Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters











Publication year range
1.
J Clin Invest ; 134(4)2024 Jan 09.
Article in English | MEDLINE | ID: mdl-38357922

ABSTRACT

Chronic and elevated levels of the antiviral cytokine IFN-α in the brain are neurotoxic. This is best observed in patients with genetic cerebral interferonopathies such as Aicardi-Goutières syndrome. Cerebral interferonopathies typically manifest in early childhood and lead to debilitating disease and premature death. There is no cure for these diseases with existing treatments largely aimed at managing symptoms. Thus, an effective therapeutic strategy is urgently needed. Here, we investigated the effect of antisense oligonucleotides targeting the murine IFN-α receptor (Ifnar1 ASOs) in a transgenic mouse model of cerebral interferonopathy. Intracerebroventricular injection of Ifnar1 ASOs into transgenic mice with brain-targeted chronic IFN-α production resulted in a blunted cerebral interferon signature, reduced neuroinflammation, restoration of blood-brain barrier integrity, absence of tissue destruction, and lessened neuronal damage. Remarkably, Ifnar1 ASO treatment was also effective when given after the onset of neuropathological changes, as it reversed such disease-related features. We conclude that ASOs targeting the IFN-α receptor halt and reverse progression of IFN-α-mediated neuroinflammation and neurotoxicity, opening what we believe to be a new and promising approach for the treatment of patients with cerebral interferonopathies.


Subject(s)
Interferon Type I , Nervous System Diseases , Child, Preschool , Humans , Mice , Animals , Neuroinflammatory Diseases , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/pharmacology , Interferon-alpha/genetics , Mice, Transgenic
2.
Toxicol Pathol ; 51(6): 375-389, 2023 08.
Article in English | MEDLINE | ID: mdl-38179962

ABSTRACT

Direct delivery of therapeutics to the central nervous system (CNS) greatly expands opportunities to treat neurological diseases but is technically challenging. This opinion outlines principal technical aspects of direct CNS delivery via intracerebroventricular (ICV) or intrathecal (IT) injection to common nonclinical test species (rodents, dogs, and nonhuman primates) and describes procedure-related clinical and histopathological effects that confound interpretation of test article-related effects. Direct dosing is by ICV injection in mice due to their small body size, while other species are dosed IT in the lumbar cistern. The most frequent procedure-related functional effects are transient absence of lower spinal reflexes after IT injection or death soon after ICV dosing. Common procedure-related microscopic findings in all species include leukocyte infiltrates in CNS meninges or perivascular (Virchow-Robin) spaces; nerve fiber degeneration in the spinal cord white matter (especially dorsal and lateral tracts compressed by dosing needles or indwelling catheters), spinal nerve roots, and sciatic nerve; meningeal fibrosis at or near IT injection sites; hemorrhage; and gliosis. Findings typically are minimal to occasionally mild. Findings tend to be more severe and/or have a higher incidence in the spinal cord segments and spinal nerve roots at or close to the site of administration.


Subject(s)
Oligonucleotides , Rodentia , Dogs , Mice , Animals , Central Nervous System/pathology , Spinal Cord/pathology , Nerve Degeneration/pathology , Primates
3.
JCI Insight ; 7(6)2022 03 22.
Article in English | MEDLINE | ID: mdl-35133987

ABSTRACT

Prion protein (PrP) concentration controls the kinetics of prion replication and is a genetically and pharmacologically validated therapeutic target for prion disease. In order to evaluate PrP concentration as a pharmacodynamic biomarker and assess its contribution to known prion disease risk factors, we developed and validated a plate-based immunoassay reactive for PrP across 6 species of interest and applicable to brain and cerebrospinal fluid (CSF). PrP concentration varied dramatically across different brain regions in mice, cynomolgus macaques, and humans. PrP expression did not appear to contribute to the known risk factors of age, sex, or common PRNP genetic variants. CSF PrP was lowered in the presence of rare pathogenic PRNP variants, with heterozygous carriers of P102L displaying 55%, and D178N just 31%, of the CSF PrP concentration of mutation-negative controls. In rodents, pharmacologic reduction of brain Prnp RNA was reflected in brain parenchyma PrP and, in turn in CSF PrP, validating CSF as a sampling compartment for the effect of PrP-lowering therapy. Our findings support the use of CSF PrP as a pharmacodynamic biomarker for PrP-lowering drugs and suggest that relative reduction from individual baseline CSF PrP concentration may be an appropriate marker for target engagement.


Subject(s)
Prion Diseases , Prion Proteins , Prions , Animals , Biomarkers/cerebrospinal fluid , Genotype , Humans , Mice , Prion Diseases/diagnosis , Prion Diseases/drug therapy , Prion Proteins/cerebrospinal fluid , Prion Proteins/genetics , Prion Proteins/pharmacology , Prions/genetics , Prions/metabolism
4.
Sci Transl Med ; 13(620): eabg4711, 2021 11 17.
Article in English | MEDLINE | ID: mdl-34788075

ABSTRACT

Alexander disease (AxD) is a devastating leukodystrophy caused by gain-of-function mutations in GFAP, and the only available treatments are supportive. Recent advances in antisense oligonucleotide (ASO) therapy have demonstrated that transcript targeting can be a successful strategy for human neurodegenerative diseases amenable to this approach. We have previously used mouse models of AxD to show that Gfap-targeted ASO suppresses protein accumulation and reverses pathology; however, the mice have a mild phenotype with no apparent leukodystrophy or overt clinical features and are therefore limited for assessing functional outcomes. In this report, we introduce a rat model of AxD that exhibits hallmark pathology with GFAP aggregation in the form of Rosenthal fibers, widespread astrogliosis, and white matter deficits. These animals develop normally during the first postnatal weeks but fail to thrive after weaning and develop severe motor deficits as they mature, with about 14% dying of unknown cause between 6 and 12 weeks of age. In this model, a single treatment with Gfap-targeted ASO provides long-lasting suppression, reverses GFAP pathology, and, depending on age of treatment, prevents or mitigates white matter deficits and motor impairment. In this report, we characterize an improved animal model of AxD with myelin pathology and motor impairment, recapitulating prominent features of the human disease, and use this model to show that ASO therapy has the potential to not only prevent but also reverse many aspects of disease.


Subject(s)
Alexander Disease , Glial Fibrillary Acidic Protein , Motor Disorders , White Matter , Alexander Disease/genetics , Alexander Disease/metabolism , Alexander Disease/pathology , Animals , Astrocytes/metabolism , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Gliosis/pathology , Motor Disorders/metabolism , Motor Disorders/pathology , Mutation/genetics , Rats , White Matter/pathology
5.
Nucleic Acids Res ; 49(2): 657-673, 2021 01 25.
Article in English | MEDLINE | ID: mdl-33367834

ABSTRACT

Antisense oligonucleotides (ASOs) have emerged as a new class of drugs to treat a wide range of diseases, including neurological indications. Spinraza, an ASO that modulates splicing of SMN2 RNA, has shown profound disease modifying effects in Spinal Muscular Atrophy (SMA) patients, energizing efforts to develop ASOs for other neurological diseases. While SMA specifically affects spinal motor neurons, other neurological diseases affect different central nervous system (CNS) regions, neuronal and non-neuronal cells. Therefore, it is important to characterize ASO distribution and activity in all major CNS structures and cell types to have a better understanding of which neurological diseases are amenable to ASO therapy. Here we present for the first time the atlas of ASO distribution and activity in the CNS of mice, rats, and non-human primates (NHP), species commonly used in preclinical therapeutic development. Following central administration of an ASO to rodents, we observe widespread distribution and target RNA reduction throughout the CNS in neurons, oligodendrocytes, astrocytes and microglia. This is also the case in NHP, despite a larger CNS volume and more complex neuroarchitecture. Our results demonstrate that ASO drugs are well suited for treating a wide range of neurological diseases for which no effective treatments are available.


Subject(s)
Central Nervous System/chemistry , Mice/metabolism , Oligonucleotides, Antisense/pharmacokinetics , Primates/metabolism , Rats/metabolism , Animals , Central Nervous System/cytology , Female , In Situ Hybridization , Injections, Intraventricular , Injections, Spinal , Macaca fascicularis , Male , Neuroglia/chemistry , Neurons/chemistry , Oligonucleotides, Antisense/administration & dosage , Organ Specificity , RNA, Long Noncoding/analysis , RNA, Long Noncoding/antagonists & inhibitors , RNA, Long Noncoding/genetics , Rats, Sprague-Dawley , Ribonuclease H , Tissue Distribution
6.
J Transl Med ; 18(1): 309, 2020 08 08.
Article in English | MEDLINE | ID: mdl-32771027

ABSTRACT

BACKGROUND: The intrathecal (IT) dosing route introduces drugs directly into the CSF to bypass the blood-brain barrier and gain direct access to the CNS. We evaluated the use of convective forces acting on the cerebrospinal fluid as a means for increasing rostral delivery of IT dosed radioactive tracer molecules and antisense oligonucleotides (ASO) in the monkey CNS. We also measured the cerebral spinal fluid (CSF) volume in a group of cynomolgus monkeys. METHODS: There are three studies presented, in each of which cynomolgus monkeys were injected into the IT space with radioactive tracer molecules and/or ASO by lumbar puncture in either a low or high volume. The first study used the radioactive tracer 64Cu-DOTA and PET imaging to evaluate the effect of the convective forces. The second study combined the injection of the radioactive tracer 99mTc-DTPA and ASO, then used SPECT imaging and ex vivo tissue analysis of the effects of convective forces to bridge between the tracer and the ASO distributions. The third experiment evaluated the effects of different injection volumes on the distribution of an ASO. In the course of performing these studies we also measured the CSF volume in the subject monkeys by Magnetic Resonance Imaging. RESULTS: It was consistently found that larger bolus dose volumes produced greater rostral distribution along the neuraxis. Thoracic percussive treatment also increased rostral distribution of low volume injections. There was little added benefit on distribution by combining the thoracic percussive treatment with the high-volume injection. The CSF volume of the monkeys was found to be 11.9 ± 1.6 cm3. CONCLUSIONS: These results indicate that increasing convective forces after IT injection increases distribution of molecules up the neuraxis. In particular, the use of high IT injection volumes will be useful to increase rostral CNS distribution of therapeutic ASOs for CNS diseases in the clinic.


Subject(s)
Central Nervous System , Oligonucleotides, Antisense , Animals , Blood-Brain Barrier , Injections, Spinal , Macaca fascicularis
7.
J Vis Exp ; (152)2019 10 29.
Article in English | MEDLINE | ID: mdl-31736489

ABSTRACT

The blood brain barrier (BBB) is an important defense against the entrance of potentially toxic or pathogenic agents from the blood into the central nervous system (CNS). However, its existence also dramatically lowers the accessibility of systemically administered therapeutic agents to the CNS. One method to overcome this, is to inject those agents directly into the cerebrospinal fluid (CSF), thus bypassing the BBB. This can be done via implantation of a catheter for either continuous infusion using an osmotic pump, or for single bolus delivery. In this article, we describe a surgical protocol for delivery of CNS-targeting antisense oligonucleotides (ASOs) via a catheter implanted directly into the cauda equina space of the adult rat spine. As representative results, we show the efficacy of a single bolus ASO intrathecal (IT) injection via this catheterization system in knocking down the target RNA in different regions of the rat CNS. The procedure is safe, effective and does not require expensive equipment or surgical tools. The technique described here can be adapted to deliver drugs in other modalities as well.


Subject(s)
Blood-Brain Barrier/metabolism , Catheterization/methods , Central Nervous System/metabolism , Drug Delivery Systems/methods , Injections, Spinal/methods , Oligonucleotides, Antisense/administration & dosage , Animals , Biological Transport , Female , Male , Rats , Rats, Sprague-Dawley
8.
JCI Insight ; 4(20)2019 10 17.
Article in English | MEDLINE | ID: mdl-31619586

ABSTRACT

Intrathecal (IT) delivery and pharmacology of antisense oligonucleotides (ASOs) for the CNS have been successfully developed to treat spinal muscular atrophy. However, ASO pharmacokinetic (PK) and pharmacodynamic (PD) properties remain poorly understood in the IT compartment. We applied multimodal imaging techniques to elucidate the IT PK and PD of unlabeled, radioactively labeled, or fluorescently labeled ASOs targeting ubiquitously expressed or neuron-specific RNAs. Following lumbar IT bolus injection in rats, all ASOs spread rostrally along the neuraxis, adhered to meninges, and were partially cleared to peripheral lymph nodes and kidneys. Rapid association with the pia and arterial walls preceded passage of ASOs across the glia limitans, along arterial intramural basement membranes, and along white-matter axonal bundles. Several neuronal and glial cell types accumulated ASOs over time, with evidence of probable glial accumulation preceding neuronal uptake. IT doses of anti-GluR1 and anti-Gabra1 ASOs markedly reduced the mRNA and protein levels of their respective neurotransmitter receptor protein targets by 2 weeks and anti-Gabra1 ASOs also reduced binding of the GABAA receptor PET ligand 18F-flumazenil in the brain over 4 weeks. Our multimodal imaging approaches elucidate multiple transport routes underlying the CNS distribution, clearance, and efficacy of IT-dosed ASOs.


Subject(s)
Brain/metabolism , GABA-A Receptor Antagonists/pharmacokinetics , Muscular Atrophy, Spinal/drug therapy , Oligonucleotides, Antisense/pharmacokinetics , Animals , Arteries/diagnostic imaging , Arteries/metabolism , Brain/blood supply , Brain/cytology , Brain/diagnostic imaging , Flumazenil/administration & dosage , Flumazenil/analogs & derivatives , GABA-A Receptor Antagonists/administration & dosage , Gene Knockdown Techniques , Humans , Injections, Spinal , Intravital Microscopy , Male , Molecular Targeted Therapy/methods , Neuroglia/metabolism , Neurons/metabolism , Oligonucleotides, Antisense/administration & dosage , Pia Mater/diagnostic imaging , Pia Mater/metabolism , RNA, Messenger/analysis , RNA, Messenger/genetics , Rats , Receptors, AMPA/analysis , Receptors, AMPA/antagonists & inhibitors , Receptors, AMPA/genetics , Receptors, GABA-A/analysis , Receptors, GABA-A/genetics , Single Photon Emission Computed Tomography Computed Tomography , Spatio-Temporal Analysis , Thionucleotides/administration & dosage , Thionucleotides/pharmacokinetics , Tissue Distribution
9.
JCI Insight ; 52019 07 30.
Article in English | MEDLINE | ID: mdl-31361599

ABSTRACT

Prion disease is a fatal, incurable neurodegenerative disease of humans and other mammals caused by conversion of cellular prion protein (PrP; PrPC) into a self-propagating neurotoxic conformer (prions; PrPSc). Strong genetic proofs of concept support lowering PrP expression as a therapeutic strategy. Antisense oligonucleotides (ASOs) can provide a practical route to lowering one target mRNA in the brain, but their development for prion disease has been hindered by three unresolved questions from prior work: uncertainty about mechanism of action, unclear potential for efficacy against established prion infection, and poor tolerability of drug delivery by osmotic pumps. Here we test antisense oligonucleotides (ASOs) delivered by bolus intracerebroventricular injection to intracerebrally prion-infected wild-type mice. Prophylactic treatments given every 2-3 months extended survival times 61-98%, and a single injection at 120 days post-infection, near the onset of clinical signs, extended survival 55% (87 days). In contrast, a non-targeting control ASO was ineffective. Thus, PrP lowering is the mechanism of action of ASOs effective against prion disease in vivo, and infrequent, or even single, bolus injections of ASOs can slow prion neuropathogenesis and markedly extend survival, even when initiated near clinical signs. These findings should empower development of PrP-lowering therapy for prion disease.


Subject(s)
Oligonucleotides, Antisense/pharmacology , Oligonucleotides, Antisense/therapeutic use , Prion Diseases/drug therapy , Animals , Brain/pathology , Disease Models, Animal , Drug Discovery , Female , Genetic Therapy , Mice , Mice, Inbred C57BL , Prion Diseases/pathology , Survival Rate
10.
BMC Genomics ; 20(1): 164, 2019 Feb 28.
Article in English | MEDLINE | ID: mdl-30819113

ABSTRACT

BACKGROUND: Microglia are multifunctional cells that are key players in brain development and homeostasis. Recent years have seen tremendous growth in our understanding of the role microglia play in neurodegeneration, CNS injury, and developmental disorders. Given that microglia show diverse functional phenotypes, there is a need for more precise tools to characterize microglial states. Here, we experimentally define gene modules as the foundation for describing microglial functional states. RESULTS: In an effort to develop a comprehensive classification scheme, we profiled transcriptomes of mouse microglia in a stimulus panel with 96 different conditions. Using the transcriptomic data, we generated fine-resolution gene modules that are robustly preserved across datasets. These modules served as the basis for a combinatorial code that we then used to characterize microglial activation under various inflammatory stimulus conditions. CONCLUSIONS: The microglial gene modules described here were robustly preserved, and could be applied to in vivo as well as in vitro conditions to dissociate the signaling pathways that distinguish acutely inflamed microglia from aged microglia. The microglial gene modules presented here are a novel resource for classifying and characterizing microglial states in health and disease.


Subject(s)
Cellular Senescence/genetics , Microglia/metabolism , Transcriptome , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cells, Cultured , Down-Regulation , Inflammation/genetics , Inflammation/metabolism , Interferon Type I/pharmacology , Interferon-gamma/pharmacology , Mice , Phenotype , Resveratrol/pharmacology , Signal Transduction , Toll-Like Receptor 2/metabolism , Transcription Factors/metabolism , Transcriptome/drug effects
11.
Exp Neurol ; 305: 66-75, 2018 07.
Article in English | MEDLINE | ID: mdl-29608917

ABSTRACT

The loss of local spinal glycine-ergic tone has been postulated as one of the mechanisms contributing to the development of spinal injury-induced spasticity. In our present study using a model of spinal transection-induced muscle spasticity, we characterize the effect of spinally-targeted GlyT2 downregulation once initiated at chronic stages after induction of spasticity in rats. In animals with identified hyper-reflexia, the anti-spasticity effect was studied after intrathecal treatment with: i) glycine, ii) GlyT2 inhibitor (ALX 1393), and iii) GlyT2 antisense oligonucleotide (GlyT2-ASO). Administration of glycine and GlyT2 inhibitor led to significant suppression of spasticity lasting for a minimum of 45-60 min. Treatment with GlyT2-ASO led to progressive suppression of muscle spasticity seen at 2-3 weeks after treatment. Over the subsequent 4-12 weeks, however, the gradual appearance of profound spinal hyper-reflexia was seen. This was presented as spontaneous or slight-tactile stimulus-evoked muscle oscillations in the hind limbs (but not in upper limbs) with individual hyper-reflexive episodes lasting between 3 and 5 min. Chronic hyper-reflexia induced by GlyT2-ASO treatment was effectively blocked by intrathecal glycine. Immunofluorescence staining and Q-PCR analysis of the lumbar spinal cord region showed a significant (>90%) decrease in GlyT2 mRNA and GlyT2 protein. These data demonstrate that spinal GlyT2 downregulation provides only a time-limited therapeutic benefit and that subsequent loss of glycine vesicular synthesis resulting from chronic GlyT2 downregulation near completely eliminates the tonic glycine-ergic activity and is functionally expressed as profound spinal hyper-reflexia. These characteristics also suggest that chronic spinal GlyT2 silencing may be associated with pro-nociceptive activity.


Subject(s)
Down-Regulation/physiology , Glycine Plasma Membrane Transport Proteins/metabolism , Muscle Spasticity/metabolism , Reflex, Abnormal/physiology , Spinal Cord Injuries/metabolism , Spinal Cord/metabolism , Animals , Female , Muscle Spasticity/physiopathology , Rats , Rats, Sprague-Dawley , Spinal Cord/physiopathology , Spinal Cord Injuries/physiopathology , Thoracic Vertebrae , Time Factors
12.
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
13.
Pain ; 159(1): 139-149, 2018 01.
Article in English | MEDLINE | ID: mdl-28976422

ABSTRACT

There is an urgent need for better treatments for chronic pain, which affects more than 1 billion people worldwide. Antisense oligonucleotides (ASOs) have proven successful in treating children with spinal muscular atrophy, a severe infantile neurological disorder, and several ASOs are currently being tested in clinical trials for various neurological disorders. Here, we characterize the pharmacodynamic activity of ASOs in spinal cord and dorsal root ganglia (DRG), key tissues for pain signaling. We demonstrate that activity of ASOs lasts up to 2 months after a single intrathecal bolus dose. Interestingly, comparison of subcutaneous, intracerebroventricular, and intrathecal administration shows that DRGs are targetable by systemic and central delivery of ASOs, while target reduction in the spinal cord is achieved only after direct central delivery. Upon detailed characterization of ASO activity in individual cell populations in DRG, we observe robust target suppression in all neuronal populations, thereby establishing that ASOs are effective in the cell populations involved in pain propagation. Furthermore, we confirm that ASOs are selective and do not modulate basal pain sensation. We also demonstrate that ASOs targeting the sodium channel Nav1.7 induce sustained analgesia up to 4 weeks. Taken together, our findings support the idea that ASOs possess the required pharmacodynamic properties, along with a long duration of action beneficial for treating pain.


Subject(s)
Ganglia, Spinal/drug effects , Nociception/physiology , Oligonucleotides, Antisense/therapeutic use , Pain/drug therapy , Spinal Cord/drug effects , Animals , Disease Models, Animal , Ganglia, Spinal/physiopathology , Male , Neurons/drug effects , Neurons/physiology , Pain/physiopathology , Rats , Rats, Sprague-Dawley , Spinal Cord/physiopathology
14.
J Neurosci Methods ; 280: 36-46, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28163066

ABSTRACT

BACKGROUND: The blood brain barrier (BBB) is an impediment to the development of large and highly charged molecules as therapeutics for diseases and injuries of the central nervous system (CNS). Antisense oligonucleotides (ASOs) are large (6000-8000MW) and highly charged and therefore do not cross the BBB. A method of circumventing the blood brain barrier to test ASOs, and other non-BBB penetrant molecules, as CNS therapeutics is the direct administration of these molecules to the CNS tissue or cerebral spinal fluid. NEW METHOD: We developed a rapid, simple and robust method for the intrathecal catheterization of rats to test putatively therapeutic antisense oligonucleotides. This method utilizes 23-gauge needles, simply constructed ½in. long 19-gauge guide cannulas and 8cm long plastic PE-10 sized catheters. COMPARISON WITH EXISTING METHODS: Unlike the cisterna magna approach, this method uses a lumbar approach for intrathecal catheterization with the catheter residing entirely in the cauda equina space minimizing spinal cord compression. Readily available materials and only a few specialized pieces of equipment, which are easily manufactured, are used for this intrathecal catheterization method. CONCLUSIONS: This method is easy to learn and has been taught to multiple in house surgeons, collaborators and contract laboratories. Greater than 90% catheterization success is routinely achieved with this method and as many as 100 catheters can be placed and test substance administered in one 6-h period. This method has allowed the pre-clinical testing of hundreds of ASOs as therapeutics for CNS indications.


Subject(s)
Catheterization/methods , Models, Animal , Animals , Catheterization/adverse effects , Catheterization/instrumentation , Catheters, Indwelling/adverse effects , Central Nervous System Agents/administration & dosage , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Coloring Agents , Enzyme-Linked Immunosorbent Assay , Female , Hyperalgesia/drug therapy , Immunohistochemistry , Injections, Spinal/instrumentation , Injections, Spinal/methods , Lumbar Vertebrae , Male , Oligonucleotides, Antisense/administration & dosage , RNA, Messenger/metabolism , Random Allocation , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Receptors, AMPA/metabolism , Spinal Cord/cytology , Spinal Cord/drug effects , Spinal Cord/metabolism
15.
Nucleic Acid Ther ; 23(6): 389-400, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24329414

ABSTRACT

The repressor element-1 silencing transcription factor/neuron-restrictive silencer factor (REST/NRSF) is one of the first negative-acting transcriptional regulators implicated in vertebrate development thought to regulate hundreds of neuron-specific genes. However, its function in the adult system remains elusive. Here we employ second-generation antisense oligonucleotides (ASOs) to study the impact of rest-mediated suppression on gene expression. We demonstrate specific reductions in REST levels in vitro, and in vivo in mouse liver following treatment with ASOs, and we show that ASO mediated-REST suppression results in the elevation in expression of many neuronal genes including brain-derived neurotrophic factor, Synapsin1 (syn1) and ß3-tubulin in BALB/c liver. Furthermore, we show the elevation of the affected proteins in plasma following ASO treatment. Finally, microarray analysis was applied to identify a broad range of genes modulated by REST suppression in mouse liver. Our findings suggest that REST may be an important target for neurodegenerative diseases like Huntington's disease, is also involved in the regulation of a broad range of additional cellular pathways, and that the antisense approach is a viable strategy for selectively modulating REST activity in vivo.


Subject(s)
Gene Expression Regulation , Repressor Proteins/genetics , Animals , Base Sequence , Binding Sites , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Gene Knockdown Techniques , Gene Regulatory Networks , Liver/metabolism , Male , Mice , Mice, Inbred BALB C , Oligonucleotide Array Sequence Analysis , Oligonucleotides, Antisense/genetics , Regulatory Sequences, Nucleic Acid , Repressor Proteins/metabolism , Transcriptome
16.
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
17.
Neuron ; 74(6): 1031-44, 2012 Jun 21.
Article in English | MEDLINE | ID: mdl-22726834

ABSTRACT

The primary cause of Huntington's disease (HD) is expression of huntingtin with a polyglutamine expansion. Despite an absence of consensus on the mechanism(s) of toxicity, diminishing the synthesis of mutant huntingtin will abate toxicity if delivered to the key affected cells. With antisense oligonucleotides (ASOs) that catalyze RNase H-mediated degradation of huntingtin mRNA, we demonstrate that transient infusion into the cerebrospinal fluid of symptomatic HD mouse models not only delays disease progression but mediates a sustained reversal of disease phenotype that persists longer than the huntingtin knockdown. Reduction of wild-type huntingtin, along with mutant huntingtin, produces the same sustained disease reversal. Similar ASO infusion into nonhuman primates is shown to effectively lower huntingtin in many brain regions targeted by HD pathology. Rather than requiring continuous treatment, our findings establish a therapeutic strategy for sustained HD disease reversal produced by transient ASO-mediated diminution of huntingtin synthesis.


Subject(s)
Huntington Disease/therapy , Nerve Tissue Proteins/antagonists & inhibitors , Nuclear Proteins/antagonists & inhibitors , Oligodeoxyribonucleotides, Antisense/therapeutic use , Animals , Corpus Striatum/metabolism , Corpus Striatum/pathology , Disease Models, Animal , Disease Progression , Huntingtin Protein , Huntington Disease/genetics , Huntington Disease/pathology , Infusions, Spinal , Macaca mulatta , Mice , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neurons/metabolism , Neurons/pathology , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Oligodeoxyribonucleotides, Antisense/administration & dosage , Time , Treatment Outcome
18.
PLoS One ; 7(2): e29429, 2012.
Article in English | MEDLINE | ID: mdl-22347364

ABSTRACT

BACKGROUND: Wig-1 is a transcription factor regulated by p53 that can interact with hnRNP A2/B1, RNA Helicase A, and dsRNAs, which plays an important role in RNA and protein stabilization. in vitro studies have shown that wig-1 binds p53 mRNA and stabilizes it by protecting it from deadenylation. Furthermore, p53 has been implicated as a causal factor in neurodegenerative diseases based in part on its selective regulatory function on gene expression, including genes which, in turn, also possess regulatory functions on gene expression. In this study we focused on the wig-1 transcription factor as a downstream p53 regulated gene and characterized the effects of wig-1 down regulation on gene expression in mouse liver and brain. METHODS AND RESULTS: Antisense oligonucleotides (ASOs) were identified that specifically target mouse wig-1 mRNA and produce a dose-dependent reduction in wig-1 mRNA levels in cell culture. These wig-1 ASOs produced marked reductions in wig-1 levels in liver following intraperitoneal administration and in brain tissue following ASO administration through a single striatal bolus injection in FVB and BACHD mice. Wig-1 suppression was well tolerated and resulted in the reduction of mutant Htt protein levels in BACHD mouse brain but had no effect on normal Htt protein levels nor p53 mRNA or protein levels. Expression microarray analysis was employed to determine the effects of wig-1 suppression on genome-wide expression in mouse liver and brain. Reduction of wig-1 caused both down regulation and up regulation of several genes, and a number of wig-1 regulated genes were identified that potentially links wig-1 various signaling pathways and diseases. CONCLUSION: Antisense oligonucleotides can effectively reduce wig-1 levels in mouse liver and brain, which results in specific changes in gene expression for pathways relevant to both the nervous system and cancer.


Subject(s)
Carrier Proteins/genetics , Gene Expression Regulation , Nuclear Proteins/genetics , Oligonucleotides, Antisense/pharmacology , Signal Transduction/genetics , Animals , Brain , Carrier Proteins/drug effects , Carrier Proteins/metabolism , Genomics , Liver , Mice , Neoplasms , Nervous System , Nuclear Proteins/drug effects , Nuclear Proteins/metabolism , RNA-Binding Proteins , Transcription Factors , Tumor Suppressor Protein p53
19.
Nat Neurosci ; 14(4): 459-68, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21358643

ABSTRACT

We used cross-linking and immunoprecipitation coupled with high-throughput sequencing to identify binding sites in 6,304 genes as the brain RNA targets for TDP-43, an RNA binding protein that, when mutated, causes amyotrophic lateral sclerosis. Massively parallel sequencing and splicing-sensitive junction arrays revealed that levels of 601 mRNAs were changed (including Fus (Tls), progranulin and other transcripts encoding neurodegenerative disease-associated proteins) and 965 altered splicing events were detected (including in sortilin, the receptor for progranulin) following depletion of TDP-43 from mouse adult brain with antisense oligonucleotides. RNAs whose levels were most depleted by reduction in TDP-43 were derived from genes with very long introns and that encode proteins involved in synaptic activity. Lastly, we found that TDP-43 autoregulates its synthesis, in part by directly binding and enhancing splicing of an intron in the 3' untranslated region of its own transcript, thereby triggering nonsense-mediated RNA degradation.


Subject(s)
Alternative Splicing/genetics , Amyotrophic Lateral Sclerosis/genetics , DNA-Binding Proteins/genetics , Nerve Degeneration/genetics , Neurons/pathology , RNA Precursors/genetics , RNA, Messenger/genetics , 3' Untranslated Regions/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/physiopathology , Animals , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/deficiency , Female , Homeostasis/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Degeneration/metabolism , Nerve Degeneration/physiopathology , Neurons/metabolism , Oligonucleotides, Antisense/genetics , RNA Precursors/antagonists & inhibitors , RNA, Messenger/antagonists & inhibitors
20.
Eur J Med Chem ; 44(11): 4413-25, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19577344

ABSTRACT

A series of small molecules consisting of a heterocyclic core flanked by two basic functionalities were synthesized and screened for in vitro affinity at the human histamine H(3) receptor (hH(3)R). Nine of the twenty-eight compounds tested were found to possess a hH(3)R K(i) of less than 5 nM and consisted of a diverse range of central hetero-aromatic linkers (pyridine, pyrazine, oxazole, isoxazole, thiazole, furan, thiophene, and pyrrole). One member of this series, (4-isopropyl-piperazin-1-yl)-(6-piperidin-1-ylmethyl-pyridin-3-yl)-methanone (37), was found to be a high affinity, selective antagonist that crosses the blood-brain barrier and occupies H(3) receptors after oral administration in the rat.


Subject(s)
Diamines/chemistry , Diamines/pharmacology , Histamine H3 Antagonists/chemistry , Histamine H3 Antagonists/pharmacology , Receptors, Histamine H3/metabolism , Animals , Blood-Brain Barrier/drug effects , Cell Line , Diamines/pharmacokinetics , Histamine H3 Antagonists/pharmacokinetics , Humans , Protein Binding , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL