Network


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

Hotspot


Dive into the research topics where Alan Pestronk is active.

Publication


Featured researches published by Alan Pestronk.


Nature Genetics | 2004

Inclusion body myopathy associated with Paget disease of bone and frontotemporal dementia is caused by mutant valosin-containing protein

Giles D. J. Watts; Jill Wymer; Margaret J. Kovach; Sarju G. Mehta; Steven Mumm; Daniel Darvish; Alan Pestronk; Michael P. Whyte; Virginia E. Kimonis

Inclusion body myopathy associated with Paget disease of bone and frontotemporal dementia (IBMPFD) is a dominant progressive disorder that maps to chromosome 9p21.1–p12. We investigated 13 families with IBMPFD linked to chromosome 9 using a candidate-gene approach. We found six missense mutations in the gene encoding valosin-containing protein (VCP, a member of the AAA-ATPase superfamily) exclusively in all 61 affected individuals. Haplotype analysis indicated that descent from two founders in two separate North American kindreds accounted for IBMPFD in ∼50% of affected families. VCP is associated with a variety of cellular activities, including cell cycle control, membrane fusion and the ubiquitin-proteasome degradation pathway. Identification of VCP as causing IBMPFD has important implications for other inclusion-body diseases, including myopathies, dementias and Paget disease of bone (PDB), as it may define a new common pathological ubiquitin-based pathway.


Nature | 2013

Mutations in prion-like domains in hnRNPA2B1 and hnRNPA1 cause multisystem proteinopathy and ALS

Hong Joo Kim; Nam Chul Kim; Yong-Dong Wang; Emily A. Scarborough; Jennifer C. Moore; Zamia Diaz; Kyle S. MacLea; Brian D. Freibaum; Songqing Li; Amandine Molliex; A. Kanagaraj; Robert A. Carter; Kevin B. Boylan; Aleksandra Wojtas; Rosa Rademakers; Jack L. Pinkus; Steven A. Greenberg; John Q. Trojanowski; Bryan J. Traynor; Bradley Smith; Simon Topp; Athina-Soragia Gkazi; John Miller; Christopher Shaw; Michael Kottlors; Janbernd Kirschner; Alan Pestronk; Yun R. Li; Alice Flynn Ford; Aaron D. Gitler

Algorithms designed to identify canonical yeast prions predict that around 250 human proteins, including several RNA-binding proteins associated with neurodegenerative disease, harbour a distinctive prion-like domain (PrLD) enriched in uncharged polar amino acids and glycine. PrLDs in RNA-binding proteins are essential for the assembly of ribonucleoprotein granules. However, the interplay between human PrLD function and disease is not understood. Here we define pathogenic mutations in PrLDs of heterogeneous nuclear ribonucleoproteins (hnRNPs) A2B1 and A1 in families with inherited degeneration affecting muscle, brain, motor neuron and bone, and in one case of familial amyotrophic lateral sclerosis. Wild-type hnRNPA2 (the most abundant isoform of hnRNPA2B1) and hnRNPA1 show an intrinsic tendency to assemble into self-seeding fibrils, which is exacerbated by the disease mutations. Indeed, the pathogenic mutations strengthen a ‘steric zipper’ motif in the PrLD, which accelerates the formation of self-seeding fibrils that cross-seed polymerization of wild-type hnRNP. Notably, the disease mutations promote excess incorporation of hnRNPA2 and hnRNPA1 into stress granules and drive the formation of cytoplasmic inclusions in animal models that recapitulate the human pathology. Thus, dysregulated polymerization caused by a potent mutant steric zipper motif in a PrLD can initiate degenerative disease. Related proteins with PrLDs should therefore be considered candidates for initiating and perhaps propagating proteinopathies of muscle, brain, motor neuron and bone.


Annals of Neurology | 2008

TDP-43 A315T mutation in familial motor neuron disease

Michael A. Gitcho; Robert H. Baloh; Sumi Chakraverty; Kevin Mayo; Joanne Norton; Denise Levitch; Kimmo J. Hatanpaa; Charles L. White; Eileen H. Bigio; Richard J. Caselli; Matt Baker; Muhammad Al-Lozi; John C. Morris; Alan Pestronk; Rosa Rademakers; Alison Goate; Nigel J. Cairns

To identify novel causes of familial neurodegenerative diseases, we extended our previous studies of TAR DNA‐binding protein 43 (TDP‐43) proteinopathies to investigate TDP‐43 as a candidate gene in familial cases of motor neuron disease. Sequencing of the TDP‐43 gene led to the identification of a novel missense mutation, Ala‐315‐Thr, which segregates with all affected members of an autosomal dominant motor neuron disease family. The mutation was not found in 1,505 healthy control subjects. The discovery of a missense mutation in TDP‐43 in a family with dominantly inherited motor neuron disease provides evidence of a direct link between altered TDP‐43 function and neurodegeneration. Ann Neurol 2008


The New England Journal of Medicine | 2010

A Randomized Study of Alglucosidase Alfa in Late-Onset Pompe's Disease

Ans T. van der Ploeg; Paula R. Clemens; Deyanira Corzo; Diana M. Escolar; Julaine Florence; Geert Jan Groeneveld; Serge Herson; Priya S. Kishnani; P. Laforêt; Stephen Lake; Dale J. Lange; Robert Leshner; J. Mayhew; Claire Morgan; Kenkichi Nozaki; Dorothy J. Park; Alan Pestronk; Barry E. Rosenbloom; Alison Skrinar; Carine I. van Capelle; Nadine A. van der Beek; Melissa P. Wasserstein; Sasa Zivkovic

BACKGROUND Pompes disease is a metabolic myopathy caused by a deficiency of acid alpha glucosidase (GAA), an enzyme that degrades lysosomal glycogen. Late-onset Pompes disease is characterized by progressive muscle weakness and loss of respiratory function, leading to early death. We conducted a randomized, placebo-controlled trial of alglucosidase alfa, a recombinant human GAA, for the treatment of late-onset Pompes disease. METHODS Ninety patients who were 8 years of age or older, ambulatory, and free of invasive ventilation were randomly assigned to receive biweekly intravenous alglucosidase alfa (20 mg per kilogram of body weight) or placebo for 78 weeks at eight centers in the United States and Europe. The two primary end points were distance walked during a 6-minute walk test and percentage of predicted forced vital capacity (FVC). RESULTS At 78 weeks, the estimated mean changes from baseline in the primary end points favored alglucosidase alfa (an increase of 28.1+/-13.1 m on the 6-minute walk test and an absolute increase of 3.4+/-1.2 percentage points in FVC; P=0.03 and P=0.006, respectively). Similar proportions of patients in the two groups had adverse events, serious adverse events, and infusion-associated reactions; events that occurred only in patients who received the active study drug included anaphylactic reactions and infusion-associated reactions of urticaria, flushing, hyperhidrosis, chest discomfort, vomiting, and increased blood pressure (each of which occurred in 5 to 8% of the patients). CONCLUSIONS In this study population, treatment with alglucosidase alfa was associated with improved walking distance and stabilization of pulmonary function over an 18-month period. (ClinicalTrials.gov number, NCT00158600.)


Science | 1975

Myasthenia gravis: passive transfer from man to mouse

Klaus V. Toyka; Db Brachman; Alan Pestronk; Ing Kao

Daily injections into mice of an ammonium sulfate-precipitated immunoglobulin fraction of serum from patients with myasthenia gravis were carried out for up to 14 days. The mice showed reduced amplitudes of miniature endplate potentials and reduced numbers of acetylcholine receptors at the neuromuscular junctions. Some mice showed typical decremental responses on repetitive nerve stimulation, with reversal by neostigmine. This represents the first evidence of a circulating factor in the serum of patients with myasthenia gravis which on passive transfer reproduces features of the disease in experimental animals.


The New England Journal of Medicine | 1977

Myasthenia gravis. Study of humoral immune mechanisms by passive transfer to mice

Klaus V. Toyka; Daniel B. Drachman; Diane E. Griffin; Alan Pestronk; Winkelstein Ja; Fishbeck Kh; Ing Kao

To study the role of humoral factors in the pathogenesis of myasthenia gravis, we employed passive transfer of human serum fractions to mice. Immunoglobulins from 16 patients with myasthenia gravis were injected into mice daily for one to 14 days. Typical myasthenic features of reduction in amplitude of miniature end-plate potentials (mean change more than 50 per cent, P less than 0.005) or reduction in acetylcholine receptors at neuromuscular junctions (mean change more than 50 per cent, P less than 0.005) (or both) were produced by immunoglobulin from 15 of the 16 patients. Some mice showed weakness or decremental responses to repetitive nerve stimulation as well. The active fraction was identified as IgG by three different purification methods. Its effect was enhanced by the third component (C3) of the complement system, but the fifth component (C5) had no effect. These data suggest that the pathogenesis of myasthenia gravis often involves and antibody-mediated autoimmune attack on the acetylcholine receptors of the neuromuscular junction.


Annals of Neurology | 2008

A Phase I/II trial of MYO-029 in Adult Subjects with Muscular Dystrophy

Kathryn R. Wagner; James L. Fleckenstein; Anthony A. Amato; Richard J. Barohn; K. Bushby; Diana M. Escolar; Kevin M. Flanigan; Alan Pestronk; Rabi Tawil; Gil I. Wolfe; Michelle Eagle; Julaine Florence; Wendy M. King; Shree Pandya; Volker Straub; Paul Juneau; Kathleen Meyers; Cristina Csimma; Tracey Araujo; Robert Allen; Stephanie A. Parsons; John M. Wozney; Edward R. LaVallie

Myostatin is an endogenous negative regulator of muscle growth and a novel target for muscle diseases. We conducted a safety trial of a neutralizing antibody to myostatin, MYO‐029, in adult muscular dystrophies (Becker muscular dystrophy, facioscapulohumeral dystrophy, and limb‐girdle muscular dystrophy).


Neurology | 1999

IgM antibody-related polyneuropathies: B-cell depletion chemotherapy using Rituximab

Todd Levine; Alan Pestronk

Article abstract Current treatments for anti-GM1 ganglioside or antimyelin-associated glycoprotein (anti-MAG) antibody-associated polyneuropathies are toxic or very costly. In this preliminary study the authors treated five patients with neuropathy and immunoglobulin M antibodies to GM1 ganglioside or MAG by depleting B cells using Rituximab—a monoclonal antibody directed against the B-cell surface membrane marker CD20. Within 3 to 6 months after treatment, all five patients had improved function, significantly increased quantitative strength measurements, and reduced titers of serum autoantibodies.


Lancet Neurology | 2013

An antisense oligonucleotide against SOD1 delivered intrathecally for patients with SOD1 familial amyotrophic lateral sclerosis: A phase 1, randomised, first-in-man study

Timothy M. Miller; Alan Pestronk; William S. David; Jeff rey Rothstein; Ericka Simpson; Stanley H. Appel; Patricia L. Andres; Katy Mahoney; Peggy Allred; Katie Alexander; Lyle W. Ostrow; David A. Schoenfeld; Eric A. Macklin; Daniel Norris; Georgios Manousakis; Matthew J. Crisp; Richard Smith; C. Frank Bennett; Kathie M. Bishop; Merit Cudkowicz

BACKGROUND Mutations in SOD1 cause 13% of familial amyotrophic lateral sclerosis. In the SOD1 Gly93Ala rat model of amyotrophic lateral sclerosis, the antisense oligonucleotide ISIS 333611 delivered to CSF decreased SOD1 mRNA and protein concentrations in spinal cord tissue and prolonged survival. We aimed to assess the safety, tolerability, and pharmacokinetics of ISIS 333611 after intrathecal administration in patients with SOD1-related familial amyotrophic lateral sclerosis. METHODS In this randomised, placebo-controlled, phase 1 trial, we delivered ISIS 333611 by intrathecal infusion using an external pump over 11·5 h at increasing doses (0·15 mg, 0·50 mg, 1·50 mg, 3·00 mg) to four cohorts of eight patients with SOD1-positive amyotrophic lateral sclerosis (six patients assigned to ISIS 333611, two to placebo in each cohort). We did the randomisation with a web-based system, assigning patients in blocks of four. Patients and investigators were masked to treatment assignment. Participants were allowed to re-enrol in subsequent cohorts. Our primary objective was to assess the safety and tolerability of ISIS 333611. Assessments were done during infusion and over 28 days after infusion. This study was registered with Clinicaltrials.gov, number NCT01041222. FINDINGS Seven of eight (88%) patients in the placebo group versus 20 of 24 (83%) in the ISIS 333611 group had adverse events. The most common events were post-lumbar puncture syndrome (3/8 [38%] vs 8/24 [33%]), back pain (4/8 [50%] vs 4/24 [17%]), and nausea (0/8 [0%] vs 3/24 [13%]). We recorded no dose-limiting toxic effects or any safety or tolerability concerns related to ISIS 333611. No serious adverse events occurred in patients given ISIS 333611. Re-enrolment and re-treatment were also well tolerated. INTERPRETATION This trial is the first clinical study of intrathecal delivery of an antisense oligonucleotide. ISIS 333611 was well tolerated when administered as an intrathecal infusion. Antisense oligonucleotides delivered to the CNS might be a feasible treatment for neurological disorders. FUNDING The ALS Association, Muscular Dystrophy Association, Isis Pharmaceuticals.


The Journal of Neuroscience | 2007

Altered Axonal Mitochondrial Transport in the Pathogenesis of Charcot-Marie-Tooth Disease from Mitofusin 2 Mutations

Robert H. Baloh; Robert E. Schmidt; Alan Pestronk; Jeffrey Milbrandt

Mutations in the mitochondrial fusion protein mitofusin 2 (MFN2) are the most commonly identified cause of Charcot-Marie-Tooth type 2 (CMT2), a dominantly inherited disease characterized by degeneration of peripheral sensory and motor axons. However, the mechanism by which mutations in this ubiquitously expressed mitochondrial fusion protein lead to neuropathy has not yet been elucidated. To explore how MFN2 mutations lead to degeneration of peripheral axons, we expressed neuropathy-associated forms of MFN2 in cultured dorsal root ganglion neurons, cells preferentially affected in CMT2. Disease-associated MFN2 mutant proteins induced abnormal clustering of small fragmented mitochondria in both neuronal cell bodies and proximal axons. Interestingly, transport of mitochondria in axons was significantly impaired in neurons expressing disease-mutated forms of MFN2. The diminished axonal mitochondrial transport was not attributable to diminished ATP levels in the neurons, and oxidative respiration was normal in mutant MFN2-expressing cells. Additionally, mitochondrial oxidative enzyme activity was normal in muscle mitochondria from a CMT2 patient with an MFN2 mutation, further supporting that abnormal mitochondrial transport in neurons is independent from an energy production defect. This abnormal mitochondrial trafficking provides a likely explanation for the selective susceptibility of the longest peripheral axons to MFN2 mutations, in which proper localization of mitochondria is critical for axonal and synaptic function.

Collaboration


Dive into the Alan Pestronk's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Glenn Lopate

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Anne M. Connolly

St. Louis Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Julaine Florence

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Robert H. Baloh

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

Conrad C. Weihl

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Matthew B. Harms

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Rati Choksi

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Craig M. Zaidman

Washington University in St. Louis

View shared research outputs
Researchain Logo
Decentralizing Knowledge