 siblings with an asymmetric axonal motor neuropathy. Electrophysiologic studies revealed an axonal, predominantly motor, length-dependent neuropathy. The mutation, HSPB3(R7S), is located in the N-terminal domain and involves the loss of a conserved arginine. Conclusions: The discovery of an HSPB3 mutation associated with an axonal motor neuropathy using a candidate gene approach supports the notion that the small heat shock protein gene family coordinately plays an important role in motor neuron viability.


Annals of Neurology | 2015

A large animal model of spinal muscular atrophy and correction of phenotype.

S. Duque; W. David Arnold; Philipp Odermatt; Xiaohui Li; Paul Porensky; Leah Schmelzer; Kathrin Meyer; Stephen J. Kolb; Daniel Schümperli; Brian K. Kaspar; Arthur H.M. Burghes

Spinal muscular atrophy (SMA) is caused by reduced levels of survival motor neuron (SMN) protein, which results in motoneuron loss. Therapeutic strategies to increase SMN levels including drug compounds, antisense oligonucleotides, and scAAV9 gene therapy have proved effective in mice. We wished to determine whether reduction of SMN in postnatal motoneurons resulted in SMA in a large animal model, whether SMA could be corrected after development of muscle weakness, and the response of clinically relevant biomarkers.


Neurologic Clinics | 2015

Spinal Muscular Atrophy

Stephen J. Kolb; John T. Kissel

Spinal muscular atrophy is an autosomal-recessive disorder characterized by degeneration of motor neurons in the spinal cord and caused by mutations in the survival motor neuron 1 gene, SMN1. The severity of SMA is variable. The SMN2 gene produces a fraction of the SMN messenger RNA (mRNA) transcript produced by the SMN1 gene. There is an inverse correlation between SMN2 gene copy number and clinical severity. Clinical management focuses on multidisciplinary care. Preclinical models of SMA have led to an explosion of SMA clinical trials that hold great promise of effective therapy in the future.


Muscle & Nerve | 2009

An analysis of disease severity based on SMN2 copy number in adults with spinal muscular atrophy

Bakri Elsheikh; Thomas W. Prior; Xiaoli Zhang; Robert G. Miller; Stephen J. Kolb; D. A N Moore; Walter G. Bradley; Richard J. Barohn; Wilson W. Bryan; Deborah Gelinas; Susan T. Iannaccone; Robert Leshner; Michelle Mendoza; Barry S. Russman; Stephen Smith; Wendy M. King; John T. Kissel

To evaluate the effect of SMN2 copy number on disease severity in spinal muscular atrophy (SMA), we stratified 45 adult SMA patients based on SMN2 copy number (3 vs. 4 copies). Patients with 3 copies had an earlier age of onset and lower spinal muscular atrophy functional rating scale (SMAFRS) scores and were more likely to be non‐ambulatory. There was, however, no difference between the groups in quantitative muscle strength or pulmonary function testing. Functional scale may be a more discriminating outcome measure for SMA clinical trials. Muscle Nerve, 2009


Annals of clinical and translational neurology | 2016

Baseline results of the NeuroNEXT spinal muscular atrophy infant biomarker study

Stephen J. Kolb; Christopher S. Coffey; Jon W. Yankey; Kristin J. Krosschell; W. David Arnold; Seward B. Rutkove; Kathryn J. Swoboda; Sandra P. Reyna; Ai Sakonju; Basil T. Darras; Richard Shell; Nancy L. Kuntz; Diana Castro; Susan T. Iannaccone; Julie Parsons; Anne M. Connolly; Claudia A. Chiriboga; Craig M. McDonald; W. Bryan Burnette; Klaus Werner; Mathula Thangarajh; Perry B. Shieh; Erika Finanger; Merit Cudkowicz; Michelle McGovern; D. Elizabeth McNeil; Richard S. Finkel; Edward M. Kaye; Allison Kingsley; Samantha R. Renusch

This study prospectively assessed putative promising biomarkers for use in assessing infants with spinal muscular atrophy (SMA).


Muscle & Nerve | 2014

SMA valiant trial: A prospective, double-blind, placebo-controlled trial of valproic acid in ambulatory adults with spinal muscular atrophy

John T. Kissel; Bakri Elsheikh; Wendy M. King; Miriam Freimer; Charles B. Scott; Stephen J. Kolb; Sandra P. Reyna; Thomas O. Crawford; Louise R. Simard; Kristin J. Krosschell; Gyula Acsadi; Mary K. Schroth; Guy D'Anjou; Bernard LaSalle; Thomas W. Prior; Susan Sorenson; Jo Anne Maczulski; Kathryn J. Swoboda

Introduction: An open‐label trial suggested that valproic acid (VPA) improved strength in adults with spinal muscular atrophy (SMA). We report a 12‐month, double‐blind, cross‐over study of VPA in ambulatory SMA adults. Methods: There were 33 subjects, aged 20–55 years, included in this investigation. After baseline assessment, subjects were randomized to receive VPA (10–20 mg/kg/day) or placebo. At 6 months, patients were switched to the other group. Assessments were performed at 3, 6, and 12 months. The primary outcome was the 6‐month change in maximum voluntary isometric contraction testing with pulmonary, electrophysiological, and functional secondary outcomes. Results: Thirty subjects completed the study. VPA was well tolerated, and compliance was good. There was no change in primary or secondary outcomes at 6 or 12 months. Conclusions: VPA did not improve strength or function in SMA adults. The outcomes used are feasible and reliable and can be employed in future trials in SMA adults. Muscle Nerve 49: 187–192, 2014


PLOS ONE | 2014

Safety, Pharmacokinetic, and Functional Effects of the Nogo-A Monoclonal Antibody in Amyotrophic Lateral Sclerosis: A Randomized, First-In-Human Clinical Trial

Vincent Meininger; Pierre François Pradat; Andrea M. Corse; Safa Al-Sarraj; Benjamin Rix Brooks; James B. Caress; Merit Cudkowicz; Stephen J. Kolb; Dale J. Lange; P. Nigel Leigh; Thomas Meyer; Stefano Milleri; Karen E. Morrison; Richard W. Orrell; Gary Peters; Jeffrey D. Rothstein; Jeremy M. Shefner; Arseniy Lavrov; Nicola Williams; Phil Overend; Jeffrey Price; Stewart Bates; Jonathan Bullman; David Krull; Alienor Berges; Bams Abila; Guy Meno-Tetang; Jens Würthner

The neurite outgrowth inhibitor, Nogo-A, has been shown to be overexpressed in skeletal muscle in amyotrophic lateral sclerosis (ALS); it is both a potential biomarker and therapeutic target. We performed a double-blind, two-part, dose-escalation study, in subjects with ALS, assessing safety, pharmacokinetics (PK) and functional effects of ozanezumab, a humanized monoclonal antibody against Nogo-A. In Part 1, 40 subjects were randomized (3∶1) to receive single dose intravenous ozanezumab (0.01, 0.1, 1, 5, or 15 mg/kg) or placebo. In Part 2, 36 subjects were randomized (3∶1) to receive two repeat doses of intravenous ozanezumab (0.5, 2.5, or 15 mg/kg) or placebo, approximately 4 weeks apart. The primary endpoints were safety and tolerability (adverse events [AEs], vital signs, electrocardiogram (ECG), and clinical laboratory tests). Secondary endpoints included PK, immunogenicity, functional endpoints (clinical and electrophysiological), and biomarker parameters. Overall, ozanezumab treatment (0.01–15 mg/kg) was well tolerated. The overall incidence of AEs in the repeat dose 2.5 mg/kg and 15 mg/kg ozanezumab groups was higher than in the repeat dose placebo group and repeat dose 0.5 mg/kg ozanezumab group. The majority were considered not related to study drug by the investigators. Six serious AEs were reported in three subjects receiving ozanezumab; none were considered related to study drug. No study drug-related patterns were identified for ECG, laboratory, or vital signs parameters. One subject (repeat dose 15 mg/kg ozanezumab) showed a weak, positive anti-ozanezumab-antibody result. PK results were generally consistent with monoclonal antibody treatments. No apparent treatment effects were observed for functional endpoints or muscle biomarkers. Immunohistochemical staining showed dose-dependent co-localization of ozanezumab with Nogo-A in skeletal muscle. In conclusion, single and repeat dose ozanezumab treatment was well tolerated and demonstrated co-localization at the site of action. These findings support future studies with ozanezumab in ALS. Trial Registration ClinicalTrials.gov NCT00875446 GSK-ClinicalStudyRegister.com GSK ID 111330

Collaboration


Dive into the Stephen J. Kolb's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

W. David Arnold

The Ohio State University Wexner Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Seward B. Rutkove

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge