Network


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

Hotspot


Dive into the research topics where John R. McAnally is active.

Publication


Featured researches published by John R. McAnally.


Science | 2016

Postnatal genome editing partially restores dystrophin expression in a mouse model of muscular dystrophy

Chengzu Long; Leonela Amoasii; Alex A. Mireault; John R. McAnally; Hui Li; Efrain Sanchez-Ortiz; Samadrita Bhattacharyya; John M. Shelton; Rhonda Bassel-Duby; Eric N. Olson

Editing can help build stronger muscles Much of the controversy surrounding the gene-editing technology called CRISPR/Cas9 centers on the ethics of germline editing of human embryos to correct disease-causing mutations. For certain disorders such as muscular dystrophy, it may be possible to achieve therapeutic benefit by editing the faulty gene in somatic cells. In proof-of-concept studies, Long et al., Nelson et al., and Tabebordbar et al. used adeno-associated virus-9 to deliver the CRISPR/Cas9 gene-editing system to young mice with a mutation in the gene coding for dystrophin, a muscle protein deficient in patients with Duchenne muscular dystrophy. Gene editing partially restored dystrophin protein expression in skeletal and cardiac muscle and improved skeletal muscle function. Science, this issue p. 400, p. 403, p. 407 Gene editing via CRISPR-Cas9 restores dystrophin protein and improves muscle function in mouse models of muscular dystrophy. CRISPR/Cas9-mediated genome editing holds clinical potential for treating genetic diseases, such as Duchenne muscular dystrophy (DMD), which is caused by mutations in the dystrophin gene. To correct DMD by skipping mutant dystrophin exons in postnatal muscle tissue in vivo, we used adeno-associated virus–9 (AAV9) to deliver gene-editing components to postnatal mdx mice, a model of DMD. Different modes of AAV9 delivery were systematically tested, including intraperitoneal at postnatal day 1 (P1), intramuscular at P12, and retro-orbital at P18. Each of these methods restored dystrophin protein expression in cardiac and skeletal muscle to varying degrees, and expression increased from 3 to 12 weeks after injection. Postnatal gene editing also enhanced skeletal muscle function, as measured by grip strength tests 4 weeks after injection. This method provides a potential means of correcting mutations responsible for DMD and other monogenic disorders after birth.


Science | 2014

Prevention of muscular dystrophy in mice by CRISPR/Cas9–mediated editing of germline DNA

Chengzu Long; John R. McAnally; John M. Shelton; Alex A. Mireault; Rhonda Bassel-Duby; Eric N. Olson

Genome editing corrects a muscle disease Patients with Duchenne muscular dystrophy find their muscles growing progressively weaker. Studies identified dystrophin as the culprit gene, which galvanized research into gene-targeted therapies. Long et al. apply genome editing to “correct” the disease-causing mutation in mice genetically destined to develop the disease. This germline editing strategy kept muscles from degenerating, even in mice harboring only a small percentage of corrected cells. Although not feasible for humans, this proof of concept sets the stage for applying genome editing to specific cell types involved in the disease. Science, this issue p. 1184 A mutation that causes muscular dystrophy in mice can be corrected by genome editing, which prevents the disease from developing. Duchenne muscular dystrophy (DMD) is an inherited X-linked disease caused by mutations in the gene encoding dystrophin, a protein required for muscle fiber integrity. DMD is characterized by progressive muscle weakness and a shortened life span, and there is no effective treatment. We used clustered regularly interspaced short palindromic repeat/Cas9 (CRISPR/Cas9)–mediated genome editing to correct the dystrophin gene (Dmd) mutation in the germ line of mdx mice, a model for DMD, and then monitored muscle structure and function. Genome editing produced genetically mosaic animals containing 2 to 100% correction of the Dmd gene. The degree of muscle phenotypic rescue in mosaic mice exceeded the efficiency of gene correction, likely reflecting an advantage of the corrected cells and their contribution to regenerating muscle. With the anticipated technological advances that will facilitate genome editing of postnatal somatic cells, this strategy may one day allow correction of disease-causing mutations in the muscle tissue of patients with DMD.


Cell | 2015

A Micropeptide Encoded by a Putative Long Noncoding RNA Regulates Muscle Performance

Douglas M. Anderson; Kelly M. Anderson; Chi-Lun Chang; Catherine A. Makarewich; Benjamin R. Nelson; John R. McAnally; Prasad Kasaragod; John M. Shelton; Jen Liou; Rhonda Bassel-Duby; Eric N. Olson

Functional micropeptides can be concealed within RNAs that appear to be noncoding. We discovered a conserved micropeptide, which we named myoregulin (MLN), encoded by a skeletal muscle-specific RNA annotated as a putative long noncoding RNA. MLN shares structural and functional similarity with phospholamban (PLN) and sarcolipin (SLN), which inhibit SERCA, the membrane pump that controls muscle relaxation by regulating Ca(2+) uptake into the sarcoplasmic reticulum (SR). MLN interacts directly with SERCA and impedes Ca(2+) uptake into the SR. In contrast to PLN and SLN, which are expressed in cardiac and slow skeletal muscle in mice, MLN is robustly expressed in all skeletal muscle. Genetic deletion of MLN in mice enhances Ca(2+) handling in skeletal muscle and improves exercise performance. These findings identify MLN as an important regulator of skeletal muscle physiology and highlight the possibility that additional micropeptides are encoded in the many RNAs currently annotated as noncoding.


Science | 2016

A peptide encoded by a transcript annotated as long noncoding RNA enhances SERCA activity in muscle

Benjamin R. Nelson; Catherine A. Makarewich; Douglas M. Anderson; Benjamin R. Winders; Constantine D. Troupes; Fenfen Wu; Austin L Reese; John R. McAnally; Xiongwen Chen; Ege T. Kavalali; Stephen C. Cannon; Steven R. Houser; Rhonda Bassel-Duby; Eric N. Olson

Another micropeptide flexes its muscle Genome annotation is a complex but imperfect art. Attesting to its limitations is the growing evidence that certain transcripts annotated as long noncoding RNAs (lncRNAs) in fact code for small peptides with biologically important functions. One such lncRNA-derived micropeptide in mammals is myoregulin, which reduces muscle performance by inhibiting the activity of a key calcium pump. Nelson et al. describe the opposite activity in a second lncRNA-derived micropeptide in mammalian muscle, called DWORF (see the Perspective by Payre and Desplan). This peptide enhances muscle performance by activating the same calcium pump. DWORF may prove to be useful in improving the cardiac muscle function of mammals with heart disease. Science, this issue p. 271; see also p. 226 A long noncoding RNA encodes a small peptide that activates a calcium pump regulating muscle contraction. [Also see Perspective by Payre and Desplan] Muscle contraction depends on release of Ca2+ from the sarcoplasmic reticulum (SR) and reuptake by the Ca2+adenosine triphosphatase SERCA. We discovered a putative muscle-specific long noncoding RNA that encodes a peptide of 34 amino acids and that we named dwarf open reading frame (DWORF). DWORF localizes to the SR membrane, where it enhances SERCA activity by displacing the SERCA inhibitors, phospholamban, sarcolipin, and myoregulin. In mice, overexpression of DWORF in cardiomyocytes increases peak Ca2+ transient amplitude and SR Ca2+ load while reducing the time constant of cytosolic Ca2+ decay during each cycle of contraction-relaxation. Conversely, slow skeletal muscle lacking DWORF exhibits delayed Ca2+ clearance and relaxation and reduced SERCA activity. DWORF is the only endogenous peptide known to activate the SERCA pump by physical interaction and provides a means for enhancing muscle contractility.


Nature | 2016

Transcription of the non-coding RNA upperhand controls Hand2 expression and heart development

Kelly M. Anderson; Douglas M. Anderson; John R. McAnally; John M. Shelton; Rhonda Bassel-Duby; Eric N. Olson

HAND2 is an ancestral regulator of heart development and one of four transcription factors that control the reprogramming of fibroblasts into cardiomyocytes. Deletion of Hand2 in mice results in right ventricle hypoplasia and embryonic lethality. Hand2 expression is tightly regulated by upstream enhancers that reside within a super-enhancer delineated by histone H3 acetyl Lys27 (H3K27ac) modifications. Here we show that transcription of a Hand2-associated long non-coding RNA, which we named upperhand (Uph), is required to maintain the super-enhancer signature and elongation of RNA polymerase II through the Hand2 enhancer locus. Blockade of Uph transcription, but not knockdown of the mature transcript, abolished Hand2 expression, causing right ventricular hypoplasia and embryonic lethality in mice. Given the substantial number of uncharacterized promoter-associated long non-coding RNAs encoded by the mammalian genome, the Uph–Hand2 regulatory partnership offers a mechanism by which divergent non-coding transcription can establish a permissive chromatin environment.


Science Advances | 2017

CRISPR-Cpf1 correction of muscular dystrophy mutations in human cardiomyocytes and mice

Yu Zhang; Chengzu Long; Hui Li; John R. McAnally; Kedryn K. Baskin; John M. Shelton; Rhonda Bassel-Duby; Eric N. Olson

CRISPR-Cpf1–mediated correction of Duchenne muscular dystrophy mutations in human cells and a mouse model. Duchenne muscular dystrophy (DMD), caused by mutations in the X-linked dystrophin gene (DMD), is characterized by fatal degeneration of striated muscles. Dilated cardiomyopathy is one of the most common lethal features of the disease. We deployed Cpf1, a unique class 2 CRISPR (clustered regularly interspaced short palindromic repeats) effector, to correct DMD mutations in patient-derived induced pluripotent stem cells (iPSCs) and mdx mice, an animal model of DMD. Cpf1-mediated genomic editing of human iPSCs, either by skipping of an out-of-frame DMD exon or by correcting a nonsense mutation, restored dystrophin expression after differentiation to cardiomyocytes and enhanced contractile function. Similarly, pathophysiological hallmarks of muscular dystrophy were corrected in mdx mice following Cpf1-mediated germline editing. These findings are the first to show the efficiency of Cpf1-mediated correction of genetic mutations in human cells and an animal disease model and represent a significant step toward therapeutic translation of gene editing for correction of DMD.


Science | 2017

Control of muscle formation by the fusogenic micropeptide myomixer

Pengpeng Bi; Andres Ramirez-Martinez; Hui Li; Jessica Cannavino; John R. McAnally; John M. Shelton; Efrain Sanchez-Ortiz; Rhonda Bassel-Duby; Eric N. Olson

Micromanaging muscle cell fusion Adult skeletal muscles are characterized by long, multinucleated cells called myofibers. Myofibers form when muscle precursor cells, or myoblasts, differentiate and fuse together during embryogenesis. The fusion process is not fully understood. Studying cell culture and mouse models, Bi et al. identified an 84–amino acid peptide that promotes myoblast fusion. This small peptide, called Myomixer, physically interacts with and stimulates the activity of a fusogenic membrane protein called Myomaker. Notably, the Myomaker-Myomixer pair can also promote the fusion of nonmuscle cells, such as fibroblasts. Science, this issue p. 323 A small peptide expressed in developing skeletal muscle controls muscle cell fusion and myofiber formation. Skeletal muscle formation occurs through fusion of myoblasts to form multinucleated myofibers. From a genome-wide clustered regularly interspaced short palindromic repeats (CRISPR) loss-of-function screen for genes required for myoblast fusion and myogenesis, we discovered an 84–amino acid muscle-specific peptide that we call Myomixer. Myomixer expression coincides with myoblast differentiation and is essential for fusion and skeletal muscle formation during embryogenesis. Myomixer localizes to the plasma membrane, where it promotes myoblast fusion and associates with Myomaker, a fusogenic membrane protein. Myomixer together with Myomaker can also induce fibroblast-fibroblast fusion and fibroblast-myoblast fusion. We conclude that the Myomixer-Myomaker pair controls the critical step in myofiber formation during muscle development.


Journal of Clinical Investigation | 2015

Severe myopathy in mice lacking the MEF2/SRF-dependent gene leiomodin-3

Bercin Kutluk Cenik; Ankit Garg; John R. McAnally; John M. Shelton; James A. Richardson; Rhonda Bassel-Duby; Eric N. Olson; Ning Liu

Maintenance of skeletal muscle structure and function requires a precise stoichiometry of sarcomeric proteins for proper assembly of the contractile apparatus. Absence of components of the sarcomeric thin filaments causes nemaline myopathy, a lethal congenital muscle disorder associated with aberrant myofiber structure and contractility. Previously, we reported that deficiency of the kelch-like family member 40 (KLHL40) in mice results in nemaline myopathy and destabilization of leiomodin-3 (LMOD3). LMOD3 belongs to a family of tropomodulin-related proteins that promote actin nucleation. Here, we show that deficiency of LMOD3 in mice causes nemaline myopathy. In skeletal muscle, transcription of Lmod3 was controlled by the transcription factors SRF and MEF2. Myocardin-related transcription factors (MRTFs), which function as SRF coactivators, serve as sensors of actin polymerization and are sequestered in the cytoplasm by actin monomers. Conversely, conditions that favor actin polymerization de-repress MRTFs and activate SRF-dependent genes. We demonstrated that the actin nucleator LMOD3, together with its stabilizing partner KLHL40, enhances MRTF-SRF activity. In turn, SRF cooperated with MEF2 to sustain the expression of LMOD3 and other components of the contractile apparatus, thereby establishing a regulatory circuit to maintain skeletal muscle function. These findings provide insight into the molecular basis of the sarcomere assembly and muscle dysfunction associated with nemaline myopathy.


Science Translational Medicine | 2017

Single-cut genome editing restores dystrophin expression in a new mouse model of muscular dystrophy

Leonela Amoasii; Chengzu Long; Hui Li; Alex A. Mireault; John M. Shelton; Efrain Sanchez-Ortiz; John R. McAnally; Samadrita Bhattacharyya; Florian Schmidt; Dirk Grimm; Stephen D. Hauschka; Rhonda Bassel-Duby; Eric N. Olson

Single-cut correction of a dystrophin gene mutation with CRISPR/Cas9 restored dystrophin expression in skeletal and cardiac muscles in a mouse model of Duchenne muscular dystrophy. Making the cut Mutations in the dystrophin gene cause Duchenne muscular dystrophy (DMD), a fatal childhood muscle disease. To optimize the correction of DMD mutations by CRISPR/Cas9 gene editing, Amoasii et al. first generated mice that had exon 50 deleted, a common human mutational “hotspot” region of the dystrophin gene. The authors then reported a method in which a single cut in genomic DNA encoding dystrophin with CRISPR/Cas9 in these engineered mice resulted in up to 90% restoration of dystrophin expression in mouse skeletal and heart muscles. This method of permanently bypassing DMD mutations using a single genomic cut suggests that this strategy may have potential for efficiently correcting DMD mutations. Duchenne muscular dystrophy (DMD) is a severe, progressive muscle disease caused by mutations in the dystrophin gene. The majority of DMD mutations are deletions that prematurely terminate the dystrophin protein. Deletions of exon 50 of the dystrophin gene are among the most common single exon deletions causing DMD. Such mutations can be corrected by skipping exon 51, thereby restoring the dystrophin reading frame. Using clustered regularly interspaced short palindromic repeats/CRISPR-associated 9 (CRISPR/Cas9), we generated a DMD mouse model by deleting exon 50. These ΔEx50 mice displayed severe muscle dysfunction, which was corrected by systemic delivery of adeno-associated virus encoding CRISPR/Cas9 genome editing components. We optimized the method for dystrophin reading frame correction using a single guide RNA that created reframing mutations and allowed skipping of exon 51. In conjunction with muscle-specific expression of Cas9, this approach restored up to 90% of dystrophin protein expression throughout skeletal muscles and the heart of ΔEx50 mice. This method of permanently bypassing DMD mutations using a single cut in genomic DNA represents a step toward clinical correction of DMD mutations and potentially those of other neuromuscular disorders.


Proceedings of the National Academy of Sciences of the United States of America | 2018

Fusogenic micropeptide Myomixer is essential for satellite cell fusion and muscle regeneration

Pengpeng Bi; John R. McAnally; John M. Shelton; Efrain Sanchez-Ortiz; Rhonda Bassel-Duby; Eric N. Olson

Significance Skeletal muscle damaged by injury or disease can regenerate new muscle fibers. The regenerative properties of skeletal muscle involve fusion of activated muscle stem cells (satellite cells). We recently discovered Myomixer, a conserved micropeptide that is specifically expressed during muscle formation. Myomixer, together with its partner Myomaker, another muscle-specific membrane protein, is necessary for muscle formation during embryogenesis. Here, we show the absolute requirement of Myomixer for the fusion of satellite cells and regeneration of adult muscle in response to injury. Our findings provide insights into the mechanisms of muscle formation and suggest opportunities for enhancing muscle regeneration through manipulation of Myomixer and Myomaker. Regeneration of skeletal muscle in response to injury occurs through fusion of a population of stem cells, known as satellite cells, with injured myofibers. Myomixer, a muscle-specific membrane micropeptide, cooperates with the transmembrane protein Myomaker to regulate embryonic myoblast fusion and muscle formation. To investigate the role of Myomixer in muscle regeneration, we used CRISPR/Cas9-mediated genome editing to generate conditional knockout Myomixer alleles in mice. We show that genetic deletion of Myomixer in satellite cells using a tamoxifen-regulated Cre recombinase transgene under control of the Pax7 promoter abolishes satellite cell fusion and prevents muscle regeneration, resulting in severe muscle degeneration after injury. Satellite cells devoid of Myomixer maintain expression of Myomaker, demonstrating that Myomaker alone is insufficient to drive myoblast fusion. These findings, together with prior studies demonstrating the essentiality of Myomaker for muscle regeneration, highlight the obligatory partnership of Myomixer and Myomaker for myofiber formation throughout embryogenesis and adulthood.

Collaboration


Dive into the John R. McAnally's collaboration.

Top Co-Authors

Avatar

Eric N. Olson

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Rhonda Bassel-Duby

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

John M. Shelton

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Catherine A. Makarewich

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Douglas M. Anderson

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Benjamin R. Nelson

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Chengzu Long

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar

Efrain Sanchez-Ortiz

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Hui Li

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Alex A. Mireault

University of Texas Southwestern Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge