Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
JACC Basic Transl Sci ; 9(3): 380-395, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38559624

ABSTRACT

To solve the clinical transformation dilemma of lamin A/C (LMNA)-mutated dilated cardiomyopathy (LMD), we developed an LMNA-mutated primate model based on the similarity between the phenotype of primates and humans. We screened out patients with LMD and compared the clinical data of LMD with TTN-mutated and mutation-free dilated cardiomyopathy to obtain the unique phenotype. After establishment of the LMNA c.357-2A>G primate model, primates were continuously observed for 48 months, and echocardiographic, electrophysiological, histologic, and transcriptional data were recorded. The LMD primate model was found to highly simulate the phenotype of clinical LMD. In addition, the LMD primate model shared a similar natural history with humans.

2.
bioRxiv ; 2024 Jan 18.
Article in English | MEDLINE | ID: mdl-38293034

ABSTRACT

Altered protein conformation can cause incurable neurodegenerative disorders. Mutations in SERPINI1 , the gene encoding neuroserpin, alter protein conformation resulting in cytotoxic aggregation in neuronal endoplasmic reticulum. Aggregates cause oxidative stress impairing function, leading to neuronal death. Familial encephalopathy with neuroserpin inclusion bodies (FENIB) is a rare autosomal dominant progressive myoclonic epilepsy. Patients present with seizures and cognitive impairments that progress to dementia and premature death. We developed HEK293T and induced pluripotent stem cell (iPSC) models of FENIB, harboring the patient's pathogenic SERPINI1 variant or stably overexpressing mutant neuroserpin fused to GFP (MUT NS-GFP). FENIB cells form neuroserpin inclusions which increase in size and number. Here, we utilized a personalized adenine base editor (ABE)-mediated approach to efficiently correct the pathogenic variant and to restore neuronal dendritic morphology. ABE-treated MUT NS-GFP cells demonstrated reduced inclusion size and number. Using an inducible MUT NS-GFP neuron system, we identified early prevention of toxic protein expression allowed aggregate clearance, while late prevention halted neuronal impairments. To address several challenges for clinical applications of gene correction, we developed a neuron-specific engineered virus-like particle to optimize neuronal ABE delivery. Preventing mutant protein with altered conformation production improved toxic protein clearance. Our findings provide a targeted strategy and may treat FENIB and potentially other neurodegenerative diseases due to altered protein conformation such as Alzheimer's and Huntington's diseases.

3.
Zool Res ; 42(4): 469-477, 2021 Jul 18.
Article in English | MEDLINE | ID: mdl-34213093

ABSTRACT

Mutations of PTEN-induced kinase I (PINK1) cause early-onset Parkinson's disease (PD) with selective neurodegeneration in humans. However, current PINK1 knockout mouse and pig models are unable to recapitulate the typical neurodegenerative phenotypes observed in PD patients. This suggests that generating PINK1 disease models in non-human primates (NHPs) that are close to humans is essential to investigate the unique function of PINK1 in primate brains. Paired single guide RNA (sgRNA)/Cas9-D10A nickases and truncated sgRNA/Cas9, both of which can reduce off-target effects without compromising on-target editing, are two optimized strategies in the CRISPR/Cas9 system for establishing disease animal models. Here, we combined the two strategies and injected Cas9-D10A mRNA and two truncated sgRNAs into one-cell-stage cynomolgus zygotes to target the PINK1 gene. We achieved precise and efficient gene editing of the target site in three newborn cynomolgus monkeys. The frame shift mutations of PINK1 in mutant fibroblasts led to a reduction in mRNA. However, western blotting and immunofluorescence staining confirmed the PINK1 protein levels were comparable to that in wild-type fibroblasts. We further reprogramed mutant fibroblasts into induced pluripotent stem cells (iPSCs), which showed similar ability to differentiate into dopamine (DA) neurons. Taken together, our results showed that co-injection of Cas9-D10A nickase mRNA and sgRNA into one-cell-stage cynomolgus embryos enabled the generation of human disease models in NHPs and target editing by pair truncated sgRNA/Cas9-D10A in PINK1 gene exon 2 did not impact protein expression.


Subject(s)
Disease Models, Animal , Macaca fascicularis/genetics , Parkinson Disease/veterinary , Protein Kinases/metabolism , Animals , Animals, Newborn , CRISPR-Associated Protein 9/genetics , CRISPR-Associated Protein 9/metabolism , Embryo Culture Techniques , Embryo Transfer , Fibroblasts/physiology , Frameshift Mutation , Gene Expression Regulation , Macaca fascicularis/embryology , Monkey Diseases/genetics , Mutation , Parkinson Disease/genetics , Protein Kinases/genetics , RNA, Guide, Kinetoplastida
4.
Protein Cell ; 11(11): 809-824, 2020 11.
Article in English | MEDLINE | ID: mdl-32729022

ABSTRACT

Many human genetic diseases, including Hutchinson-Gilford progeria syndrome (HGPS), are caused by single point mutations. HGPS is a rare disorder that causes premature aging and is usually caused by a de novo point mutation in the LMNA gene. Base editors (BEs) composed of a cytidine deaminase fused to CRISPR/Cas9 nickase are highly efficient at inducing C to T base conversions in a programmable manner and can be used to generate animal disease models with single amino-acid substitutions. Here, we generated the first HGPS monkey model by delivering a BE mRNA and guide RNA (gRNA) targeting the LMNA gene via microinjection into monkey zygotes. Five out of six newborn monkeys carried the mutation specifically at the target site. HGPS monkeys expressed the toxic form of lamin A, progerin, and recapitulated the typical HGPS phenotypes including growth retardation, bone alterations, and vascular abnormalities. Thus, this monkey model genetically and clinically mimics HGPS in humans, demonstrating that the BE system can efficiently and accurately generate patient-specific disease models in non-human primates.


Subject(s)
Disease Models, Animal , Gene Editing , Lamin Type A , Progeria , Animals , Female , Humans , Lamin Type A/genetics , Lamin Type A/metabolism , Macaca fascicularis , Progeria/genetics , Progeria/metabolism , Progeria/pathology
5.
J Biomed Res ; 35(2): 148-162, 2020 Nov 27.
Article in English | MEDLINE | ID: mdl-33402545

ABSTRACT

There are an estimated 10 000 monogenic diseases affecting tens of millions of individuals worldwide. The application of CRISPR/Cas genome editing tools to treat monogenic diseases is an emerging strategy with the potential to generate personalized treatment approaches for these patients. CRISPR/Cas-based systems are programmable and sequence-specific genome editing tools with the capacity to generate base pair resolution manipulations to DNA or RNA. The complexity of genomic insults resulting in heritable disease requires patient-specific genome editing strategies with consideration of DNA repair pathways, and CRISPR/Cas systems of different types, species, and those with additional enzymatic capacity and/or delivery methods. In this review we aim to discuss broad and multifaceted therapeutic applications of CRISPR/Cas gene editing systems including in harnessing of homology directed repair, non-homologous end joining, microhomology-mediated end joining, and base editing to permanently correct diverse monogenic diseases.

6.
Nat Commun ; 10(1): 4537, 2019 10 04.
Article in English | MEDLINE | ID: mdl-31586095

ABSTRACT

Duchenne muscular dystrophy (DMD) is a fatal genetic disorder caused by mutations in the dystrophin gene. To enable the non-invasive analysis of DMD gene correction strategies in vivo, we introduced a luciferase reporter in-frame with the C-terminus of the dystrophin gene in mice. Expression of this reporter mimics endogenous dystrophin expression and DMD mutations that disrupt the dystrophin open reading frame extinguish luciferase expression. We evaluated the correction of the dystrophin reading frame coupled to luciferase in mice lacking exon 50, a common mutational hotspot, after delivery of CRISPR/Cas9 gene editing machinery with adeno-associated virus. Bioluminescence monitoring revealed efficient and rapid restoration of dystrophin protein expression in affected skeletal muscles and the heart. Our results provide a sensitive non-invasive means of monitoring dystrophin correction in mouse models of DMD and offer a platform for testing different strategies for amelioration of DMD pathogenesis.


Subject(s)
Dystrophin/genetics , Genetic Therapy/methods , Intravital Microscopy/methods , Muscle, Skeletal/diagnostic imaging , Muscular Dystrophy, Duchenne/therapy , Animals , CRISPR-Cas Systems/genetics , Dependovirus/genetics , Disease Models, Animal , Dystrophin/metabolism , Exons/genetics , Gene Editing/methods , Genes, Reporter/genetics , Genetic Vectors/chemistry , Genetic Vectors/genetics , Humans , Luciferases/chemistry , Luciferases/genetics , Luminescent Measurements , Male , Mice , Mice, Transgenic , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/diagnostic imaging , Muscular Dystrophy, Duchenne/genetics , Mutation , Treatment Outcome
7.
Natl Sci Rev ; 6(3): 393, 2019 May.
Article in English | MEDLINE | ID: mdl-34691887
8.
Physiol Rev ; 98(3): 1205-1240, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29717930

ABSTRACT

Muscular dystrophies represent a large group of genetic disorders that significantly impair quality of life and often progress to premature death. There is no effective treatment for these debilitating diseases. Most therapies, developed to date, focus on alleviating the symptoms or targeting the secondary effects, while the underlying gene mutation is still present in the human genome. The discovery and application of programmable nucleases for site-specific DNA double-stranded breaks provides a powerful tool for precise genome engineering. In particular, the CRISPR/Cas system has revolutionized the genome editing field and is providing a new path for disease treatment by targeting the disease-causing genetic mutations. In this review, we provide a historical overview of genome-editing technologies, summarize the most recent advances, and discuss potential strategies and challenges for permanently correcting genetic mutations that cause muscular dystrophies.


Subject(s)
Gene Editing/methods , Genetic Therapy , Muscular Dystrophies/prevention & control , Animals , Disease Models, Animal , Humans , Muscular Dystrophy, Animal
9.
Sci Adv ; 4(1): eaap9004, 2018 01.
Article in English | MEDLINE | ID: mdl-29404407

ABSTRACT

Genome editing with CRISPR/Cas9 is a promising new approach for correcting or mitigating disease-causing mutations. Duchenne muscular dystrophy (DMD) is associated with lethal degeneration of cardiac and skeletal muscle caused by more than 3000 different mutations in the X-linked dystrophin gene (DMD). Most of these mutations are clustered in "hotspots." There is a fortuitous correspondence between the eukaryotic splice acceptor and splice donor sequences and the protospacer adjacent motif sequences that govern prokaryotic CRISPR/Cas9 target gene recognition and cleavage. Taking advantage of this correspondence, we screened for optimal guide RNAs capable of introducing insertion/deletion (indel) mutations by nonhomologous end joining that abolish conserved RNA splice sites in 12 exons that potentially allow skipping of the most common mutant or out-of-frame DMD exons within or nearby mutational hotspots. We refer to the correction of DMD mutations by exon skipping as myoediting. In proof-of-concept studies, we performed myoediting in representative induced pluripotent stem cells from multiple patients with large deletions, point mutations, or duplications within the DMD gene and efficiently restored dystrophin protein expression in derivative cardiomyocytes. In three-dimensional engineered heart muscle (EHM), myoediting of DMD mutations restored dystrophin expression and the corresponding mechanical force of contraction. Correcting only a subset of cardiomyocytes (30 to 50%) was sufficient to rescue the mutant EHM phenotype to near-normal control levels. We conclude that abolishing conserved RNA splicing acceptor/donor sites and directing the splicing machinery to skip mutant or out-of-frame exons through myoediting allow correction of the cardiac abnormalities associated with DMD by eliminating the underlying genetic basis of the disease.


Subject(s)
Gene Editing , Genome, Human , Muscular Dystrophy, Duchenne/genetics , Mutation/genetics , Myocardium/pathology , Tissue Engineering/methods , Base Sequence , Dystrophin/genetics , Exons/genetics , Humans , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/metabolism , RNA, Guide, Kinetoplastida/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
10.
Sci Transl Med ; 9(418)2017 Nov 29.
Article in English | MEDLINE | ID: mdl-29187645

ABSTRACT

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.


Subject(s)
Dystrophin/metabolism , Gene Editing/methods , Animals , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/physiology , Disease Models, Animal , Dystrophin/genetics , Exons/genetics , Mice , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/metabolism , Mutation/genetics
11.
JCI Insight ; 2(18)2017 09 21.
Article in English | MEDLINE | ID: mdl-28931764

ABSTRACT

Dystrophin maintains the integrity of striated muscles by linking the actin cytoskeleton with the cell membrane. Duchenne muscular dystrophy (DMD) is caused by mutations in the dystrophin gene (DMD) that result in progressive, debilitating muscle weakness, cardiomyopathy, and a shortened lifespan. Mutations of dystrophin that disrupt the amino-terminal actin-binding domain 1 (ABD-1), encoded by exons 2-8, represent the second-most common cause of DMD. In the present study, we compared three different strategies for CRISPR/Cas9 genome editing to correct mutations in the ABD-1 region of the DMD gene by deleting exons 3-9, 6-9, or 7-11 in human induced pluripotent stem cells (iPSCs) and by assessing the function of iPSC-derived cardiomyocytes. All three exon deletion strategies enabled the expression of truncated dystrophin protein and restoration of cardiomyocyte contractility and calcium transients to varying degrees. We show that deletion of exons 3-9 by genomic editing provides an especially effective means of correcting disease-causing ABD-1 mutations. These findings represent an important step toward eventual correction of common DMD mutations and provide a means of rapidly assessing the expression and function of internally truncated forms of dystrophin-lacking portions of ABD-1.


Subject(s)
Actins/metabolism , Dystrophin/metabolism , Gene Editing , Muscular Dystrophy, Duchenne/genetics , Mutation , CRISPR-Cas Systems , Dystrophin/genetics , Exons , Humans , Induced Pluripotent Stem Cells/cytology , Protein Binding , Protein Domains
12.
Sci Adv ; 3(4): e1602814, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28439558

ABSTRACT

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.


Subject(s)
CRISPR-Cas Systems , Dystrophin , Muscular Dystrophy, Animal/therapy , Muscular Dystrophy, Duchenne/therapy , Myocytes, Cardiac/metabolism , Animals , Dystrophin/genetics , Dystrophin/metabolism , Humans , Mice , Mice, Inbred mdx , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/metabolism , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology , Myocytes, Cardiac/pathology
13.
JAMA Neurol ; 73(11): 1349-1355, 2016 Nov 01.
Article in English | MEDLINE | ID: mdl-27668807

ABSTRACT

IMPORTANCE: Muscle weakness, the most common symptom of neuromuscular disease, may result from muscle dysfunction or may be caused indirectly by neuronal and neuromuscular junction abnormalities. To date, more than 780 monogenic neuromuscular diseases, linked to 417 different genes, have been identified in humans. Genome-editing methods, especially the CRISPR (clustered regularly interspaced short palindromic repeats)-Cas9 (CRISPR-associated protein 9) system, hold clinical potential for curing many monogenic disorders, including neuromuscular diseases such as Duchenne muscular dystrophy, spinal muscular atrophy, amyotrophic lateral sclerosis, and myotonic dystrophy type 1. OBJECTIVES: To provide an overview of genome-editing approaches; to summarize published reports on the feasibility, efficacy, and safety of current genome-editing methods as they relate to the potential correction of monogenic neuromuscular diseases; and to highlight scientific and clinical opportunities and obstacles toward permanent correction of disease-causing mutations responsible for monogenic neuromuscular diseases by genome editing. EVIDENCE REVIEW: PubMed and Google Scholar were searched for articles published from June 30, 1989, through June 9, 2016, using the following keywords: genome editing, CRISPR-Cas9, neuromuscular disease, Duchenne muscular dystrophy, spinal muscular atrophy, amyotrophic lateral sclerosis, and myotonic dystrophy type 1. The following sources were reviewed: 341 articles describing different approaches to edit mammalian genomes; 330 articles describing CRISPR-Cas9-mediated genome editing in cell culture lines (in vitro) and animal models (in vivo); 16 websites used to generate single-guide RNA; 4 websites for off-target effects; and 382 articles describing viral and nonviral delivery systems. Articles describing neuromuscular diseases, including Duchenne muscular dystrophy, spinal muscular atrophy, amyotrophic lateral sclerosis, and myotonic dystrophy type 1, were also reviewed. FINDINGS: Multiple proof-of-concept studies reveal the feasibility and efficacy of genome-editing-meditated correction of monogenic neuromuscular diseases in cultured cells and animal models. CONCLUSIONS AND RELEVANCE: Genome editing is a rapidly evolving technology with enormous translational potential once efficacy, delivery, and safety issues are addressed. The clinical impact of this technology is that genome editing can permanently correct disease-causing mutations and circumvent the hurdles of traditional gene- and cell-based therapies.


Subject(s)
Amyotrophic Lateral Sclerosis/therapy , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Gene Editing/methods , Genetic Therapy/methods , Muscular Atrophy, Spinal/therapy , Muscular Dystrophy, Duchenne/therapy , Myotonic Dystrophy/therapy , Amyotrophic Lateral Sclerosis/genetics , Animals , Humans , Muscular Atrophy, Spinal/genetics , Muscular Dystrophy, Duchenne/genetics , Myotonic Dystrophy/genetics
14.
Science ; 351(6271): 400-3, 2016 Jan 22.
Article in English | MEDLINE | ID: mdl-26721683

ABSTRACT

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.


Subject(s)
CRISPR-Cas Systems , Dystrophin/genetics , Genetic Therapy/methods , Muscular Dystrophy, Duchenne/therapy , Animals , Dependovirus , Disease Models, Animal , Exons/genetics , Female , Forelimb/physiopathology , Genome/genetics , Hand Strength , Male , Mice , Mice, Inbred mdx , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/genetics , Myocardium/metabolism
15.
Science ; 345(6201): 1184-1188, 2014 Sep 05.
Article in English | MEDLINE | ID: mdl-25123483

ABSTRACT

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.


Subject(s)
CRISPR-Cas Systems , Dystrophin/genetics , Gene Targeting/methods , Muscular Dystrophy, Duchenne/prevention & control , Animals , DNA/genetics , Exons/genetics , Genetic Therapy/methods , Germ Cells , Mice , Mice, Inbred mdx , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology
16.
Proc Natl Acad Sci U S A ; 111(31): 11521-6, 2014 Aug 05.
Article in English | MEDLINE | ID: mdl-25049392

ABSTRACT

Members of the calmodulin-binding transcription activator (CAMTA) family of proteins function as calcium-sensitive regulators of gene expression in multicellular organisms ranging from plants to humans. Here, we show that global or nervous system deletion of CAMTA1 in mice causes severe ataxia with Purkinje cell degeneration and cerebellar atrophy, partially resembling the consequences of haploinsufficiency of the human CAMTA1 locus. Gene-expression analysis identified a large collection of neuronal genes that were dysregulated in the brains of CAMTA1-mutant mice, and elucidation of a consensus sequence for binding of CAMTA proteins to DNA revealed the association of CAMTA-binding sites with many of these genes. We conclude that CAMTA1 plays an essential role in the control of Purkinje cell function and survival. CAMTA1-mutant mice provide a model to study the molecular mechanisms of neurodegenerative diseases and for screening potential therapeutic interventions for such disorders.


Subject(s)
Ataxia/metabolism , Ataxia/pathology , Calcium-Binding Proteins/deficiency , Purkinje Cells/metabolism , Purkinje Cells/pathology , Trans-Activators/deficiency , Transcription Factors/deficiency , AT Rich Sequence , Animals , Ataxia/physiopathology , Base Sequence , Binding Sites , Calcium-Binding Proteins/metabolism , Gene Expression Regulation , Integrases/metabolism , Inverted Repeat Sequences/genetics , Male , Mice , Mice, Knockout , Molecular Sequence Data , Motor Activity , Nestin/metabolism , Nucleotide Motifs/genetics , Protein Multimerization , Trans-Activators/metabolism , Transcription Factors/metabolism
17.
J Clin Invest ; 124(8): 3529-39, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24960163

ABSTRACT

Nemaline myopathy (NM) is a congenital myopathy that can result in lethal muscle dysfunction and is thought to be a disease of the sarcomere thin filament. Recently, several proteins of unknown function have been implicated in NM, but the mechanistic basis of their contribution to disease remains unresolved. Here, we demonstrated that loss of a muscle-specific protein, kelch-like family member 40 (KLHL40), results in a nemaline-like myopathy in mice that closely phenocopies muscle abnormalities observed in KLHL40-deficient patients. We determined that KLHL40 localizes to the sarcomere I band and A band and binds to nebulin (NEB), a protein frequently implicated in NM, as well as a putative thin filament protein, leiomodin 3 (LMOD3). KLHL40 belongs to the BTB-BACK-kelch (BBK) family of proteins, some of which have been shown to promote degradation of their substrates. In contrast, we found that KLHL40 promotes stability of NEB and LMOD3 and blocks LMOD3 ubiquitination. Accordingly, NEB and LMOD3 were reduced in skeletal muscle of both Klhl40-/- mice and KLHL40-deficient patients. Loss of sarcomere thin filament proteins is a frequent cause of NM; therefore, our data that KLHL40 stabilizes NEB and LMOD3 provide a potential basis for the development of NM in KLHL40-deficient patients.


Subject(s)
Muscle Proteins/deficiency , Myopathies, Nemaline/etiology , Myopathies, Nemaline/metabolism , Animals , Animals, Newborn , Cytoskeletal Proteins/metabolism , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Proteins/chemistry , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Myopathies, Nemaline/pathology , Protein Interaction Domains and Motifs , Protein Stability , Proteolysis , Sarcomeres/metabolism , Sarcomeres/pathology , Ubiquitination
18.
Science ; 328(5974): 94-8, 2010 Apr 02.
Article in English | MEDLINE | ID: mdl-20360108

ABSTRACT

Semiconservative DNA replication ensures the faithful duplication of genetic information during cell divisions. However, how epigenetic information carried by histone modifications propagates through mitotic divisions remains elusive. To address this question, the DNA replication-dependent nucleosome partition pattern must be clarified. Here, we report significant amounts of H3.3-H4 tetramers split in vivo, whereas most H3.1-H4 tetramers remained intact. Inhibiting DNA replication-dependent deposition greatly reduced the level of splitting events, which suggests that (i) the replication-independent H3.3 deposition pathway proceeds largely by cooperatively incorporating two new H3.3-H4 dimers and (ii) the majority of splitting events occurred during replication-dependent deposition. Our results support the idea that "silent" histone modifications within large heterochromatic regions are maintained by copying modifications from neighboring preexisting histones without the need for H3-H4 splitting events.


Subject(s)
Chromatin Assembly and Disassembly , DNA Replication , Histones/chemistry , Histones/metabolism , Nucleosomes/metabolism , Amino Acid Sequence , Aphidicolin/pharmacology , Cell Cycle , Chromatin/metabolism , Epigenesis, Genetic , HeLa Cells , Heterochromatin/metabolism , Humans , Hydroxyurea/pharmacology , Mass Spectrometry , Molecular Sequence Data , Protein Multimerization , S Phase , Transfection
19.
PLoS Pathog ; 5(7): e1000526, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19629177

ABSTRACT

Plant Resistance (R) proteins play an integral role in defense against pathogen infection. A unique gain-of-function mutation in the R gene SNC1, snc1, results in constitutive activation of plant immune pathways and enhanced resistance against pathogen infection. We previously found that mutations in MOS4 suppress the autoimmune phenotypes of snc1, and that MOS4 is part of a nuclear complex called the MOS4-Associated Complex (MAC) along with the transcription factor AtCDC5 and the WD-40 protein PRL1. Here we report the immuno-affinity purification of the MAC using HA-tagged MOS4 followed by protein sequence analysis by mass spectrometry. A total of 24 MAC proteins were identified, 19 of which have predicted roles in RNA processing based on their homology to proteins in the Prp19-Complex, an evolutionarily conserved spliceosome-associated complex containing homologs of MOS4, AtCDC5, and PRL1. Among these were two highly similar U-box proteins with homology to the yeast and human E3 ubiquitin ligase Prp19, which we named MAC3A and MAC3B. MAC3B was recently shown to exhibit E3 ligase activity in vitro. Through reverse genetics analysis we show that MAC3A and MAC3B are functionally redundant and are required for basal and R protein-mediated resistance in Arabidopsis. Like mos4-1 and Atcdc5-1, mac3a mac3b suppresses snc1-mediated autoimmunity. MAC3 localizes to the nucleus and interacts with AtCDC5 in planta. Our results suggest that MAC3A and MAC3B are members of the MAC that function redundantly in the regulation of plant innate immunity.


Subject(s)
Arabidopsis Proteins/immunology , Arabidopsis/immunology , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis Proteins/metabolism , Autoimmunity , Cell Cycle Proteins/metabolism , Cell Nucleus/metabolism , DNA Repair Enzymes/immunology , DNA Repair Enzymes/metabolism , Immunity, Innate/physiology , Mass Spectrometry , Nuclear Proteins/immunology , Nuclear Proteins/metabolism , Phenotype , Systems Biology/methods
20.
Proc Natl Acad Sci U S A ; 106(26): 10722-7, 2009 Jun 30.
Article in English | MEDLINE | ID: mdl-19506251

ABSTRACT

Protein phosphorylation plays essential roles in eukaryotic circadian clocks. Like PERIOD in animals, the Neurospora core circadian protein FRQ is progressively phosphorylated and becomes extensively phosphorylated before its degradation. In this study, by using purified FRQ protein from Neurospora, we identified 43 in vivo FRQ phosphorylation sites by mass spectrometry analysis. In addition, we show that CK-1a and CKII are responsible for most FRQ phosphorylation events and identify an additional 33 phosphorylation sites by in vitro kinase assays. Whole-cell metabolic isotope labeling and quantitative MS analyses suggest that circadian oscillation of the FRQ phosphorylation profile is primarily due to progressive phosphorylation at the majority of these newly discovered phosphorylation sites. Furthermore, systematic mutations of the identified FRQ phosphorylation sites led to either long or short period phenotypes. These changes in circadian period are attributed to increases or decreases in FRQ stability, respectively. Together, this comprehensive study of FRQ phosphorylation reveals that regulation of FRQ stability by multiple independent phosphorylation events is a major factor that determines the period length of the clock. A model is proposed to explain how FRQ stability is regulated by multiple phosphorylation events.


Subject(s)
Circadian Rhythm/physiology , Fungal Proteins/metabolism , Neurospora/metabolism , Amino Acid Sequence , Binding Sites/genetics , Blotting, Western , Circadian Rhythm/genetics , Cycloheximide/pharmacology , Fungal Proteins/genetics , Mass Spectrometry/methods , Models, Biological , Molecular Sequence Data , Mutation , Mycelium/drug effects , Mycelium/genetics , Mycelium/metabolism , Neurospora/drug effects , Neurospora/genetics , Phosphorylation , Protein Stability/drug effects , Protein Synthesis Inhibitors/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...