Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Genes Dev ; 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39142832

ABSTRACT

Neurological and neuromuscular diseases resulting from familial, sporadic, or de novo mutations have devasting personal, familial, and societal impacts. As the initial product of DNA transcription, RNA transcripts and their associated ribonucleoprotein complexes provide attractive targets for modulation by increasing wild-type or blocking mutant allele expression, thus relieving downstream pathological consequences. Therefore, it is unsurprising that many existing and under-development therapeutics have focused on targeting disease-associated RNA transcripts as a frontline drug strategy for these genetic disorders. This review focuses on the current range of RNA targeting modalities using examples of both dominant and recessive neurological and neuromuscular diseases.

2.
NPJ Microgravity ; 10(1): 41, 2024 Mar 28.
Article in English | MEDLINE | ID: mdl-38548798

ABSTRACT

The cardiovascular system is strongly impacted by the hazards of spaceflight. Astronauts spending steadily increasing lengths of time in microgravity are subject to cardiovascular deconditioning resulting in loss of vascular tone, reduced total blood volume, and diminished cardiac output. Appreciating the mechanisms by which the cells of the vasculature are altered during spaceflight will be integral to understanding and combating these deleterious effects as the human presence in space advances. In this study, we performed RNA-Seq analysis coupled with review by QIAGEN Ingenuity Pathway Analysis software on human aortic smooth muscle cells (HASMCs) cultured for 3 days in microgravity and aboard the International Space Station to assess the transcriptomic changes that occur during spaceflight. The results of our RNA-Seq analysis show that SMCs undergo a wide range of transcriptional alteration while in space, significantly affecting 4422 genes. SMCs largely down-regulate markers of the contractile, synthetic, and osteogenic phenotypes including smooth muscle alpha actin (αSMA), matrix metalloproteinases (MMPs), and bone morphogenic proteins (BMPs). Additionally, components of several cellular signaling pathways were strongly impacted including the STAT3, NFκB, PI3K/AKT, HIF1α, and Endothelin pathways. This study highlights the significant changes in transcriptional behavior SMCs exhibit during spaceflight and puts these changes in context to better understand vascular function in space.

3.
Hum Mol Genet ; 29(24): 3900-3918, 2021 02 25.
Article in English | MEDLINE | ID: mdl-33378537

ABSTRACT

C9orf72 ALS/FTD patients show remarkable clinical heterogeneity, but the complex biology of the repeat expansion mutation has limited our understanding of the disease. BAC transgenic mice were used to better understand the molecular mechanisms and repeat length effects of C9orf72 ALS/FTD. Genetic analyses of these mice demonstrate that the BAC transgene and not integration site effects cause ALS/FTD phenotypes. Transcriptomic changes in cell proliferation, inflammation and neuronal pathways are found late in disease and alternative splicing changes provide early molecular markers that worsen with disease progression. Isogenic sublines of mice with 800, 500 or 50 G4C2 repeats generated from the single-copy C9-500 line show longer repeats result in earlier onset, increased disease penetrance and increased levels of RNA foci and dipeptide RAN protein aggregates. These data demonstrate G4C2 repeat length is an important driver of disease and identify alternative splicing changes as early biomarkers of C9orf72 ALS/FTD.


Subject(s)
Alternative Splicing , Amyotrophic Lateral Sclerosis/pathology , C9orf72 Protein/metabolism , DNA Repeat Expansion , Disease Models, Animal , Frontotemporal Dementia/pathology , Penetrance , Amyotrophic Lateral Sclerosis/etiology , Amyotrophic Lateral Sclerosis/metabolism , Animals , C9orf72 Protein/genetics , Frontotemporal Dementia/etiology , Frontotemporal Dementia/metabolism , Humans , Mice , Mice, Transgenic , Mutation , Phenotype
4.
Nat Commun ; 11(1): 2022, 2020 04 24.
Article in English | MEDLINE | ID: mdl-32332745

ABSTRACT

The thymus is a primary lymphoid organ that plays an essential role in T lymphocyte maturation and selection during development of one arm of the mammalian adaptive immune response. Although transcriptional mechanisms have been well documented in thymocyte development, co-/post-transcriptional modifications are also important but have received less attention. Here we demonstrate that the RNA alternative splicing factor MBNL1, which is sequestered in nuclear RNA foci by C(C)UG microsatellite expansions in myotonic dystrophy (DM), is essential for normal thymus development and function. Mbnl1 129S1 knockout mice develop postnatal thymic hyperplasia with thymocyte accumulation. Transcriptome analysis indicates numerous gene expression and RNA mis-splicing events, including transcription factors from the TCF/LEF family. CNBP, the gene containing an intronic CCTG microsatellite expansion in DM type 2 (DM2), is coordinately expressed with MBNL1 in the developing thymus and DM2 CCTG expansions induce similar transcriptome alterations in DM2 blood, which thus serve as disease-specific biomarkers.


Subject(s)
DNA-Binding Proteins/genetics , Myotonic Dystrophy/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Thymus Gland/growth & development , Adult , Aged , Aged, 80 and over , Animals , DNA Repeat Expansion , DNA-Binding Proteins/metabolism , Female , Humans , Introns/genetics , Male , Mice , Mice, Knockout , Microsatellite Repeats/genetics , Middle Aged , Myotonic Dystrophy/blood , Myotonic Dystrophy/immunology , RNA Splicing/immunology , RNA-Seq , Thymus Gland/immunology , Young Adult
5.
Proc Natl Acad Sci U S A ; 117(10): 5472-5477, 2020 03 10.
Article in English | MEDLINE | ID: mdl-32086392

ABSTRACT

Studies on myotonic dystrophy type 1 (DM1) have led to the RNA-mediated disease model for hereditary disorders caused by noncoding microsatellite expansions. This model proposes that DM1 disease manifestations are caused by a reversion to fetal RNA processing patterns in adult tissues due to the expression of toxic CUG RNA expansions (CUGexp) leading to decreased muscleblind-like, but increased CUGBP1/ETR3-like factor 1 (CELF1), alternative splicing activities. Here, we test this model in vivo, using the mouse HSALR poly(CUG) model for DM1 and recombinant adeno-associated virus (rAAV)-mediated transduction of specific splicing factors. Surprisingly, systemic overexpression of HNRNPA1, not previously linked to DM1, also shifted DM1-relevant splicing targets to fetal isoforms, resulting in more severe muscle weakness/myopathy as early as 4 to 6 wk posttransduction, whereas rAAV controls were unaffected. Overexpression of HNRNPA1 promotes fetal exon inclusion of representative DM1-relevant splicing targets in differentiated myoblasts, and HITS-CLIP of rAAV-mycHnrnpa1-injected muscle revealed direct interactions of HNRNPA1 with these targets in vivo. Similar to CELF1, HNRNPA1 protein levels decrease during postnatal development, but are elevated in both regenerating mouse muscle and DM1 skeletal muscle. Our studies suggest that CUGexp RNA triggers abnormal expression of multiple nuclear RNA binding proteins, including CELF1 and HNRNPA1, that antagonize MBNL activity to promote fetal splicing patterns.


Subject(s)
Alternative Splicing , Heterogeneous Nuclear Ribonucleoprotein A1/genetics , Heterogeneous Nuclear Ribonucleoprotein A1/metabolism , Myotonic Dystrophy/genetics , Animals , CELF1 Protein/genetics , DNA-Binding Proteins/metabolism , Disease Models, Animal , Fetus , Humans , Mice , Mice, Transgenic , Myotonic Dystrophy/metabolism , Myotonic Dystrophy/pathology , RNA-Binding Proteins/metabolism
6.
Genes Dev ; 31(11): 1122-1133, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28698297

ABSTRACT

Myotonic dystrophy type 1 (DM1) is a CTG microsatellite expansion (CTGexp) disorder caused by expression of CUGexp RNAs. These mutant RNAs alter the activities of RNA processing factors, including MBNL proteins, leading to re-expression of fetal isoforms in adult tissues and DM1 pathology. While this pathogenesis model accounts for adult-onset disease, the molecular basis of congenital DM (CDM) is unknown. Here, we test the hypothesis that disruption of developmentally regulated RNA alternative processing pathways contributes to CDM disease. We identify prominent alternative splicing and polyadenylation abnormalities in infant CDM muscle, and, although most are also misregulated in adult-onset DM1, dysregulation is significantly more severe in CDM. Furthermore, analysis of alternative splicing during human myogenesis reveals that CDM-relevant exons undergo prenatal RNA isoform transitions and are predicted to be disrupted by CUGexp-associated mechanisms in utero. To test this possibility and the contribution of MBNLs to CDM pathogenesis, we generated mouse Mbnl double (Mbnl1; Mbnl2) and triple (Mbnl1; Mbnl2; Mbnl3) muscle-specific knockout models that recapitulate the congenital myopathy, gene expression, and spliceopathy defects characteristic of CDM. This study demonstrates that RNA misprocessing is a major pathogenic factor in CDM and provides novel mouse models to further examine roles for cotranscriptional/post-transcriptional gene regulation during development.


Subject(s)
Muscle Development/genetics , Myotonic Dystrophy/genetics , Myotonic Dystrophy/physiopathology , RNA Processing, Post-Transcriptional/genetics , RNA Splicing , RNA-Binding Proteins/genetics , Animals , Carrier Proteins/genetics , Cells, Cultured , Child, Preschool , DNA-Binding Proteins/genetics , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation, Developmental/genetics , Gene Knockout Techniques , Humans , Infant , Mice , RNA-Binding Proteins/metabolism
7.
Nat Rev Genet ; 17(1): 19-32, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26593421

ABSTRACT

The human transcriptome is composed of a vast RNA population that undergoes further diversification by splicing. Detecting specific splice sites in this large sequence pool is the responsibility of the major and minor spliceosomes in collaboration with numerous splicing factors. This complexity makes splicing susceptible to sequence polymorphisms and deleterious mutations. Indeed, RNA mis-splicing underlies a growing number of human diseases with substantial societal consequences. Here, we provide an overview of RNA splicing mechanisms followed by a discussion of disease-associated errors, with an emphasis on recently described mutations that have provided new insights into splicing regulation. We also discuss emerging strategies for splicing-modulating therapy.


Subject(s)
Alternative Splicing , Animals , Base Sequence , Genetic Therapy , Humans , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/therapy , Mutation , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/therapy , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Spliceosomes/physiology
8.
Hum Gene Ther Clin Dev ; 25(4): 212-7, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25357151

ABSTRACT

Gene delivery vectors based on adeno-associated virus (AAV) have potential utility for treatment of many genetic disorders. Current AAV vector manufacturing methods employ helper viruses to deliver functions needed to produce replication-defective recombinant AAV (rAAV) vectors, and clearance of infectious helper virus from the drug substance is essential for ensuring the safety of rAAV-based therapies. We have developed a manufacturing method for the production of rAAV vectors using a recombinant herpes simplex virus type 1 (rHSV) complementation system in suspension baby hamster kidney cells. The manufacturing process includes three primary unit operations, detergent lysis of the cell harvest and two downstream column chromatography steps, which achieve viral clearance. These unit operations inactivate and remove HSV, including replication-competent HSV present at low levels in rHSV helper stocks. Here we report results quantifying the reduction in HSV achieved during rAAV vector purification. Clearance of HSV was at least 6.84 log10 with 1% Triton X-100, 4.34 log10 with CIM Q column chromatography, and 2.86 log10 with AVB affinity chromatography. Combined, these three orthogonal methods achieved clearance of at least 14.04 log10 of HSV. The total input quantity of rHSV in a 100-liter production batch is approximately 1.2×10(12) plaque-forming units (pfu), and after purification, the concentration of residual rHSV in the resulting drug substance of approximately 450 ml would be less than 2.42×10(-5) pfu/ml. A rAAV vector produced using this method was used in a clinical trial in which subjects receive up to 100 intramuscular injections of 1.35 ml each, which would contain a maximum of 3.27×10(-3) pfu of HSV. These results support the safety of rAAV vectors produced using our rHSV complementation method.


Subject(s)
DNA, Recombinant/isolation & purification , Dependovirus/genetics , Genetic Engineering/methods , Genetic Vectors/isolation & purification , Herpesvirus 1, Human/genetics , Animals , Cell Line , Chlorocebus aethiops , Cricetinae , DNA, Recombinant/chemistry , DNA, Recombinant/genetics , Genetic Vectors/chemistry , Genetic Vectors/genetics , Vero Cells
9.
Neuron ; 75(3): 437-50, 2012 Aug 09.
Article in English | MEDLINE | ID: mdl-22884328

ABSTRACT

The RNA-mediated disease model for myotonic dystrophy (DM) proposes that microsatellite C(C)TG expansions express toxic RNAs that disrupt splicing regulation by altering MBNL1 and CELF1 activities. While this model explains DM manifestations in muscle, less is known about the effects of C(C)UG expression on the brain. Here, we report that Mbnl2 knockout mice develop several DM-associated central nervous system (CNS) features including abnormal REM sleep propensity and deficits in spatial memory. Mbnl2 is prominently expressed in the hippocampus and Mbnl2 knockouts show a decrease in NMDA receptor (NMDAR) synaptic transmission and impaired hippocampal synaptic plasticity. While Mbnl2 loss did not significantly alter target transcript levels in the hippocampus, misregulated splicing of hundreds of exons was detected using splicing microarrays, RNA-seq, and HITS-CLIP. Importantly, the majority of the Mbnl2-regulated exons examined were similarly misregulated in DM. We propose that major pathological features of the DM brain result from disruption of the MBNL2-mediated developmental splicing program.


Subject(s)
Alternative Splicing , Brain/physiopathology , Myotonic Dystrophy/genetics , RNA-Binding Proteins/genetics , Animals , Base Sequence , Disease Models, Animal , Humans , Mice , Mice, Knockout , Molecular Sequence Data , Myotonic Dystrophy/metabolism , Myotonic Dystrophy/physiopathology , Neuronal Plasticity/genetics , Oligonucleotide Array Sequence Analysis , RNA-Binding Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Synaptic Transmission/genetics
10.
Hum Gene Ther ; 20(8): 861-70, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19419276

ABSTRACT

Recombinant adeno-associated virus (rAAV) production systems capable of meeting clinical or anticipated commercial-scale manufacturing needs have received relatively little scrutiny compared with the intense research activity afforded the in vivo and in vitro evaluation of rAAV for gene transfer. Previously we have reported a highly efficient recombinant herpes simplex virus type 1 (rHSV) complementation system for rAAV production in multiple adherent cell lines; however, production in a scalable format was not demonstrated. Here we report rAAV production by rHSV coinfection of baby hamster kidney (BHK) cells grown in suspension (sBHK cells), using two ICP27-deficient rHSV vectors, one harboring a transgene flanked by the AAV2 inverted terminal repeats and a second bearing the AAV rep2 and capX genes (where X is any rAAV serotype). The rHSV coinfection of sBHK cells produced similar rAAV1/AAT-specific yields (85,400 DNase-resistant particles [DRP]/cell) compared with coinfection of adherent HEK-293 cells (74,600 DRP/cell); however, sBHK cells permitted a 3-fold reduction in the rHSV-rep2/capX vector multiplicity of infection, grew faster than HEK-293 cells, retained specific yields (DRP/cell) at higher cell densities, and had a decreased virus production cycle. Furthermore, sBHK cells were able to produce AAV serotypes 1, 2, 5, and 8 at similar specific yields, using multiple therapeutic genes. rAAV1/AAT production in sBHK cells was scaled to 10-liter disposable bioreactors, using optimized spinner flask infection conditions, and resulted in average volumetric productivities as high as 2.4 x 10(14) DRP/liter.


Subject(s)
Dependovirus/growth & development , Dependovirus/genetics , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/physiology , Animals , Bioreactors , Buffers , Cell Count , Cell Line , Cell Proliferation , Cell Survival , Chromatography , Dependovirus/classification , Humans , Superinfection , Time Factors , Transgenes
SELECTION OF CITATIONS
SEARCH DETAIL