Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Exp Neurol ; 305: 66-75, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29608917

RESUMO

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.


Assuntos
Regulação para Baixo/fisiologia , Proteínas da Membrana Plasmática de Transporte de Glicina/metabolismo , Espasticidade Muscular/metabolismo , Reflexo Anormal/fisiologia , Traumatismos da Medula Espinal/metabolismo , Medula Espinal/metabolismo , Animais , Feminino , Espasticidade Muscular/fisiopatologia , Ratos , Ratos Sprague-Dawley , Medula Espinal/fisiopatologia , Traumatismos da Medula Espinal/fisiopatologia , Vértebras Torácicas , Fatores de Tempo
2.
J Neurosci Methods ; 280: 36-46, 2017 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-28163066

RESUMO

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.


Assuntos
Cateterismo/métodos , Modelos Animais , Animais , Cateterismo/efeitos adversos , Cateterismo/instrumentação , Cateteres de Demora/efeitos adversos , Fármacos do Sistema Nervoso Central/administração & dosagem , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Corantes , Ensaio de Imunoadsorção Enzimática , Feminino , Hiperalgesia/tratamento farmacológico , Imuno-Histoquímica , Injeções Espinhais/instrumentação , Injeções Espinhais/métodos , Vértebras Lombares , Masculino , Oligonucleotídeos Antissenso/administração & dosagem , RNA Mensageiro/metabolismo , Distribuição Aleatória , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Receptores de AMPA/metabolismo , Medula Espinal/citologia , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo
3.
JAMA Neurol ; 70(2): 201-7, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23147550

RESUMO

BACKGROUND: Therapies designed to decrease the level of SOD1 are currently in a clinical trial for patients with superoxide dismutase (SOD1)-linked familial amyotrophic lateral sclerosis (ALS). OBJECTIVE: To determine whether the SOD1 protein in cerebral spinal fluid (CSF) may be a pharmacodynamic marker for antisense oligonucleotide therapy and a disease marker for ALS. DESIGN: Antisense oligonucleotides targeting human SOD1 were administered to rats expressing SOD1G93A. The human SOD1 protein levels were measured in the rats' brain and CSF samples. In human CSF samples, the following proteins were measured: SOD1, tau, phosphorylated tau, VILIP-1, and YKL-40. PARTICIPANTS: Ninety-three participants with ALS, 88 healthy controls, and 89 controls with a neurological disease (55 with dementia of the Alzheimer type, 19 with multiple sclerosis, and 15 with peripheral neuropathy). RESULTS: Antisense oligonucleotide-treated SOD1G93A rats had decreased human SOD1 messenger RNA levels (mean [SD] decrease of 69% [4%]) and decreased protein levels (mean [SD] decrease of 48% [14%]) in the brain. The rats' CSF samples showed a similar decrease in hSOD1 levels (mean [SD] decrease of 42% [14%]). In human CSF samples, the SOD1 levels varied a mean (SD) 7.1% (5.7%) after additional measurements, separated by months, were performed. The CSF SOD1 levels were higher in the participants with ALS (mean [SE] level, 172 [8] ng/mL; P<.05) and the controls with a neurological disease (mean [SE] level, 172 [6] ng/mL; P<.05) than in the healthy controls (mean [SE] level, 134 [4] ng/mL). Elevated CSF SOD1 levels did not correlate with disease characteristics in participants with ALS or controls with dementia of the Alzheimer type, but they did correlate with tau, phosphorylated tau, VILIP-1 and YKL-40 levels in controls with dementia of the Alzheimer type. CONCLUSIONS: SOD1 in CSF may be an excellent pharmacodynamic marker for SOD1-lowering therapies because antisense oligonucleotide therapy lowers protein levels in the rat brain and rat CSF samples and because SOD1 levels in CSF samples from humans are stable over time.


Assuntos
Encéfalo/efeitos dos fármacos , Encéfalo/enzimologia , Oligonucleotídeos Antissenso/administração & dosagem , Superóxido Dismutase/líquido cefalorraquidiano , Idoso , Esclerose Lateral Amiotrófica/líquido cefalorraquidiano , Esclerose Lateral Amiotrófica/enzimologia , Esclerose Lateral Amiotrófica/terapia , Animais , Biomarcadores/líquido cefalorraquidiano , Sistemas de Liberação de Medicamentos/métodos , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Oligonucleotídeos Antissenso/uso terapêutico , Ratos , Superóxido Dismutase/antagonistas & inibidores , Superóxido Dismutase-1
4.
Mol Ther Nucleic Acids ; 1: e9, 2012 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-23344724

RESUMO

Mice deficient for the cellular prion protein (PrP(C)) do not develop prion disease; accordingly, gene-based strategies to diminish PrP(C) expression are of interest. We synthesized a series of chemically modified antisense oligonucleotides (ASOs) targeted against mouse Prnp messenger RNA (mRNA) and identified those that were most effective in decreasing PrP(C) expression. Those ASOs were also evaluated in scrapie-infected cultured cells (ScN2a) for their efficacy in diminishing the levels of the disease-causing prion protein (PrP(Sc)). When the optimal ASO was infused intracerebrally into FVB mice over a 14-day period beginning 1 day after infection with the Rocky Mountain Laboratory (RML) strain of mouse prions, a prolongation of the incubation period of almost 2 months was observed. Whether ASOs can be used to develop an effective therapy for patients dying of Creutzfeldt-Jakob disease remains to be established.

5.
Biochem Pharmacol ; 78(3): 284-91, 2009 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-19393225

RESUMO

The role of dose and plasma concentration on liver tissue uptake and resulting antisense pharmacology using a chemically modified antisense oligonucleotide (ASO) targeting PTEN was assessed in mice. A single bolus s.c. dose of 60 mg/kg in mice showed a time-dependent reduction in liver PTEN mRNA that was maximal at 48-72 h and returned to near control levels by 20 days after administration. These pharmacodynamics are in good agreement with liver concentrations of ASO and are consistent with slow elimination (t(1/2)=8 days) of the PTEN ASO from Balb/C mouse liver. As expected, highest ASO concentrations in liver resulted from the s.c. slow infusion at all doses tested. Unexpectedly, the liver EC(50) for the 24-h s.c. slow infusion was approximately twofold higher than the two bolus routes of administration. Based on plasma concentration analysis it appears that 1-2 microg/mL ASO plasma concentration is a threshold that, if exceeded, results in robust antisense effects and below which there is reduced or complete loss of antisense pharmacology in liver even though bulk uptake in the organ is improved. Co-administration of a nonsense ASO competed for liver uptake, but unexpectedly increased pharmacodynamic response for the active oligonucleotide (ISIS 116847) supporting inhibition of a nonproductive bulk uptake pathway while simultaneously improving productive uptake (pharmacodynamics). This competition effect was similar whether the nonsense oligonucleotide was co-administered with ASO or administered up to 24 h prior to active ASO injection.


Assuntos
Oligonucleotídeos Antissenso/farmacologia , PTEN Fosfo-Hidrolase/efeitos dos fármacos , Animais , Sequência de Bases , Primers do DNA , Meia-Vida , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Oligonucleotídeos Antissenso/administração & dosagem , Oligonucleotídeos Antissenso/sangue , Reação em Cadeia da Polimerase Via Transcriptase Reversa
6.
J Clin Invest ; 116(8): 2290-6, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16878173

RESUMO

Neurotoxicity from accumulation of misfolded/mutant proteins is thought to drive pathogenesis in neurodegenerative diseases. Since decreasing levels of proteins responsible for such accumulations is likely to ameliorate disease, a therapeutic strategy has been developed to downregulate almost any gene in the CNS. Modified antisense oligonucleotides, continuously infused intraventricularly, have been demonstrated to distribute widely throughout the CNS of rodents and primates, including the regions affected in the major neurodegenerative diseases. Using this route of administration, we found that antisense oligonucleotides to superoxide dismutase 1 (SOD1), one of the most abundant brain proteins, reduced both SOD1 protein and mRNA levels throughout the brain and spinal cord. Treatment initiated near onset significantly slowed disease progression in a model of amyotrophic lateral sclerosis (ALS) caused by a mutation in SOD1. This suggests that direct delivery of antisense oligonucleotides could be an effective, dosage-regulatable means of treating neurodegenerative diseases, including ALS, where appropriate target proteins are known.


Assuntos
Doenças Neurodegenerativas/tratamento farmacológico , Oligonucleotídeos Antissenso/uso terapêutico , Animais , Sequência de Bases , Fibroblastos/efeitos dos fármacos , Fibroblastos/enzimologia , Macaca mulatta , Doença dos Neurônios Motores/enzimologia , Oligonucleotídeos Antissenso/metabolismo , Oligonucleotídeos Antissenso/farmacologia , Dobramento de Proteína , RNA Mensageiro/genética , Ratos , Superóxido Dismutase/genética , Superóxido Dismutase-1 , Função Ventricular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA