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










Publication year range
3.
Sci Transl Med ; 10(465)2018 10 31.
Article in English | MEDLINE | ID: mdl-30381411

ABSTRACT

Spinocerebellar ataxia type 7 (SCA7) is an autosomal dominant neurodegenerative disorder characterized by cerebellar and retinal degeneration, and is caused by a CAG-polyglutamine repeat expansion in the ATAXIN-7 gene. Patients with SCA7 develop progressive cone-rod dystrophy, typically resulting in blindness. Antisense oligonucleotides (ASOs) are single-stranded chemically modified nucleic acids designed to mediate the destruction, prevent the translation, or modify the processing of targeted RNAs. Here, we evaluated ASOs as treatments for SCA7 retinal degeneration in representative mouse models of the disease after injection into the vitreous humor of the eye. Using Ataxin-7 aggregation, visual function, retinal histopathology, gene expression, and epigenetic dysregulation as outcome measures, we found that ASO-mediated Ataxin-7 knockdown yielded improvements in treated SCA7 mice. In SCA7 mice with retinal disease, intravitreal injection of Ataxin-7 ASOs also improved visual function despite initiating treatment after symptom onset. Using color fundus photography and autofluorescence imaging, we also determined the nature of retinal degeneration in human SCA7 patients. We observed variable disease severity and cataloged rapidly progressive retinal degeneration. Given the accessibility of neural retina, availability of objective, quantitative readouts for monitoring therapeutic response, and the rapid disease progression in SCA7, ASOs targeting ATAXIN-7 might represent a viable treatment for SCA7 retinal degeneration.


Subject(s)
Ataxin-7/metabolism , Mutant Proteins/metabolism , Oligonucleotides, Antisense/pharmacology , Spinocerebellar Ataxias/physiopathology , Vision, Ocular/drug effects , Animals , Ataxin-7/genetics , Chromatin Assembly and Disassembly/drug effects , Disease Models, Animal , Disease Progression , Epigenesis, Genetic/drug effects , Gene Expression Regulation/drug effects , Humans , Intravitreal Injections , Mice , Oligonucleotides, Antisense/administration & dosage , Peptides/metabolism , Phenotype , Photoreceptor Cells, Vertebrate/drug effects , Photoreceptor Cells, Vertebrate/metabolism , Protein Aggregates/drug effects , Retina/drug effects , Retina/metabolism , Retinal Degeneration/complications , Retinal Degeneration/pathology , Retinal Degeneration/physiopathology , Spinocerebellar Ataxias/complications , Spinocerebellar Ataxias/pathology
4.
J Clin Invest ; 128(1): 359-368, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29202483

ABSTRACT

Charcot-Marie-Tooth disease type 1A (CMT1A) is caused by duplication of peripheral myelin protein 22 (PMP22) and is the most common hereditary peripheral neuropathy. CMT1A is characterized by demyelination and axonal loss, which underlie slowed motor nerve conduction velocity (MNCV) and reduced compound muscle action potentials (CMAP) in patients. There is currently no known treatment for this disease. Here, we show that antisense oligonucleotides (ASOs) effectively suppress PMP22 mRNA in affected nerves in 2 murine CMT1A models. Notably, initiation of ASO treatment after disease onset restored myelination, MNCV, and CMAP almost to levels seen in WT animals. In addition to disease-associated gene expression networks that were restored with ASO treatment, we also identified potential disease biomarkers through transcriptomic profiling. Furthermore, we demonstrated that reduction of PMP22 mRNA in skin biopsies from ASO-treated rats is a suitable biomarker for evaluating target engagement in response to ASO therapy. These results support the use of ASOs as a potential treatment for CMT1A and elucidate potential disease and target engagement biomarkers for use in future clinical trials.


Subject(s)
Action Potentials/drug effects , Charcot-Marie-Tooth Disease/drug therapy , Motor Neurons/metabolism , Myelin Proteins/antagonists & inhibitors , Oligodeoxyribonucleotides, Antisense/pharmacology , Skin/metabolism , Action Potentials/genetics , Animals , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/metabolism , Charcot-Marie-Tooth Disease/pathology , Disease Models, Animal , Female , Male , Mice , Mice, Transgenic , Motor Neurons/pathology , Myelin Proteins/biosynthesis , Myelin Proteins/genetics , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Skin/pathology
5.
Ann Neurol ; 83(1): 27-39, 2018 01.
Article in English | MEDLINE | ID: mdl-29226998

ABSTRACT

OBJECTIVE: Alexander disease is a fatal leukodystrophy caused by autosomal dominant gain-of-function mutations in the gene for glial fibrillary acidic protein (GFAP), an intermediate filament protein primarily expressed in astrocytes of the central nervous system. A key feature of pathogenesis is overexpression and accumulation of GFAP, with formation of characteristic cytoplasmic aggregates known as Rosenthal fibers. Here we investigate whether suppressing GFAP with antisense oligonucleotides could provide a therapeutic strategy for treating Alexander disease. METHODS: In this study, we use GFAP mutant mouse models of Alexander disease to test the efficacy of antisense suppression and evaluate the effects on molecular and cellular phenotypes and non-cell-autonomous toxicity. Antisense oligonucleotides were designed to target the murine Gfap transcript, and screened using primary mouse cortical cultures. Lead oligonucleotides were then tested for their ability to reduce GFAP transcripts and protein, first in wild-type mice with normal levels of GFAP, and then in adult mutant mice with established pathology and elevated levels of GFAP. RESULTS: Nearly complete and long-lasting elimination of GFAP occurred in brain and spinal cord following single bolus intracerebroventricular injections, with a striking reversal of Rosenthal fibers and downstream markers of microglial and other stress-related responses. GFAP protein was also cleared from cerebrospinal fluid, demonstrating its potential utility as a biomarker in future clinical applications. Finally, treatment led to improved body condition and rescue of hippocampal neurogenesis. INTERPRETATION: These results demonstrate the efficacy of antisense suppression for an astrocyte target, and provide a compelling therapeutic approach for Alexander disease. Ann Neurol 2018;83:27-39.


Subject(s)
Alexander Disease/drug therapy , Glial Fibrillary Acidic Protein/antagonists & inhibitors , Oligonucleotides, Antisense/therapeutic use , Alexander Disease/genetics , Alexander Disease/pathology , Animals , Biomarkers/cerebrospinal fluid , Brain Chemistry/drug effects , Gene Expression Regulation/drug effects , Glial Fibrillary Acidic Protein/biosynthesis , Glial Fibrillary Acidic Protein/genetics , Hippocampus/drug effects , Hippocampus/growth & development , Hippocampus/pathology , Humans , Injections, Intraventricular , Mice , Mice, Inbred C57BL , Mutation/genetics , Neurogenesis/drug effects , Spinal Cord/drug effects , Spinal Cord/metabolism
6.
Mol Ther Nucleic Acids ; 8: 508-519, 2017 Sep 15.
Article in English | MEDLINE | ID: mdl-28918051

ABSTRACT

No treatments exist to slow or halt Parkinson's disease (PD) progression; however, inhibition of leucine-rich repeat kinase 2 (LRRK2) activity represents one of the most promising therapeutic strategies. Genetic ablation and pharmacological LRRK2 inhibition have demonstrated promise in blocking α-synuclein (α-syn) pathology. However, LRRK2 kinase inhibitors may reduce LRRK2 activity in several tissues and induce systemic phenotypes in the kidney and lung that are undesirable. Here, we test whether antisense oligonucleotides (ASOs) provide an alternative therapeutic strategy, as they can be restricted to the CNS and provide a stable, long-lasting reduction of protein throughout the brain. Administration of LRRK2 ASOs to the brain reduces LRRK2 protein levels and fibril-induced α-syn inclusions. Mice exposed to α-syn fibrils treated with LRRK2 ASOs show more tyrosine hydroxylase (TH)-positive neurons compared to control mice. Furthermore, intracerebral injection of LRRK2 ASOs avoids unwanted phenotypes associated with loss of LRRK2 expression in the periphery. This study further demonstrates that a reduction of endogenous levels of normal LRRK2 reduces the formation of α-syn inclusions. Importantly, this study points toward LRRK2 ASOs as a potential therapeutic strategy for preventing PD-associated pathology and phenotypes without causing potential adverse side effects in peripheral tissues associated with LRRK2 inhibition.

7.
J Hepatol ; 64(4): 781-9, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26658683

ABSTRACT

BACKGROUND & AIMS: Current treatment of chronic hepatitis B virus infection (CHB) includes interferon and nucleos(t)ide analogues, which generally do not reduce HBV surface antigen (HBsAg) production, a constellation that is associated with poor prognosis of CHB. Here we evaluated the efficacy of an antisense approach using antisense oligonucleotide (ASO) technology already in clinical use for liver targeted therapy to specifically inhibit HBsAg production and viremia in a preclinical setting. METHODS: A lead ASO was identified and characterized in vitro and subsequently tested for efficacy in vivo and in vitro using HBV transgenic and hydrodynamic transfection mouse and a cell culture HBV infection model, respectively. RESULTS: ASO treatment decreased serum HBsAg levels ⩾2 logs in a dose and time-dependent manner; HBsAg decreased 2 logs in a week and returned to baseline 4 weeks after a single ASO injection. ASO treatment effectively reduced HBsAg in combination with entecavir, while the nucleoside analogue alone did not. ASO treatment has pan-genotypic antiviral activity in the hydrodynamic transfection system. Finally, cccDNA-driven HBV gene expression is ASO sensitive in HBV infected cells in vitro. CONCLUSION: Our results demonstrate in a preclinical setting the efficacy of an antisense approach against HBV by efficiently reducing serum HBsAg (as well as viremia) across different genotypes alone or in combination with standard nucleoside therapy. Since the applied antisense technology is already in clinical use, a lead compound can be rapidly validated in a clinical setting and thus, constitutes a novel therapeutic approach targeting chronic HBV infection.


Subject(s)
Hepatitis B Surface Antigens/blood , Hepatitis B, Chronic/drug therapy , Oligonucleotides, Antisense/therapeutic use , Viremia/drug therapy , Animals , Hep G2 Cells , Hepatitis B e Antigens/blood , Hepatitis B, Chronic/virology , Humans , Mice
8.
J Pharmacol Exp Ther ; 355(2): 329-40, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26330536

ABSTRACT

Myotonic dystrophy type 1 (DM1) is the most common form of muscular dystrophy in adults. DM1 is caused by an expanded CTG repeat in the 3'-untranslated region of DMPK, the gene encoding dystrophia myotonica protein kinase (DMPK). Antisense oligonucleotides (ASOs) containing 2',4'-constrained ethyl-modified (cEt) residues exhibit a significantly increased RNA binding affinity and in vivo potency relative to those modified with other 2'-chemistries, which we speculated could translate to enhanced activity in extrahepatic tissues, such as muscle. Here, we describe the design and characterization of a cEt gapmer DMPK ASO (ISIS 486178), with potent activity in vitro and in vivo against mouse, monkey, and human DMPK. Systemic delivery of unformulated ISIS 486718 to wild-type mice decreased DMPK mRNA levels by up to 90% in liver and skeletal muscle. Similarly, treatment of either human DMPK transgenic mice or cynomolgus monkeys with ISIS 486178 led to up to 70% inhibition of DMPK in multiple skeletal muscles and ∼50% in cardiac muscle in both species. Importantly, inhibition of DMPK was well tolerated and was not associated with any skeletal muscle or cardiac toxicity. Also interesting was the demonstration that the inhibition of DMPK mRNA levels in muscle was maintained for up to 16 and 13 weeks post-treatment in mice and monkeys, respectively. These results demonstrate that cEt-modified ASOs show potent activity in skeletal muscle, and that this attractive therapeutic approach warrants further clinical investigation to inhibit the gain-of-function toxic RNA underlying the pathogenesis of DM1.


Subject(s)
Myotonic Dystrophy/drug therapy , Myotonin-Protein Kinase/metabolism , Oligonucleotides, Antisense/pharmacology , Oligonucleotides/pharmacology , Animals , Cell Line , Humans , Macaca fascicularis , Male , Mice , Mice, Transgenic , Muscle, Skeletal/enzymology , Myotonin-Protein Kinase/antagonists & inhibitors , Myotonin-Protein Kinase/genetics , Oligonucleotides/chemistry , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley
9.
Nature ; 498(7455): 511-5, 2013 Jun 27.
Article in English | MEDLINE | ID: mdl-23728303

ABSTRACT

Rev-Erb-α and Rev-Erb-ß are nuclear receptors that regulate the expression of genes involved in the control of circadian rhythm, metabolism and inflammatory responses. Rev-Erbs function as transcriptional repressors by recruiting nuclear receptor co-repressor (NCoR)-HDAC3 complexes to Rev-Erb response elements in enhancers and promoters of target genes, but the molecular basis for cell-specific programs of repression is not known. Here we present evidence that in mouse macrophages Rev-Erbs regulate target gene expression by inhibiting the functions of distal enhancers that are selected by macrophage-lineage-determining factors, thereby establishing a macrophage-specific program of repression. Remarkably, the repressive functions of Rev-Erbs are associated with their ability to inhibit the transcription of enhancer-derived RNAs (eRNAs). Furthermore, targeted degradation of eRNAs at two enhancers subject to negative regulation by Rev-Erbs resulted in reduced expression of nearby messenger RNAs, suggesting a direct role of these eRNAs in enhancer function. By precisely defining eRNA start sites using a modified form of global run-on sequencing that quantifies nascent 5' ends, we show that transfer of full enhancer activity to a target promoter requires both the sequences mediating transcription-factor binding and the specific sequences encoding the eRNA transcript. These studies provide evidence for a direct role of eRNAs in contributing to enhancer functions and suggest that Rev-Erbs act to suppress gene expression at a distance by repressing eRNA transcription.


Subject(s)
Down-Regulation/genetics , Enhancer Elements, Genetic/genetics , Macrophages/metabolism , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , Transcription, Genetic/genetics , Alleles , Animals , Base Sequence , Binding Sites , Gene Knockdown Techniques , Mice , Nuclear Receptor Subfamily 1, Group D, Member 1/deficiency , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics , Organ Specificity , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Response Elements/genetics
10.
Nat Neurosci ; 15(11): 1488-97, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23023293

ABSTRACT

FUS/TLS (fused in sarcoma/translocated in liposarcoma) and TDP-43 are integrally involved in amyotrophic lateral sclerosis (ALS) and frontotemporal dementia. We found that FUS/TLS binds to RNAs from >5,500 genes in mouse and human brain, primarily through a GUGGU-binding motif. We identified a sawtooth-like binding pattern, consistent with co-transcriptional deposition of FUS/TLS. Depletion of FUS/TLS from the adult nervous system altered the levels or splicing of >950 mRNAs, most of which are distinct from RNAs dependent on TDP-43. Abundance of only 45 RNAs was reduced after depletion of either TDP-43 or FUS/TLS from mouse brain, but among these were mRNAs that were transcribed from genes with exceptionally long introns and that encode proteins that are essential for neuronal integrity. Expression levels of a subset of these were lowered after TDP-43 or FUS/TLS depletion in stem cell-derived human neurons and in TDP-43 aggregate-containing motor neurons in sporadic ALS, supporting a common loss-of-function pathway as one component underlying motor neuron death from misregulation of TDP-43 or FUS/TLS.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , DNA-Binding Proteins/metabolism , Frontotemporal Dementia/metabolism , RNA Precursors/metabolism , RNA, Messenger/metabolism , RNA-Binding Protein FUS/metabolism , Adaptor Proteins, Signal Transducing , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Autophagy-Related Proteins , Brain/metabolism , Brain/pathology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Transformed , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Excitatory Amino Acid Transporter 2/genetics , Excitatory Amino Acid Transporter 2/metabolism , Female , Frontotemporal Dementia/genetics , Frontotemporal Dementia/pathology , Gene Expression Profiling , Gene Expression Regulation/genetics , Histone-Lysine N-Methyltransferase/metabolism , Humans , Immunoprecipitation , Kv Channel-Interacting Proteins/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Neurons/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Cell Adhesion Molecules/metabolism , Neural Stem Cells/metabolism , Neurofilament Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Protein Binding/genetics , Protein Structure, Tertiary/genetics , RNA Precursors/genetics , RNA Splicing/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA-Binding Protein FUS/deficiency , RNA-Binding Protein FUS/genetics , Shal Potassium Channels/metabolism , Spinal Cord/metabolism , Ubiquitin-Protein Ligases/metabolism , tau Proteins/genetics , tau Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...