Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
J Neurosci ; 36(3): 762-72, 2016 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-26791207

RESUMEN

The interneuronal propagation of aggregated tau is believed to play an important role in the pathogenesis of human tauopathies. It requires the uptake of seed-competent tau into cells, seeding of soluble tau in recipient neurons and release of seeded tau into the extracellular space to complete the cycle. At present, it is not known which tau species are seed-competent. Here, we have dissected the molecular characteristics of seed-competent tau species from the TgP301S tau mouse model using various biochemical techniques and assessed their seeding ability in cell and animal models. We found that sucrose gradient fractions from brain lysates seeded cellular tau aggregation only when large (>10 mer) aggregated, hyperphosphorylated (AT8- and AT100-positive) and nitrated tau was present. In contrast, there was no detectable seeding by fractions containing small, oligomeric (<6 mer) tau. Immunodepletion of the large aggregated AT8-positive tau strongly reduced seeding; moreover, fractions containing these species initiated the formation and spreading of filamentous tau pathology in vivo, whereas fractions containing tau monomers and small oligomeric assemblies did not. By electron microscopy, seed-competent sucrose gradient fractions contained aggregated tau species ranging from ring-like structures to small filaments. Together, these findings indicate that a range of filamentous tau aggregates are the major species that underlie the spreading of tau pathology in the P301S transgenic model. Significance statement: The spread of tau pathology from neuron to neuron is postulated to account for, or at least to contribute to, the overall propagation of tau pathology during the development of human tauopathies including Alzheimer's disease. It is therefore important to characterize the native tau species responsible for this process of seeding and pathology spreading. Here, we use several biochemical techniques to dissect the molecular characteristics of native tau protein conformers from TgP301S tau mice and show that seed-competent tau species comprise small fibrils capable of seeding tau pathology in cell and animal models. Characterization of seed-competent tau gives insight into disease mechanisms and therapeutic interventions.


Asunto(s)
Amiloide/genética , Encéfalo , Ovillos Neurofibrilares/genética , Tauopatías/genética , Proteínas tau/genética , Animales , Encéfalo/patología , Femenino , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Ovillos Neurofibrilares/patología , Tauopatías/patología
2.
Regul Toxicol Pharmacol ; 80: 348-57, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27155597

RESUMEN

Central Nervous System (CNS)-related safety concerns are major contributors to delays and failure during the development of new candidate drugs (CDs). CNS-related safety data on 141 small molecule CDs from five pharmaceutical companies were analyzed to identify the concordance between rodent multi-parameter neurofunctional assessments (Functional Observational Battery: FOB, or Irwin test: IT) and the five most common adverse events (AEs) in Phase I clinical trials, namely headache, nausea, dizziness, fatigue/somnolence and pain. In the context of this analysis, the FOB/IT did not predict the occurrence of these particular AEs in man. For AEs such as headache, nausea, dizziness and pain the results are perhaps unsurprising, as the FOB/IT were not originally designed to predict these AEs. More unexpected was that the FOB/IT are not adequate for predicting 'somnolence/fatigue' nonclinically. In drug development, these five most prevalent AEs are rarely responsible for delaying or stopping further progression of CDs. More serious AEs that might stop CD development occurred at too low an incidence rate in our clinical dataset to enable translational analysis.


Asunto(s)
Conducta Animal/efectos de los fármacos , Enfermedades del Sistema Nervioso Central/inducido químicamente , Sistema Nervioso Central/efectos de los fármacos , Ensayos Clínicos Fase I como Asunto , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/etiología , Pruebas de Toxicidad/métodos , Animales , Sistema Nervioso Central/fisiopatología , Enfermedades del Sistema Nervioso Central/fisiopatología , Relación Dosis-Respuesta a Droga , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/fisiopatología , Humanos , Ratones , Ratas , Reproducibilidad de los Resultados , Medición de Riesgo , Especificidad de la Especie
3.
J Neuroinflammation ; 8: 76, 2011 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-21729281

RESUMEN

INTRODUCTION: Microglial activation in multiple sclerosis has been postulated to contribute to long-term neurodegeneration during disease. Fingolimod has been shown to impact on the relapsing remitting phase of disease by modulating autoreactive T-cell egress from lymph organs. In addition, it is brain penetrant and has been shown to exert multiple effects on nervous system cells. METHODS: In this study, the impact of fingolimod and other sphingosine-1-phosphate receptor active molecules following lysophosphotidyl choline-induced demyelination was examined in the rat telencephalon reaggregate, spheroid cell culture system. The lack of immune system components allowed elucidation of the direct effects of fingolimod on CNS cell types in an organotypic situation. RESULTS: Following demyelination, fingolimod significantly augmented expression of myelin basic protein in the remyelination phase. This increase was not associated with changes in neurofilament levels, indicating de novo myelin protein expression not associated with axonal branching. Myelin wrapping was confirmed morphologically using confocal and electron microscopy. Increased remyelination was associated with down-regulation of microglial ferritin, tumor necrosis factor alpha and interleukin 1 during demyelination when fingolimod was present. In addition, nitric oxide metabolites and apoptotic effectors caspase 3 and caspase 7 were reduced during demyelination in the presence of fingolimod. The sphingosine-1-phosphate receptor 1 and 5 agonist BAF312 also increased myelin basic protein levels, whereas the sphingosine-1-phosphate receptor 1 agonist AUY954 failed to replicate this effect on remyelination. CONCLUSIONS: The results presented indicate that modulation of S1P receptors can ameliorate pathological effectors associated with microglial activation leading to a subsequent increase in protein and morphological markers of remyelination. In addition, sphingosine-1-phosphate receptor 5 is implicated in promoting remyelination in vitro. This knowledge may be of benefit for treatment of chronic microglial inflammation in multiple sclerosis.


Asunto(s)
Biomarcadores/metabolismo , Inmunosupresores/farmacología , Microglía/efectos de los fármacos , Microglía/fisiología , Vaina de Mielina/metabolismo , Glicoles de Propileno/farmacología , Esfingosina/análogos & derivados , Animales , Técnicas de Cultivo de Célula , Células Cultivadas , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/patología , Femenino , Clorhidrato de Fingolimod , Humanos , Inmunosupresores/uso terapéutico , Lisofosfatidilcolinas/farmacología , Microglía/citología , Esclerosis Múltiple Recurrente-Remitente/inmunología , Esclerosis Múltiple Recurrente-Remitente/patología , Proteína Básica de Mielina/metabolismo , Degeneración Nerviosa/tratamiento farmacológico , Degeneración Nerviosa/patología , Embarazo , Glicoles de Propileno/uso terapéutico , Ratas , Ratas Sprague-Dawley , Receptores de Lisoesfingolípidos/metabolismo , Esferoides Celulares/citología , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/fisiología , Esfingosina/farmacología , Esfingosina/uso terapéutico
4.
Mult Scler ; 17(8): 939-48, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21459808

RESUMEN

BACKGROUND: There has been poor translation for the use of immunosuppressive agents from experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS), into the treatment of MS. This may be due to the fact that most EAE studies examine prophylactic, pre-treatment regimes that prove to be therapeutically-ineffective in long-established, often progressive, MS. FTY720 (fingolimod/Gilenya) is a sphingosine-1-phosphate receptor modulator. This is a new oral agent that markedly reduces the number of relapses in people with MS, compared with currently licensed injectable agents such as the beta interferons. FTY720 has activity against lymphocytes but may also influence oligodendroglia and could therefore have the potential to influence progressive MS, by promoting remyelination. METHODS: The effect of FTY720 was assessed in relapsing-progressive EAE in mice. RESULTS: Early intervention during relapsing EAE could completely inhibit subsequent relapses, inhibited the accumulation of neurodegeneration, and facilitated motor recovery. However, when examined in secondary progressive EAE, that develops after the accumulation of deficit from relapsing disease, long-term treatment with FTY720 failed to slow deterioration when initiated late (4 months) into the disease course. CONCLUSIONS: This study indicates that early intervention with immunosuppressive agents may inhibit the generation of the neurodegenerative microenvironment, which is no longer responsive to potent immunosuppression. However, if treatment is initiated too late, progressive, neurological-disease continues unabated. This suggests that immunosuppression is insufficient to control secondary progression in animals, as has been found so far to be the case in MS, and may warrant early intervention with FTY720 for optimal treatment benefit.


Asunto(s)
Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Inmunosupresores/uso terapéutico , Degeneración Nerviosa/tratamiento farmacológico , Glicoles de Propileno/uso terapéutico , Esfingosina/análogos & derivados , Animales , Progresión de la Enfermedad , Encefalomielitis Autoinmune Experimental/patología , Femenino , Clorhidrato de Fingolimod , Masculino , Ratones , Recuperación de la Función/efectos de los fármacos , Esfingosina/uso terapéutico
5.
J Neuropathol Exp Neurol ; 68(6): 616-25, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19458548

RESUMEN

Multiple sclerosis is an immune-mediated demyelinating disease, with axonal loss underlying long-term progressive disability. In this study, we have analyzed axonal and myelin pathology in a chronic relapsing-remitting experimental autoimmune encephalomyelitis model in Biozzi ABH mice induced by immunization with a syngeneic spinal cord homogenate. The animals were followed for3 months; inflammation, T-cell infiltration, demyelination, and axonal loss were examined at various time points throughout the disease course. We found that macrophage infiltration and microglia activation preceded detectable T-cell infiltration. Axonal loss was first evident at the acute phase of disease before demyelination was detected. Demyelination and axonal loss occurred after each relapse and correlated with increasing residual motor deficits in remission. The resulting lesions displayed evidence of demyelination, remyelination, axonal degeneration, and axon loss. After a series of 3 relapses, animals entered a chronic progressive phase with permanent paralysis and a relative absence of inflammation. Axonal loss continued in this phase, although demyelinated axons persisted. These findings indicate that experimental autoimmune encephalomyelitis in Biozzi ABH mice has important similarities to multiple sclerosis with a relapsing-remitting disease course followed by a secondary progressive phase; it is thus a suitable model in which to explore remyelination and neuroprotective therapies for multiple sclerosis.


Asunto(s)
Axones/patología , Progresión de la Enfermedad , Encefalomielitis Autoinmune Experimental/patología , Vaina de Mielina/patología , Animales , Antígenos CD/metabolismo , Enfermedades Desmielinizantes/complicaciones , Enfermedades Desmielinizantes/patología , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/fisiopatología , Citometría de Flujo/métodos , Inflamación/etiología , Inflamación/patología , Linfocitos/patología , Ratones , Ratones Biozzi , Recurrencia , Médula Espinal/patología , Estadísticas no Paramétricas , Factores de Tiempo
6.
J Pharmacol Toxicol Methods ; 100: 106602, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31238094

RESUMEN

Regulatory guidelines recommend specialised safety pharmacology assessments in animals to characterise drug-induced effects on the central nervous system (CNS) prior to first-in-human trials, including the functional observational battery or Irwin test (here collectively termed neurofunctional assessments). These assessments effectively detect overtly neurotoxic drugs; however, the suitability of the in vivo assessments to readily detect more subtle drug effects on the nervous system has been questioned. A survey was formulated by an international expert working group convened by the (NC3Rs) to capture practice in CNS neurofunctional assessment tests and opinions on the perceived impact of in vivo test battery endpoints. Impact was defined as "the impact of measures alone/in combination on decision making in drug development or candidate selection when using the neurofunctional assessment". The results demonstrate that rodents are predominantly used for small molecule assessments, whereas non-rodents are frequently used to test biotherapeutics. Practice varied between respondents in terms of experimental design. Subsets of test battery endpoints were consistently considered highly impactful (e.g. convulsions, stereotypic behaviors); however, the perceived impact level of other endpoints varied depending whether drugs were designed for CNS targets. Many endpoints were considered to have no or minimal impact, whereas a subset of endpoints in CNS test batteries appears more impactful than others. A critical evaluation is required to assess whether the translational value of CNS in vivo safety pharmacology assessments could be increased by modifying or augmenting standard CNS test batteries. A revised approach to CNS safety assessment has the potential to reduce animal numbers without compromising patient safety.


Asunto(s)
Desarrollo de Medicamentos/métodos , Evaluación Preclínica de Medicamentos/métodos , Modelos Animales , Farmacología/métodos , Animales , Sistema Nervioso Central/efectos de los fármacos , Desarrollo de Medicamentos/legislación & jurisprudencia , Desarrollo de Medicamentos/estadística & datos numéricos , Evaluación Preclínica de Medicamentos/estadística & datos numéricos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/prevención & control , Humanos , Farmacología/legislación & jurisprudencia , Proyectos de Investigación/legislación & jurisprudencia , Proyectos de Investigación/estadística & datos numéricos , Encuestas y Cuestionarios
7.
J Neuroimmunol ; 193(1-2): 120-9, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18037503

RESUMEN

Cannabinoids may exhibit symptom control in multiple sclerosis (MS). We show here that cannabinoid receptor (CBR) agonists can also be immunosuppressive and neuroprotective in models of MS. Immunosuppression was associated with reduced: myelin-specific T cell responses; central nervous system infiltration and reduced clinical disease. This was found to be largely CB(1)R-dependent and only occurred at doses that induced significant cannabimimetic effects that would not be achieved clinically. Lower, non-immunosuppressive doses of cannabinoids however, slowed the accumulation of nerve loss and disability, despite failing to inhibit relapses. This further highlights the neuroprotective potential of cannabinoids to slow the progression of MS.


Asunto(s)
Cannabinoides/farmacología , Inmunosupresores/farmacología , Esclerosis Múltiple/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Animales , Benzoxazinas/farmacología , Cannabinoides/uso terapéutico , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/inmunología , Inmunosupresores/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Morfolinas/farmacología , Esclerosis Múltiple/inmunología , Naftalenos/farmacología , Fármacos Neuroprotectores/uso terapéutico , Piperidinas/farmacología , Pirazoles/farmacología , Receptor Cannabinoide CB1/fisiología , Receptor Cannabinoide CB2/fisiología , Rimonabant , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología
8.
Artículo en Inglés | MEDLINE | ID: mdl-29753134

RESUMEN

INTRODUCTION: The safety-related failure of drugs during clinical phases of development is a significant contributor to drug attrition, wasting resources and preventing treatments from reaching patients. A lack of concordance between results from animal models and adverse events in the clinic has been identified as one potential cause of attrition. In vitro models using human tissue or cells have the potential to replace some animal models and improve predictivity to humans. METHODS: To gauge the current use of human tissue models in safety pharmacology and the barriers to greater uptake, an electronic survey of the international safety assessment community was carried out and a Safety Pharmacology Society European Regional Meeting was organised entitled 'The Use of Human Tissue in Safety Assessment'. RESULTS: A greater range of human tissue models is in use in safety assessment now than four years ago, although data is still not routinely included in regulatory submissions. The barriers to increased uptake of the models have not changed over that time, with inadequate supply and characterisation of tissue being the most cited blocks. DISCUSSION: Supporting biobanking, the development of new human tissue modelling technology, and raising awareness in the scientific and regulatory communities are key ways in which the barriers to greater uptake of human tissue models can be overcome. The development of infrastructure and legislation in the UK to support the use of post-mortem or surgical discard tissue will allow scientists to locally source tissue for research.


Asunto(s)
Bancos de Muestras Biológicas/tendencias , Evaluación Preclínica de Medicamentos/métodos , Evaluación Preclínica de Medicamentos/tendencias , Animales , Bancos de Muestras Biológicas/normas , Evaluación Preclínica de Medicamentos/normas , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/prevención & control , Humanos , Modelos Animales , Obtención de Tejidos y Órganos/métodos , Obtención de Tejidos y Órganos/normas , Obtención de Tejidos y Órganos/tendencias
9.
Dis Model Mech ; 10(8): 939-942, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28768734

RESUMEN

Mouse models, including patient-derived xenograft mice, are widely used to address questions in cancer research. However, there are documented flaws in these models that can result in the misrepresentation of human tumour biology and limit the suitability of the model for translational research. A coordinated effort to promote the more widespread development and use of 'non-animal human tissue' models could provide a clinically relevant platform for many cancer studies, maximising the opportunities presented by human tissue resources such as biobanks. A number of key factors limit the wide adoption of non-animal human tissue models in cancer research, including deficiencies in the infrastructure and the technical tools required to collect, transport, store and maintain human tissue for lab use. Another obstacle is the long-standing cultural reliance on animal models, which can make researchers resistant to change, often because of concerns about historical data compatibility and losing ground in a competitive environment while new approaches are embedded in lab practice. There are a wide range of initiatives that aim to address these issues by facilitating data sharing and promoting collaborations between organisations and researchers who work with human tissue. The importance of coordinating biobanks and introducing quality standards is gaining momentum. There is an exciting opportunity to transform cancer drug discovery by optimising the use of human tissue and reducing the reliance on potentially less predictive animal models.


Asunto(s)
Investigación Biomédica , Modelos Biológicos , Neoplasias/patología , Animales , Humanos , Ratones , Técnicas de Cultivo de Tejidos , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Lab Anim ; 51(2): 160-169, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27307423

RESUMEN

Rodent models produce data which underpin biomedical research and non-clinical drug trials, but translation from rodents into successful clinical outcomes is often lacking. There is a growing body of evidence showing that improving experimental design is key to improving the predictive nature of rodent studies and reducing the number of animals used in research. Age, one important factor in experimental design, is often poorly reported and can be overlooked. The authors conducted a survey to assess the age used for a range of models, and the reasoning for age choice. From 297 respondents providing 611 responses, researchers reported using rodents most often in the 6-20 week age range regardless of the biology being studied. The age referred to as 'adult' by respondents varied between six and 20 weeks. Practical reasons for the choice of rodent age were frequently given, with increased cost associated with using older animals and maintenance of historical data comparability being two important limiting factors. These results highlight that choice of age is inconsistent across the research community and often not based on the development or cellular ageing of the system being studied. This could potentially result in decreased scientific validity and increased experimental variability. In some cases the use of older animals may be beneficial. Increased scientific rigour in the choice of the age of rodent may increase the translation of rodent models to humans.


Asunto(s)
Envejecimiento , Investigación Biomédica/métodos , Ratones/fisiología , Ratas/fisiología , Factores de Edad , Animales , Investigación Biomédica/normas , Terminología como Asunto
11.
Br J Pharmacol ; 174(16): 2662-2681, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28677901

RESUMEN

BACKGROUND AND PURPOSE: Our initial aim was to generate cannabinoid agents that control spasticity, occurring as a consequence of multiple sclerosis (MS), whilst avoiding the sedative side effects associated with cannabis. VSN16R was synthesized as an anandamide (endocannabinoid) analogue in an anti-metabolite approach to identify drugs that target spasticity. EXPERIMENTAL APPROACH: Following the initial chemistry, a variety of biochemical, pharmacological and electrophysiological approaches, using isolated cells, tissue-based assays and in vivo animal models, were used to demonstrate the activity, efficacy, pharmacokinetics and mechanism of action of VSN16R. Toxicological and safety studies were performed in animals and humans. KEY RESULTS: VSN16R had nanomolar activity in tissue-based, functional assays and dose-dependently inhibited spasticity in a mouse experimental encephalomyelitis model of MS. This effect occurred with over 1000-fold therapeutic window, without affecting normal muscle tone. Efficacy was achieved at plasma levels that are feasible and safe in humans. VSN16R did not bind to known CB1 /CB2 /GPPR55 cannabinoid-related receptors in receptor-based assays but acted on a vascular cannabinoid target. This was identified as the major neuronal form of the big conductance, calcium-activated potassium (BKCa ) channel. Drug-induced opening of neuronal BKCa channels induced membrane hyperpolarization, limiting excessive neural-excitability and controlling spasticity. CONCLUSIONS AND IMPLICATIONS: We identified the neuronal form of the BKCa channel as the target for VSN16R and demonstrated that its activation alleviates neuronal excitability and spasticity in an experimental model of MS, revealing a novel mechanism to control spasticity. VSN16R is a potential, safe and selective ligand for controlling neural hyper-excitability in spasticity.


Asunto(s)
Benzamidas/uso terapéutico , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Canales de Potasio de Gran Conductancia Activados por el Calcio/fisiología , Espasticidad Muscular/tratamiento farmacológico , Animales , Benzamidas/química , Benzamidas/farmacocinética , Benzamidas/farmacología , Perros , Método Doble Ciego , Endocannabinoides/química , Endocannabinoides/farmacocinética , Endocannabinoides/farmacología , Endocannabinoides/uso terapéutico , Femenino , Hepatocitos/metabolismo , Isomerismo , Macaca , Masculino , Arterias Mesentéricas/efectos de los fármacos , Arterias Mesentéricas/fisiología , Ratones , Ratones Noqueados , Conejos , Ratas Sprague-Dawley , Ratas Wistar , Receptor Cannabinoide CB1/genética , Receptores de Cannabinoides/genética , Conducto Deferente/efectos de los fármacos , Conducto Deferente/fisiología
12.
J Neurol Sci ; 233(1-2): 21-5, 2005 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-15894331

RESUMEN

The current failure of potent immunosuppressive agents to control progressive disease in multiple sclerosis has moved a focus from immunotherapy towards the need for neuroprotection. There is increasing evidence for cannabinoid-mediated control of symptoms, which is being more supported by the underlying biology. However there is accumulating evidence in vitro and in vivo to support the hypothesis that the cannabinoid system can limit the neurodegenerative possesses that drive progressive disease, and may provide a new avenue for disease control.


Asunto(s)
Cannabinoides/uso terapéutico , Enfermedades del Sistema Nervioso Central/prevención & control , Fármacos Neuroprotectores/uso terapéutico , Animales , Modelos Animales de Enfermedad , Humanos , Inflamación/etiología , Inflamación/prevención & control , Esclerosis Múltiple/tratamiento farmacológico
13.
PLoS One ; 8(10): e76907, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24130809

RESUMEN

Endocannabinoids and some phytocannabinoids bind to CB1 and CB2 cannabinoid receptors, transient receptor potential vanilloid one (TRPV1) receptor and the orphan G protein receptor fifty-five (GPR55). Studies using C57BL/10 and C57BL/6 (Cnr2 (tm1Zim)) CB2 cannabinoid receptor knockout mice have demonstrated an immune-augmenting effect in experimental autoimmune encephalomyelitis (EAE) models of multiple sclerosis. However, other EAE studies in Biozzi ABH mice often failed to show any treatment effect of either CB2 receptor agonism or antagonism on inhibition of T cell autoimmunity. The influence of genetic background on the induction of EAE in endocannabinoid system-related gene knockout mice was examined. It was found that C57BL/6.GPR55 knockout mice developed less severe disease, notably in female mice, following active induction with myelin oligodendrocyte glycoprotein 35-55 peptide. In contrast C57BL/6.CB2 (Cnr2 (Dgen)) receptor knockout mice developed augmented severity of disease consistent with the genetically and pharmacologically-distinct, Cnr2 (tm1Zim) mice. However, when the knockout gene was bred into the ABH mouse background and EAE induced with spinal cord autoantigens the immune-enhancing effect of CB2 receptor deletion was lost. Likewise CB1 receptor and transient receptor potential vanilloid one knockout mice on the ABH background demonstrated no alteration in immune-susceptibility, in terms of disease incidence and severity of EAE, in contrast to that reported in some C57BL/6 mouse studies. Furthermore the immune-modulating influence of GPR55 was marginal on the ABH mouse background. Whilst sedative doses of tetrahydrocannabinol could induce immunosuppression, this was associated with a CB1 receptor rather than a CB2 receptor-mediated effect. These data support the fact that non-psychoactive doses of medicinal cannabis have a marginal influence on the immune response in MS. Importantly, it adds a note of caution for the translational value of some transgenic/gene knockout and other studies on low-EAE susceptibility backgrounds with inconsistent disease course and susceptibility.


Asunto(s)
Encefalomielitis Autoinmune Experimental/genética , Técnicas de Inactivación de Genes , Esclerosis Múltiple/genética , Receptor Cannabinoide CB2/deficiencia , Receptor Cannabinoide CB2/genética , Receptores de Cannabinoides/deficiencia , Receptores de Cannabinoides/genética , Animales , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Eliminación de Gen , Inmunomodulación/genética , Masculino , Ratones , Esclerosis Múltiple/inmunología , Fenotipo , Especificidad de la Especie
14.
PLoS One ; 8(11): e79188, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24223903

RESUMEN

Multiple sclerosis is an immune-mediated, demyelinating and neurodegenerative disease that currently lacks any neuroprotective treatments. Innovative neuroprotective trial designs are required to hasten the translational process of drug development. An ideal target to monitor the efficacy of strategies aimed at treating multiple sclerosis is the visual system, which is the most accessible part of the human central nervous system. A novel C57BL/6 mouse line was generated that expressed transgenes for a myelin oligodendrocyte glycoprotein-specific T cell receptor and a retinal ganglion cell restricted-Thy1 promoter-controlled cyan fluorescent protein. This model develops spontaneous or induced optic neuritis, in the absence of paralytic disease normally associated with most rodent autoimmune models of multiple sclerosis. Demyelination and neurodegeneration could be monitored longitudinally in the living animal using electrophysiology, visual sensitivity, confocal scanning laser ophthalmoscopy and optical coherence tomography all of which are relevant to human trials. This model offers many advantages, from a 3Rs, economic and scientific perspective, over classical experimental autoimmune encephalomyelitis models that are associated with substantial suffering of animals. Optic neuritis in this model led to inflammatory damage of axons in the optic nerve and subsequent loss of retinal ganglion cells in the retina. This was inhibited by the systemic administration of a sodium channel blocker (oxcarbazepine) or intraocular treatment with siRNA targeting caspase-2. These novel approaches have relevance to the future treatment of neurodegeneration of MS, which has so far evaded treatment.


Asunto(s)
Carbamazepina/análogos & derivados , Modelos Animales de Enfermedad , Esclerosis Múltiple/terapia , Neuritis Óptica/terapia , Interferencia de ARN , Animales , Anticonvulsivantes/farmacología , Carbamazepina/farmacología , Caspasa 2/genética , Caspasa 2/inmunología , Caspasa 2/metabolismo , Potenciales Evocados Visuales/inmunología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Confocal , Esclerosis Múltiple/genética , Esclerosis Múltiple/inmunología , Glicoproteína Mielina-Oligodendrócito/inmunología , Oftalmoscopía , Nervio Óptico/inmunología , Nervio Óptico/metabolismo , Neuritis Óptica/genética , Neuritis Óptica/inmunología , Oxcarbazepina , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Retina/inmunología , Retina/metabolismo , Células Ganglionares de la Retina/inmunología , Células Ganglionares de la Retina/metabolismo , Tomografía de Coherencia Óptica
15.
Mult Scler Relat Disord ; 1(2): 64-75, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25876933

RESUMEN

Cannabis-based medicines have recently been approved for the treatment of pain and spasticity in multiple sclerosis (MS). This supports the original perceptions of people with MS, who were using illegal street cannabis for symptom control and pre-clinical testing in animal models of MS. This activity is supported both by the biology of the disease and the biology of the cannabis plant and the endocannabinoid system. MS results from disease that impairs neurotransmission and this is controlled by cannabinoid receptors and endogenous cannabinoid ligands. This can limit spasticity and may also influence the processes that drive the accumulation of progressive disability.

16.
J Biol Chem ; 282(20): 15208-16, 2007 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-17395590

RESUMEN

Experimental allergic encephalomyelitis, an autoimmune disorder mediated by T cells, results in demyelination, inflammation, and axonal loss in the central nervous system (CNS). Microglia play a critical role in major histocompatibility complex class II (MHC II)-dependent antigen presentation and in reactivation of CNS-infiltrated encephalitogenic T cells. Minocycline, a tetracycline anti-biotic, has profound anti-inflammatory properties and is experimentally used for treatment of many CNS disorders; however, the mechanisms involved in minocycline effects remain unknown. We show that administration of minocycline for 2 weeks ameliorated clinical severity of experimental allergic encephalomyelitis, an effect that partially involves the down-regulation of MHC II proteins in the spinal cord. Therefore, we sought to elucidate the molecular mechanisms of minocycline inhibitory effects on MHC II expression in microglia. Although complex, the co-activator class II transactivator (CIITA) is a key regulator of MHC II expression. Here we show that minocycline inhibited interferongamma (IFNgamma)-induced CIITA and MHC II mRNA. Interestingly, however, it was without effect on STAT1 phosphorylation or IRF-1 expression, transcription factors that are activated by IFNgamma and necessary for CIITA expression. Further experiments revealed that MHC II expression is down-regulated in the presence of the PKC(alpha) inhibitor Gö6976. Minocycline inhibited IFNgamma-induced PKC(alpha/betaII) phosphorylation and the nuclear translocation of both PKC(alpha/betaII) and IRF-1 that subsequently inhibits CIITA expression. Our present data delineate a molecular pathway of minocycline action that includes inhibitory effects on PKC(alpha/betaII) and transcription factors that regulate the expression of critical inflammatory genes such as MHC II. Such a fundamental mechanism may underlie the pleiotropic effects of minocycline in CNS inflammatory disorders.


Asunto(s)
Antibacterianos/farmacología , Enfermedades Autoinmunes Desmielinizantes SNC/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Antígenos de Histocompatibilidad Clase II/biosíntesis , Macrófagos/metabolismo , Microglía/metabolismo , Minociclina/farmacología , Proteína Quinasa C-alfa/metabolismo , Proteína Quinasa C/metabolismo , Animales , Antibacterianos/uso terapéutico , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Presentación de Antígeno/efectos de los fármacos , Presentación de Antígeno/inmunología , Carbazoles/farmacología , Enfermedades Autoinmunes Desmielinizantes SNC/tratamiento farmacológico , Enfermedades Autoinmunes Desmielinizantes SNC/inmunología , Enfermedades Autoinmunes Desmielinizantes SNC/patología , Modelos Animales de Enfermedad , Regulación hacia Abajo/inmunología , Inhibidores Enzimáticos/farmacología , Femenino , Antígenos de Histocompatibilidad Clase II/inmunología , Indoles/farmacología , Factor 1 Regulador del Interferón/inmunología , Factor 1 Regulador del Interferón/metabolismo , Interferón gamma/biosíntesis , Interferón gamma/inmunología , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Macrófagos/inmunología , Macrófagos/fisiología , Microglía/inmunología , Microglía/patología , Minociclina/uso terapéutico , Proteínas Nucleares/inmunología , Proteínas Nucleares/metabolismo , Fosforilación/efectos de los fármacos , Proteína Quinasa C/inmunología , Proteína Quinasa C beta , Proteína Quinasa C-alfa/inmunología , Ratas , Ratas Sprague-Dawley , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/patología , Factores de Tiempo , Transactivadores/inmunología , Transactivadores/metabolismo
17.
J Neurosci Res ; 74(6): 858-67, 2003 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-14648590

RESUMEN

The increase in myelin basic protein (MBP) synthesis observed in brain aggregate cultures supplemented with macrophages is reflected in elevated supernatant protein levels of the key promoters of oligodendrocyte proliferation, fibroblast growth factor-2 (FGF-2) and platelet-derived growth factor-AA (PDGF-AA), during the premyelinating phase. Although supernatant levels of transforming growth factor-beta1 (TGF-beta1), the most abundant growth factor produced at the transcriptional and translational levels by phagocytic macrophages, were reduced at this stage, it was the only growth factor for which mRNA expression was increased significantly in macrophage-enriched cultures. TGF-beta1, which supports oligodendrocyte differentiation, was increased in the supernatant of macrophage-enriched cultures only after the onset of myelinogenesis. Hence, standard cultures treated with TGF-beta1 during the premyelinating period reproduced effects of macrophage supplementation, inducing an increase in MBP synthesis and in PDGF-AA and FGF-2 bioavailability. A similar increase in MBP synthesis in PDGF-AA treated cultures emphasises its central role in oligodendrocyte progenitor proliferation. In contrast, FGF-2 blocked MBP synthesis in the cultures. In cultures treated with anti-TGF-beta1 antibody before or after the first detection of MBP, supernatant levels of TGF-beta1, FGF-2, and PDGF-AA were reduced with resultant inhibition of myelination. Paradoxically, supraphysiological TGF-beta1 treatment after the onset of myelination had the same effect on myelin accumulation. These results indicate an enabling and regulatory role for TGF-beta1 in oligodendrocyte development and, as a source of TGF-beta1, macrophages in the inflammatory multiple sclerosis lesion, may have the potential to promote remyelination by modulating the growth factor repertoire in demyelinating disease.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos/fisiología , Macrófagos/fisiología , Vaina de Mielina/fisiología , Factor de Crecimiento Derivado de Plaquetas/fisiología , Subunidades de Proteína/fisiología , Factor de Crecimiento Transformador beta/fisiología , Animales , Agregación Celular/fisiología , Células Cultivadas , Sistema Nervioso Central/citología , Sistema Nervioso Central/fisiología , Humanos , Ratas , Ratas Sprague-Dawley , Factor de Crecimiento Transformador beta1
18.
Eur J Neurosci ; 20(9): 2267-75, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15525268

RESUMEN

Multiple sclerosis is increasingly recognized as a neurodegenerative disease which is triggered by inflammation in the central nervous system (CNS). Demyelination-associated axonal or neuronal damage is a primary cause of disability and has thus far not been successfully targeted by available drug therapies. The neuroprotective properties of both endogenous and administered cannabinoids have been shown in in vivo and in vitro models of CNS damage following excitotoxic, oxidative, traumatic and ischaemic insults, with a predominantly apoptotic effector mechanism. In this study a foetal mouse telencephalon aggregate cell culture system was developed to compare tissue from cannabinoid receptor 1 knockout mice with wildtype counterparts. Aggregate formation and neurofilament/myelin basic protein accumulation were dependent on the age of foetal dissection and species used. Following treatment with interferon-gamma, levels of myelin basic protein, neurofilament, neuronal dephosphorylation and caspase 3 activation were assessed in telencephalon tissue in vitro. Cytokine treatment resulted in significant loss of the neuronal marker neurofilament-H in cannabinoid receptor 1 knockout cultures but not in wildtypes, indicating that presence of the cannabinoid receptor 1 gene can be neuroprotective. Caspase 3 activation was higher in cultures from knockout animals, indicating an apoptotic mechanism of cell death. Dephosphorylated neurofilament levels were significantly elevated in knockout mice, lending support to the premise that neurofilament dephosphorylation is a marker for neuronal damage. Taken together, these results indicate that neuroprotection could be elicited through the cannabinoid receptor 1, and point towards a potential therapeutic role for cannabinoid compounds in demyelinating conditions such as multiple sclerosis.


Asunto(s)
Cannabinoides/farmacología , Interferón gamma/farmacología , Degeneración Nerviosa/tratamiento farmacológico , Receptor Cannabinoide CB1/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/fisiopatología , Caspasa 3 , Caspasas/metabolismo , Agregación Celular/efectos de los fármacos , Agregación Celular/genética , Células Cultivadas , Interferón gamma/metabolismo , Ratones , Ratones Noqueados , Modelos Biológicos , Proteína Básica de Mielina/metabolismo , Degeneración Nerviosa/genética , Degeneración Nerviosa/prevención & control , Proteínas de Neurofilamentos/metabolismo , Fármacos Neuroprotectores/farmacología , Técnicas de Cultivo de Órganos/métodos , Fosforilación/efectos de los fármacos , Receptor Cannabinoide CB1/agonistas , Receptor Cannabinoide CB1/genética
19.
Glia ; 45(3): 278-86, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14730701

RESUMEN

Remyelination in CNS aggregate cultures is determined both by macrophage enrichment and the mode of demyelination. Despite the same degree of myelin loss, accumulation of MBP in anti-MOG antibody-demyelinated aggregates overtakes that of controls, while recovery is significantly delayed following IFN-gamma-induced demyelination. In antibody-treated cultures, remyelination was associated with a significant increase in culture supernatant levels of TGF-beta1, FGF-2, and PDGF-AA as well as an induction of TNF-alpha immediately following removal of the demyelinating insult. The impaired recovery in IFN-gamma-treated cultures, denoted by a significant reduction in TGF-beta1, was reversed by treatment with hrTGF-beta1. Macrophage supplementation of the cultures prior to the addition of either demyelinating agent induced a greater degree of myelin loss followed by incomplete remyelination in both cases. This failure to remyelinate was associated in both groups with a several-fold elevation in TNF-alpha and with modest increases in PDGF-AA and FGF-2 in the antibody-treated cultures. In contrast, macrophage supplementation to mature cultures in the absence of any demyelinating treatment resulted in enhanced accumulation of MBP associated with a promyelinative growth factor and TNF-alpha profile similar to that in aggregates enriched with macrophages at the outset of the culture period. Hence, effector elements of the adaptive immune response appear to override promyelinogenic in favor of proinflammatory macrophage factors in mature CNS aggregates, counteracting the potential for myelin repair.


Asunto(s)
Autoanticuerpos/farmacología , Sistema Nervioso Central/metabolismo , Citocinas/farmacología , Enfermedades Desmielinizantes/metabolismo , Macrófagos Peritoneales/metabolismo , Inhibición Neural , Animales , Agregación Celular/inmunología , Células Cultivadas , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/patología , Enfermedades Desmielinizantes/inmunología , Enfermedades Desmielinizantes/patología , Proteínas de la Mielina , Glicoproteína Asociada a Mielina/inmunología , Glicoproteína Mielina-Oligodendrócito , Inhibición Neural/inmunología , Ratas , Ratas Sprague-Dawley , Telencéfalo/metabolismo , Telencéfalo/patología
20.
Brain ; 126(Pt 10): 2191-202, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12876144

RESUMEN

Multiple sclerosis is increasingly being recognized as a neurodegenerative disease that is triggered by inflammatory attack of the CNS. As yet there is no satisfactory treatment. Using experimental allergic encephalo myelitis (EAE), an animal model of multiple sclerosis, we demonstrate that the cannabinoid system is neuroprotective during EAE. Mice deficient in the cannabinoid receptor CB1 tolerate inflammatory and excitotoxic insults poorly and develop substantial neurodegeneration following immune attack in EAE. In addition, exogenous CB1 agonists can provide significant neuroprotection from the consequences of inflammatory CNS disease in an experimental allergic uveitis model. Therefore, in addition to symptom management, cannabis may also slow the neurodegenerative processes that ultimately lead to chronic disability in multiple sclerosis and probably other diseases.


Asunto(s)
Ácido Aspártico/análogos & derivados , Cannabinoides/uso terapéutico , Esclerosis Múltiple/tratamiento farmacológico , Proteínas de Saccharomyces cerevisiae , Animales , Ácido Aspártico/análisis , Axones/química , Axones/patología , Benzoxazinas , Ciclohexanoles/uso terapéutico , Maleato de Dizocilpina/uso terapéutico , Encefalomielitis Autoinmune Experimental , Agonistas de Aminoácidos Excitadores/uso terapéutico , Eliminación de Gen , Humanos , Ratones , Ratones Transgénicos , Proteínas de Unión al GTP Monoméricas/genética , Morfolinas/uso terapéutico , Esclerosis Múltiple/patología , N-Metilaspartato/uso terapéutico , Naftalenos/uso terapéutico , Degeneración Nerviosa , Proteínas Nucleares/genética , Receptores de Cannabinoides , Receptores de Droga/agonistas , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Retina/efectos de los fármacos , Retina/patología , Médula Espinal/patología , Uveítis/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA