Network


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

Hotspot


Dive into the research topics where Lindsay M. Wallace is active.

Publication


Featured researches published by Lindsay M. Wallace.


Annals of Neurology | 2011

DUX4, a candidate gene for facioscapulohumeral muscular dystrophy, causes p53-dependent myopathy in vivo

Lindsay M. Wallace; Sara E. Garwick; Wenyan Mei; Alexandra Belayew; Frédérique Coppée; Katherine J. Ladner; Denis C. Guttridge; Jing Yang; Scott Q. Harper

Facioscapulohumeral muscular dystrophy (FSHD) is associated with D4Z4 repeat contraction on human chromosome 4q35. This genetic lesion does not result in complete loss or mutation of any gene. Consequently, the pathogenic mechanisms underlying FSHD have been difficult to discern. In leading FSHD pathogenesis models, D4Z4 contractions are proposed to cause epigenetic changes, which ultimately increase expression of genes with myopathic potential. Although no gene has been conclusively linked to FSHD development, recent evidence supports a role for the D4Z4‐encoded DUX4 gene in FSHD. In this study, our objective was to test the in vivo myopathic potential of DUX4.


Molecular Therapy | 2012

RNA Interference Inhibits DUX4-induced Muscle Toxicity In Vivo: Implications for a Targeted FSHD Therapy

Lindsay M. Wallace; Jian Liu; Jacqueline S. Domire; Sara E Garwick-Coppens; S. Guckes; Kevin M. Flanigan; Scott Q. Harper

No treatment exists for facioscapulohumeral muscular dystrophy (FSHD), one of the most common inherited muscle diseases. Although FSHD can be debilitating, little effort has been made to develop targeted therapies. This lack of focus on targeted FSHD therapy perpetuated because the genes and pathways involved in the disorder were not understood. Now, more than 2 decades after efforts to decipher the root cause of FSHD began, this barrier to translation is finally lowering. Specifically, several recent studies support an FSHD pathogenesis model involving overexpression of the myopathic DUX4 gene. DUX4 inhibition has therefore emerged as a promising therapeutic strategy for FSHD. In this study, we tested a preclinical RNA interference (RNAi)-based DUX4 gene silencing approach as a prospective treatment for FSHD. We found that adeno-associated viral (AAV) vector-delivered therapeutic microRNAs corrected DUX4-associated myopathy in mouse muscle. These results provide proof-of-principle for RNAi therapy of FSHD through DUX4 inhibition.


Journal of Biological Chemistry | 2010

PP2A:B56ϵ, a Substrate of Caspase-3, Regulates p53-dependent and p53-independent Apoptosis during Development

Zhigang Jin; Lindsay M. Wallace; Scott Q. Harper; Jing Yang

Protein phosphatase 2A (PP2A) is one of the most abundantly expressed serine/threonine protein phosphatases. A large body of evidence suggests that PP2A is a tumor suppressor and plays critical roles in regulating apoptosis. PP2A is a heterotrimeric protein complex. Its substrate specificity, localization, and activity are regulated by regulatory subunits of PP2A. A recent study has demonstrated that single nucleotide polymorphism in B56ϵ (PPP2R5E), a B56 family regulatory subunit of PP2A, is associated with human soft tissue sarcoma. This raises the possibility that B56ϵ is involved in tumorigenesis and plays important roles in regulating apoptosis. However, this hypothesis has not been tested experimentally. Our previous studies revealed that B56ϵ regulates a number of developmental signaling pathways during early embryonic patterning. Here we report novel functions of B56ϵ in regulating apoptosis. We provide evidence that B56ϵ has both anti- and pro-apoptotic functions. B56ϵ suppresses p53-independent apoptosis during neural development, but triggers p53-dependent apoptosis. Mechanistically, B56ϵ regulates the p53-dependent apoptotic pathway solely through controlling the stability of p53 protein. In addition to its function in regulating apoptosis, we show that B56ϵ undergoes proteolytic cleavage. The cleavage of B56ϵ is mediated by caspase-3 and occurs on the carboxyl side of an evolutionarily conserved N-terminal “DKXD” motif. These results demonstrate that B56ϵ, a substrate of caspase-3, is an essential regulator of apoptosis. So far, we have identified an alternative translation isoform and a caspase cleavage product of B56ϵ. The significance of post-transcriptional regulation of B56ϵ is discussed.


Molecular Therapy | 2011

RNA Interference Improves Myopathic Phenotypes in Mice Over-expressing FSHD Region Gene 1 (FRG1)

Lindsay M. Wallace; Sara E Garwick-Coppens; Rossella Tupler; Scott Q. Harper

Muscular dystrophies, and other diseases of muscle, arise from recessive and dominant gene mutations. Gene replacement strategies may be beneficial for the former, while gene silencing approaches may provide treatment for the latter. In the last two decades, muscle-directed gene therapies were primarily focused on treating recessive disorders. This disparity at least partly arose because feasible mechanisms to silence dominant disease genes lagged behind gene replacement strategies. With the discovery of RNA interference (RNAi) and its subsequent development as a promising new gene silencing tool, the landscape has changed. In this study, our objective was to demonstrate proof-of-principle for RNAi therapy of a dominant myopathy in vivo. We tested the potential of adeno-associated viral (AAV)-delivered therapeutic microRNAs, targeting the human Facioscapulohumeral muscular dystrophy (FSHD) region gene 1 (FRG1), to correct myopathic features in mice expressing toxic levels of human FRG1 (FRG1(-high) mice). We found that FRG1 gene silencing improved muscle mass, strength, and histopathological abnormalities associated with muscular dystrophy in FRG1(-high) mice, thereby demonstrating therapeutic promise for treatment of dominantly inherited myopathies using RNAi. This approach potentially applies to as many as 29 different gene mutations responsible for myopathies inherited as dominant disorders.


Archive | 2010

RNAi Therapy for Dominant Muscular Dystrophies and Other Myopathies

Lindsay M. Wallace; Sara E. Garwick; Scott Q. Harper

Over the last ∼15 years, muscular dystrophy gene therapy strategies have been primarily aimed at replacing defective or missing genes underlying recessive disorders, such as Duchenne muscular dystrophy. These gene replacement strategies are typically not indicated for treating dominant diseases; instead, patients bearing dominant mutations would likely benefit from reduction or elimination of the abnormal allele. Until very recently, there was no feasible mechanism to reduce or eliminate disease genes, and molecular therapy development for dominant muscular dystrophies was largely unexplored. RNA interference (RNAi) has recently emerged as a powerful tool to suppress any gene of interest in a sequence-specific manner. As such, RNAi is a leading candidate strategy to silence dominant disease genes, including those involved in muscular dystrophy and related myopathies. Here, we discuss the potential for RNAi-mediated gene therapy of dominant muscular dystrophies and other myopathies.


Molecular therapy. Nucleic acids | 2014

RNAi-mediated Gene Silencing of Mutant Myotilin Improves Myopathy in LGMD1A Mice

Jian Liu; Lindsay M. Wallace; Sara E Garwick-Coppens; Darcée D. Sloboda; Carol S. Davis; Chady H. Hakim; Michael A. Hauser; Susan V. Brooks; Scott Q. Harper

Recent progress suggests gene therapy may one day be an option for treating some forms of limb girdle muscular dystrophy (LGMD). Nevertheless, approaches targeting LGMD have so far focused on gene replacement strategies for recessive forms of the disease. In contrast, no attempts have been made to develop molecular therapies for any of the eight dominantly inherited forms of LGMD. Importantly, the emergence of RNA interference (RNAi) therapeutics in the last decade provided new tools to combat dominantly inherited LGMDs with molecular therapy. In this study, we describe the first RNAi-based, preclinical gene therapy approach for silencing a gene associated with dominant LGMD. To do this, we developed adeno-associated viral vectors (AAV6) carrying designed therapeutic microRNAs targeting mutant myotilin (MYOT), which is the underlying cause of LGMD type 1A (LGMD1A). Our best MYOT-targeted microRNA vector (called miMYOT) significantly reduced mutant myotilin mRNA and soluble protein expression in muscles of LGMD1A mice (the TgT57I model) both 3 and 9 months after delivery, demonstrating short- and long-term silencing effects. This MYOT gene silencing subsequently decreased deposition of MYOT-seeded intramuscular protein aggregates, which is the hallmark feature of LGMD1A. Histological improvements were accompanied by significant functional correction, as miMYOT-treated animals showed increased muscle weight and improved specific force in the gastrocnemius, which is one of the most severely affected muscles in TgT57I mice and patients with dominant myotilin mutations. These promising results in a preclinical model of LGMD1A support the further development of RNAi-based molecular therapy as a prospective treatment for LGMD1A. Furthermore, this study sets a foundation that may be refined and adapted to treat other dominant LGMD and related disorders.


Molecular therapy. Nucleic acids | 2013

Dose-dependent Toxicity of Humanized Renilla reniformis GFP (hrGFP) Limits Its Utility as a Reporter Gene in Mouse Muscle.

Lindsay M. Wallace; Andrew Moreo; K. Reed Clark; Scott Q. Harper

Gene therapy has historically focused on delivering protein-coding genes to target cells or tissues using a variety of vectors. In recent years, the field has expanded to include gene-silencing strategies involving delivery of noncoding inhibitory RNAs, such as short hairpin RNAs or microRNAs (miRNAs). Often called RNA interference (RNAi) triggers, these small inhibitory RNAs are difficult or impossible to visualize in living cells or tissues. To circumvent this detection problem and ensure efficient delivery in preclinical studies, vectors can be engineered to coexpress a fluorescent reporter gene to serve as a marker of transduction. In this study, we set out to optimize adeno-associated viral (AAV) vectors capable of delivering engineered miRNAs and green fluorescent protein (GFP) reporter genes to skeletal muscle. Although the more broadly utilized enhanced GFP (eGFP) gene derived from the jellyfish, Aequorea victoria was a conventional choice, we were concerned about some previous studies suggesting this protein was myotoxic. We thus opted to test vectors carrying the humanized Renilla reniformis-derived GFP (hrGFP) gene, which has not seen as extensive usage as eGFP but was purported to be a safer and less cytotoxic alternative. Employing AAV6 vector dosages typically used in preclinical gene transfer studies (3×1010 –1 × 1011 particles), we found that hrGFP caused dose-dependent myopathy when delivered to wild-type (wt) mouse muscle, whereas identical titers of AAV6 carrying eGFP were relatively benign. Dose de-escalation at or below 8 × 109 AAV particles effectively reduced or eliminated hrGFP-associated myotoxicity, but also had dampening effects on green fluorescence and miRNA-mediated gene silencing in whole muscles. We conclude that hrGFP is impractical for use as a transduction marker in preclinical, AAV-based RNA interference therapy studies where adult mouse muscle is the target organ. Moreover, our data support that eGFP is superior to hrGFP as a reporter gene in mouse muscle. These results may impact the design of future preclinical gene therapy studies targeting muscles and non-muscle tissues alike.


Human Molecular Genetics | 2016

Mouse Dux is myotoxic and shares partial functional homology with its human paralog DUX4

Jocelyn O. Eidahl; Carlee R. Giesige; Jacqueline S. Domire; Lindsay M. Wallace; Allison M. Fowler; S. Guckes; Sara E Garwick-Coppens; Paul Labhart; Scott Q. Harper

Abstract D4Z4 repeats are present in at least 11 different mammalian species, including humans and mice. Each repeat contains an open reading frame encoding a double homeodomain (DUX) family transcription factor. Aberrant expression of the D4Z4 ORF called DUX4 is associated with the pathogenesis of Facioscapulohumeral muscular dystrophy (FSHD). DUX4 is toxic to numerous cell types of different species, and over-expression caused dysmorphism and developmental arrest in frogs and zebrafish, embryonic lethality in transgenic mice, and lesions in mouse muscle. Because DUX4 is a primate-specific gene, questions have been raised about the biological relevance of over-expressing it in non-primate models, as DUX4 toxicity could be related to non-specific cellular stress induced by over-expressing a DUX family transcription factor in organisms that did not co-evolve its regulated transcriptional networks. We assessed toxic phenotypes of DUX family genes, including DUX4, DUX1, DUX5, DUXA, DUX4-s, Dux-bl and mouse Dux. We found that DUX proteins were not universally toxic, and only the mouse Dux gene caused similar toxic phenotypes as human DUX4. Using RNA-seq, we found that 80% of genes upregulated by Dux were similarly increased in DUX4-expressing cells. Moreover, 43% of Dux-responsive genes contained ChIP-seq binding sites for both Dux and DUX4, and both proteins had similar consensus binding site sequences. These results suggested DUX4 and Dux may regulate some common pathways, and despite diverging from a common progenitor under different selective pressures for millions of years, the two genes maintain partial functional homology.


Archive | 2011

Rapid Cloning and Validation of MicroRNA Shuttle Vectors: A Practical Guide

Ryan L. Boudreau; Sara E Garwick-Coppens; Jian Liu; Lindsay M. Wallace; Scott Q. Harper

MicroRNAs (miRNAs) have emerged as important modulators of eukaryotic gene expression through a process called RNA interference (RNAi). Over the last several years, a large amount of work has focused on understanding how miRNAs are expressed and processed to a biologically functional form. This knowledge has enabled the development of RNAi as a molecular tool for investigating basic biological questions or as a therapeutic technique. Artificial miRNA shuttle vectors can be engineered to mimic natural miRNAs and subsequently used to suppress any gene of interest. Here, we describe a simple method to build and functionally validate artificial miRNA shuttles.


PLOS ONE | 2015

Aberrant splicing in transgenes containing introns, exons, and V5 epitopes: lessons from developing an FSHD mouse model expressing a D4Z4 repeat with flanking genomic sequences.

Eugénie Ansseau; Jacqueline S. Domire; Lindsay M. Wallace; Jocelyn O. Eidahl; S. Guckes; Carlee R. Giesige; Nettie K. Pyne; Alexandra Belayew; Scott Q. Harper

The DUX4 gene, encoded within D4Z4 repeats on human chromosome 4q35, has recently emerged as a key factor in the pathogenic mechanisms underlying Facioscapulohumeral muscular dystrophy (FSHD). This recognition prompted development of animal models expressing the DUX4 open reading frame (ORF) alone or embedded within D4Z4 repeats. In the first published model, we used adeno-associated viral vectors (AAV) and strong viral control elements (CMV promoter, SV40 poly A) to demonstrate that the DUX4 cDNA caused dose-dependent toxicity in mouse muscles. As a follow-up, we designed a second generation of DUX4-expressing AAV vectors to more faithfully genocopy the FSHD-permissive D4Z4 repeat region located at 4q35. This new vector (called AAV.D4Z4.V5.pLAM) contained the D4Z4/DUX4 promoter region, a V5 epitope-tagged DUX4 ORF, and the natural 3’ untranslated region (pLAM) harboring two small introns, DUX4 exons 2 and 3, and the non-canonical poly A signal required for stabilizing DUX4 mRNA in FSHD. AAV.D4Z4.V5.pLAM failed to recapitulate the robust pathology of our first generation vectors following delivery to mouse muscle. We found that the DUX4.V5 junction sequence created an unexpected splice donor in the pre-mRNA that was preferentially utilized to remove the V5 coding sequence and DUX4 stop codon, yielding non-functional DUX4 protein with 55 additional residues on its carboxyl-terminus. Importantly, we further found that aberrant splicing could occur in any expression construct containing a functional splice acceptor and sequences resembling minimal splice donors. Our findings represent an interesting case study with respect to AAV.D4Z4.V5.pLAM, but more broadly serve as a note of caution for designing constructs containing V5 epitope tags and/or transgenes with downstream introns and exons.

Collaboration


Dive into the Lindsay M. Wallace's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sara E Garwick-Coppens

The Research Institute at Nationwide Children's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jian Liu

The Research Institute at Nationwide Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

S. Guckes

The Research Institute at Nationwide Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Jocelyn O. Eidahl

The Research Institute at Nationwide Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

S.Q. Harper

The Research Institute at Nationwide Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Allison M. Fowler

The Research Institute at Nationwide Children's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nettie K. Pyne

The Research Institute at Nationwide Children's Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge