Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Mov Disord ; 39(6): 936-944, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38494847

RESUMEN

BACKGROUND: Immunotherapy targeting pathological α-synuclein (α-syn) species is a promising strategy for slowing disease progression in neurodegenerative synucleinopathies, including Parkinson's disease (PD). OBJECTIVE: The aim was to evaluate the safety, tolerability, pharmacokinetics, and target engagement of ascending doses of Lu AF82422. METHODS: In this first-in-human study (NCT03611569), healthy participants (18-55 years, cohort A) and patients with PD (40-80 years, Hoehn and Yahr stage ≤3, cohort B) were enrolled in ascending-dose cohorts and randomly assigned to receive single intravenous infusions of Lu AF82422 (cohorts A1-A6: 75, 225, 750, 2250 4500, and 9000 mg, respectively; cohorts B1 and B2: 2250 and 9000 mg, respectively) or placebo. Participants were monitored during a 12-week observational period. RESULTS: Overall, single intravenous infusions of Lu AF82422 were safe and well tolerated, and no serious adverse events (AE) were observed; the most common AEs were related to the study on lumbar punctures, headache, and common infections. Lu AF82422 concentrations (in plasma and cerebrospinal fluid [CSF]) increased in a dose-proportional manner with no observable differences between cohorts; mean plasma half-life was 700 h. Plasma concentrations of Lu AF82422 had an immediate, concentration-dependent lowering effect on the plasma concentration of free α-syn and on the ratio of free to total α-syn in all cohorts and lowered the free-to-total α-syn ratio in CSF in the high-dose PD cohort. CONCLUSIONS: The safety and pharmacokinetic profile of Lu AF82422 were appropriate for further clinical development, and results indicated peripheral target engagement. The central target engagement observed in participants with PD indicates that the doses of Lu AF82422 tested may provide CSF concentrations sufficient to target aggregated forms of α-syn. © 2024 H. Lundbeck A/S. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.


Asunto(s)
Enfermedad de Parkinson , alfa-Sinucleína , Humanos , Persona de Mediana Edad , Masculino , Femenino , Adulto , Enfermedad de Parkinson/tratamiento farmacológico , Anciano , Adolescente , Adulto Joven , Anciano de 80 o más Años , Relación Dosis-Respuesta a Droga , Método Doble Ciego
3.
Transl Psychiatry ; 7(11): 1261, 2017 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-29187755

RESUMEN

1q21.1 hemizygous microdeletion is a copy number variant leading to eightfold increased risk of schizophrenia. In order to investigate biological alterations induced by this microdeletion, we generated a novel mouse model (Df(h1q21)/+) and characterized it in a broad test battery focusing on schizophrenia-related assays. Df(h1q21)/+ mice displayed increased hyperactivity in response to amphetamine challenge and increased sensitivity to the disruptive effects of amphetamine and phencyclidine hydrochloride (PCP) on prepulse inhibition. Probing of the direct dopamine (DA) pathway using the DA D1 receptor agonist SKF-81297 revealed no differences in induced locomotor activity compared to wild-type mice, but Df(h1q21)/+ mice showed increased sensitivity to the DA D2 receptor agonist quinpirole and the D1/D2 agonist apomorphine. Electrophysiological characterization of DA neuron firing in the ventral tegmental area revealed more spontaneously active DA neurons and increased firing variability in Df(h1q21)/+ mice, and decreased feedback reduction of DA neuron firing in response to amphetamine. In a range of other assays, Df(h1q21)/+ mice showed no difference from wild-type mice: gross brain morphology and basic functions such as reflexes, ASR, thermal pain sensitivity, and motor performance were unaltered. Similarly, anxiety related measures, baseline prepulse inhibition, and seizure threshold were unaltered. In addition to the central nervous system-related phenotypes, Df(h1q21)/+ mice exhibited reduced head-to tail length, which is reminiscent of the short stature reported in humans with 1q21.1 deletion. With aspects of both construct and face validity, the Df(h1q21)/+ model may be used to gain insight into schizophrenia-relevant alterations in dopaminergic transmission.


Asunto(s)
Anomalías Múltiples , Conducta Animal , Deleción Cromosómica , Agonistas de Dopamina/farmacología , Inhibidores de Captación de Dopamina/farmacología , Neuronas Dopaminérgicas/metabolismo , Antagonistas de Aminoácidos Excitadores/farmacología , Megalencefalia , Núcleo Accumbens/metabolismo , Inhibición Prepulso , Receptores Dopaminérgicos/metabolismo , Esquizofrenia , Área Tegmental Ventral/metabolismo , Anomalías Múltiples/metabolismo , Anomalías Múltiples/patología , Anomalías Múltiples/fisiopatología , Anfetamina/farmacología , Animales , Apomorfina/farmacología , Conducta Animal/efectos de los fármacos , Benzazepinas/farmacología , Cromosomas Humanos Par 1/metabolismo , Modelos Animales de Enfermedad , Agonistas de Dopamina/administración & dosificación , Inhibidores de Captación de Dopamina/administración & dosificación , Neuronas Dopaminérgicas/efectos de los fármacos , Antagonistas de Aminoácidos Excitadores/administración & dosificación , Megalencefalia/metabolismo , Megalencefalia/patología , Megalencefalia/fisiopatología , Ratones , Ratones Endogámicos C57BL , Núcleo Accumbens/efectos de los fármacos , Fenciclidina/farmacología , Fenotipo , Inhibición Prepulso/efectos de los fármacos , Quinpirol/farmacología , Receptores Dopaminérgicos/efectos de los fármacos , Esquizofrenia/metabolismo , Esquizofrenia/patología , Esquizofrenia/fisiopatología , Área Tegmental Ventral/efectos de los fármacos
4.
J Psychiatry Neurosci ; 42(1): 48-58, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27391101

RESUMEN

BACKGROUND: The hemizygous 22q11.2 microdeletion is a common copy number variant in humans. The deletion confers high risk for neurodevelopmental disorders, including autism and schizophrenia. Up to 41% of deletion carriers experience psychotic symptoms. METHODS: We present a new mouse model (Df(h22q11)/+) of the deletion syndrome (22q11.2DS) and report on, to our knowledge, the most comprehensive study undertaken to date in 22q11.2DS models. The study was conducted in male mice. RESULTS: We found elevated postpubertal N-methyl-D-aspartate (NMDA) receptor antagonist-induced hyperlocomotion, age-independent prepulse inhibition (PPI) deficits and increased acoustic startle response (ASR). The PPI deficit and increased ASR were resistant to antipsychotic treatment. The PPI deficit was not a consequence of impaired hearing measured by auditory brain stem responses. The Df(h22q11)/+ mice also displayed increased amplitude of loudness-dependent auditory evoked potentials. Prefrontal cortex and dorsal striatal elevations of the dopamine metabolite DOPAC and increased dorsal striatal expression of the AMPA receptor subunit GluR1 was found. The Df(h22q11)/+ mice did not deviate from wild-type mice in a wide range of other behavioural and biochemical assays. LIMITATIONS: The 22q11.2 microdeletion has incomplete penetrance in humans, and the severity of disease depends on the complete genetic makeup in concert with environmental factors. In order to obtain more marked phenotypes reflecting the severe conditions related to 22q11.2DS it is suggested to expose the Df(h22q11)/+ mice to environmental stressors that may unmask latent psychopathology. CONCLUSION: The Df(h22q11)/+ model will be a valuable tool for increasing our understanding of the etiology of schizophrenia and other psychiatric disorders associated with the 22q11DS.


Asunto(s)
Envejecimiento/fisiología , Síndrome de DiGeorge/fisiopatología , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Filtrado Sensorial/fisiología , Ácido 3,4-Dihidroxifenilacético/metabolismo , Envejecimiento/efectos de los fármacos , Animales , Percepción Auditiva/fisiología , Cuerpo Estriado/metabolismo , Modelos Animales de Enfermedad , Potenciales Evocados Auditivos del Tronco Encefálico , Antagonistas de Aminoácidos Excitadores/farmacología , Masculino , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , Corteza Prefrontal/metabolismo , Receptores AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Reflejo de Sobresalto/fisiología
5.
Biophys Chem ; 220: 34-41, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27863716

RESUMEN

The 140-residue natively disordered protein α-synuclein (aSN) is a central component in the development of a family of neurodegenerative diseases termed synucleinopathies. This is attributed to its ability to form cytotoxic aggregates such as oligomers and amyloid fibrils. Consequently there have been intense efforts to avoid aggregation or reroute the aggregation pathway using pharmaceutical agents such as small molecules, chaperones and antibodies. aSN's lack of persistent structure in the monomeric state, as well as the multitude of different oligomeric and even different fibrillar states, makes it difficult to raise antibodies that would be efficacious in neutralizing all conformations of aSN. However, the C-terminal 20-40 residues of aSN are a promising epitope for antibody development. It is primarily disordered in both monomeric and aggregated forms, and an anti-C-terminal antibody will therefore be able to bind all forms. Furthermore, it might not interfere with the folding of aSN into membranes, which could be important for its physiological role. Here we report a screen of a series of monoclonal antibodies, which all target the C-terminal of aSN. According to dot blot analyses, different antibodies bound different forms of aSN with different preferences and showed reduced binding to monomeric compared to aggregated (oligomeric and fibrillary) aSN. Consequently they have different effects on aSN's ability to fibrillate and permeabilize membranes. Generally, the antibodies with strongest binding to aggregated aSN in dot blot, also inhibited fibrillation and membrane permeabilization the most, and promoted formation of amorphous aggregates surrounded by small and thin fibers. This suggests that the development of antibodies that targets the C-terminus, exposed in the aggregated forms of aSN, may be beneficial for improved immunotherapy against PD.


Asunto(s)
Amiloide/efectos de los fármacos , Anticuerpos Monoclonales/farmacología , Agregación Patológica de Proteínas/prevención & control , alfa-Sinucleína/inmunología , Animales , Permeabilidad de la Membrana Celular/efectos de los fármacos , Humanos , Ratones , Enfermedad de Parkinson/tratamiento farmacológico , Agregado de Proteínas/efectos de los fármacos , Agregación Patológica de Proteínas/tratamiento farmacológico
6.
Mov Disord ; 31(2): 203-13, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26704735

RESUMEN

Immunotherapy using antibodies targeting alpha-synuclein has proven to be an effective strategy for ameliorating pathological and behavioral deficits induced by excess pathogenic alpha-synuclein in various animal and/or cellular models. However, the process of selecting the anti-alpha-synuclein antibody with the best potential to treat synucleinopathies in humans is not trivial. Critical to this process is a better understanding of the pathological processes involved in the synucleinopathies and how antibodies are able to influence these. We will give an overview of the first proof-of-concept studies in rodent disease models and discuss challenges associated with developing antibodies against alpha-synuclein resulting from the distribution and structural characteristics of the protein. We will also provide a status on the passive immunization approaches targeting alpha-synuclein that have entered, or are expected to enter, clinical evaluation.


Asunto(s)
Modelos Animales de Enfermedad , Inmunización Pasiva/métodos , Enfermedad por Cuerpos de Lewy/terapia , Atrofia de Múltiples Sistemas/terapia , Enfermedad de Parkinson/terapia , alfa-Sinucleína/química , alfa-Sinucleína/inmunología , Animales
7.
Xenobiotica ; 45(2): 177-87, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25183402

RESUMEN

1. Regulation of hepatic metabolism or transport may lead to increase in drug clearance and compromise efficacy or safety. In this study, cryopreserved human hepatocytes were used to assess the effect of 309 compounds on the activity and mRNA expression (using qPCR techniques) of CYP1A2, CYP2B6 and CYP3A4, as well as mRNA expression of six hepatic transport proteins: OATP1B1 (SCLO1B1), OCT1 (SLC22A1), MDR1 (ABCB1), MRP2 (ABCC2), MRP3 (ABCC3) and BCRP (ABCG2). 2. The results showed that 6% of compounds induced CYP1A2 activity (1.5-fold increase); 30% induced CYP2B6 while 23% induced CYP3A4. qPCR data identified 16, 33 or 32% inducers of CYP1A2, CYP2B6 or CYP3A4, respectively. MRP2 was induced by 27 compounds followed by MDR1 (16)>BCRP (9)>OCT1 (8)>OATP1B1 (5)>MRP3 (2). 3. CYP3A4 appeared to be down-regulated (≥2-fold decrease in mRNA expression) by 53 compounds, 10 for CYP2B6, 6 for OCT1, 4 for BCRP, 2 for CYP1A2 and OATP1B1 and 1 for MDR1 and MRP2. 4. Structure-activity relationship analysis showed that CYP2B6 and CYP3A4 inducers are bulky lipophilic molecules with a higher number of heavy atoms and a lower number of hydrogen bond donors. Finally, a strategy for testing CYP inducers in drug discovery is proposed.


Asunto(s)
Inductores del Citocromo P-450 CYP1A2/farmacología , Inductores del Citocromo P-450 CYP2B6/farmacología , Inductores del Citocromo P-450 CYP3A/farmacología , Hepatocitos/efectos de los fármacos , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/química , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/química , Transportadoras de Casetes de Unión a ATP/metabolismo , Técnicas de Cultivo de Célula , Inductores del Citocromo P-450 CYP1A2/química , Inductores del Citocromo P-450 CYP2B6/química , Citocromo P-450 CYP3A/metabolismo , Inductores del Citocromo P-450 CYP3A/química , Descubrimiento de Drogas/métodos , Hepatocitos/enzimología , Humanos , Transportador 1 de Anión Orgánico Específico del Hígado , Proteína 2 Asociada a Resistencia a Múltiples Medicamentos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/química , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Transportadores de Anión Orgánico/química , Transportadores de Anión Orgánico/metabolismo , Transportador 1 de Catión Orgánico/química , Transportador 1 de Catión Orgánico/metabolismo , Relación Estructura-Actividad
8.
Biol Psychiatry ; 76(2): 128-37, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24090792

RESUMEN

BACKGROUND: Genome-wide scans have uncovered rare copy number variants conferring high risk of psychiatric disorders. The 15q13.3 microdeletion is associated with a considerably increased risk of idiopathic generalized epilepsy, intellectual disability, and schizophrenia. METHODS: A 15q13.3 microdeletion mouse model (Df[h15q13]/+) was generated by hemizygous deletion of the orthologous region and characterized with focus on schizophrenia- and epilepsy-relevant parameters. RESULTS: Df(h15q13)/+ mice showed marked changes in neuronal excitability in acute seizure assays, with increased propensity to develop myoclonic and absence-like seizures but decreased propensity for clonic and tonic seizures. Furthermore, they had impaired long-term spatial reference memory and a decreased theta frequency in hippocampus and prefrontal cortex. Electroencephalogram characterization revealed auditory processing deficits similar to those observed in schizophrenia. Gamma band power was increased during active state, but evoked gamma power following auditory stimulus (40 Hz) was dramatically reduced, mirroring observations in patients with schizophrenia. In addition, Df(h15q13)/+ mice showed schizophrenia-like decreases in amplitudes of auditory evoked potentials. Although displaying a grossly normal behavior, Df(h15q13)/+ mice are more aggressive following exposure to mild stressors, similar to what is described in human deletion carriers. Furthermore, Df(h15q13)/+ mice have increased body weight, and a similar increase in body weight was subsequently found in a sample of human subjects with 15q13.3 deletion. CONCLUSIONS: The Df(h15q13)/+ mouse shows similarities to several alterations related to the 15q13.3 microdeletion syndrome, epilepsy, and schizophrenia, offering a novel tool for addressing the underlying biology of these diseases.


Asunto(s)
Encéfalo/fisiopatología , Trastornos de los Cromosomas/genética , Modelos Animales de Enfermedad , Epilepsia/genética , Discapacidad Intelectual/genética , Ratones , Esquizofrenia/genética , Convulsiones/genética , Animales , Conducta Animal/fisiología , Peso Corporal/genética , Deleción Cromosómica , Cromosomas Humanos Par 15/genética , Electroencefalografía , Femenino , Humanos , Masculino , Ratones Endogámicos C57BL , Persona de Mediana Edad
9.
Neurosci Lett ; 554: 141-5, 2013 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-24021803

RESUMEN

Alpha-synuclein (α-syn) is mainly a presynaptic protein that has been implicated in Parkinson's disease and various other neurodegenerative disorders. Evidence obtained in knockout mice suggests that α-syn controls plasticity of dopamine (DA) overflow in presynaptic terminals. It is also believed that α-syn spreads and may seed its aggregates from cell to cell. The effects of exogenously applied α-syn on dopaminergic neurotransmission have not been studied. We addressed this issue by microinjecting human α-syn protein into the dorsal striatum of wild-type and α-syn knockout mice and monitoring stimulated DA overflow with constant potential amperometry. The evoked DA overflow was decreased in knockout mice six days after α-syn microinjection. The maximal velocity of DA re-uptake was reduced in both genotypes. Similar results were not seen when the effects of microinjected α-syn were studied immediately after the treatment, but instead there was a trend toward an increase in both stimulated DA overflow and maximal velocity of DA re-uptake. We conclude that locally applied human α-syn affects DA overflow and the effects depend on the presence of endogenous α-syn.


Asunto(s)
Cuerpo Estriado/metabolismo , Dopamina/metabolismo , alfa-Sinucleína/metabolismo , Animales , Cuerpo Estriado/efectos de los fármacos , Estimulación Eléctrica , Humanos , Masculino , Ratones Noqueados , Microinyecciones , Mutación , Proteínas Recombinantes/farmacología , alfa-Sinucleína/genética , alfa-Sinucleína/farmacología
10.
PLoS One ; 8(4): e60691, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23573275

RESUMEN

Impaired olfaction is an early pre-motor symptom of Parkinson's disease. The neuropathology underlying olfactory dysfunction in Parkinson's disease is unknown, however α-synuclein accumulation/aggregation and altered neurogenesis might play a role. We characterized olfactory deficits in a transgenic mouse model of Parkinson's disease expressing human wild-type α-synuclein under the control of the mouse α-synuclein promoter. Preliminary clinical observations suggest that rasagiline, a monoamine oxidase-B inhibitor, improves olfaction in Parkinson's disease. We therefore examined whether rasagiline ameliorates olfactory deficits in this Parkinson's disease model and investigated the role of olfactory bulb neurogenesis. α-Synuclein mice were progressively impaired in their ability to detect odors, to discriminate between odors, and exhibited alterations in short-term olfactory memory. Rasagiline treatment rescued odor detection and odor discrimination abilities. However, rasagiline did not affect short-term olfactory memory. Finally, olfactory changes were not coupled to alterations in olfactory bulb neurogenesis. We conclude that rasagiline reverses select olfactory deficits in a transgenic mouse model of Parkinson's disease. The findings correlate with preliminary clinical observations suggesting that rasagiline ameliorates olfactory deficits in Parkinson's disease.


Asunto(s)
Indanos/farmacología , Inhibidores de la Monoaminooxidasa/farmacología , Trastornos del Olfato/tratamiento farmacológico , Enfermedad de Parkinson/tratamiento farmacológico , alfa-Sinucleína/metabolismo , Factores de Edad , Animales , Modelos Animales de Enfermedad , Humanos , Indanos/uso terapéutico , Memoria a Corto Plazo , Ratones , Ratones Transgénicos , Inhibidores de la Monoaminooxidasa/uso terapéutico , Bulbo Olfatorio/efectos de los fármacos , Bulbo Olfatorio/patología , Olfato/efectos de los fármacos
11.
Psychopharmacology (Berl) ; 217(4): 525-38, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21537941

RESUMEN

RATIONALE: Psychosis susceptibility is mediated in part by the dopaminergic neurotransmitter system. In humans, individual differences in vulnerability for psychosis are reflected in differential sensitivity for psychostimulants such as amphetamine. We hypothesize that the same genes and pathways underlying behavioral sensitization in mice are also involved in the vulnerability to psychosis. OBJECTIVES: The aim of the current study was to investigate which genes and pathways may contribute to behavioral sensitization in different dopaminergic output areas in the mouse brain. METHODS: We took advantage of the naturally occurring difference in psychostimulant sensitivity in DBA/2 mice and selected animals displaying extremes in behavioral sensitization to amphetamine. Subsequently, the dopamine output areas, prefrontal cortex, nucleus accumbens, and cornu ammonis 1 (CA1) area of the hippocampus, were isolated by laser microdissection and subjected to DNA microarray analysis 1 h after a challenge dose of amphetamine. RESULTS: A large number of genes with differential expression between high and low responders were identified, with no overlap between brain regions. Validation of these gene expression changes with real-time quantitative polymerase chain reaction demonstrated that the most robust and reproducible effects on gene expression were in the CA1 region of the hippocampus. Interestingly, many of the validated genes in CA1 are members of the cAMP response element (CRE) family and targets of the glucocorticoid receptor (GR) and myocyte enhancer factor 2 (Mef2) transcription factors. CONCLUSION: We hypothesize that CRE, Mef2, and GR signaling form a transcription regulating network, which underlies differential amphetamine sensitivity, and therefore, may play an important role in susceptibility to psychosis.


Asunto(s)
Anfetamina/efectos adversos , Conducta Animal/efectos de los fármacos , Región CA1 Hipocampal/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Psicosis Inducidas por Sustancias/genética , Anfetamina/farmacocinética , Animales , Región CA1 Hipocampal/metabolismo , Interacción Gen-Ambiente , Genes Inmediatos-Precoces/efectos de los fármacos , Captura por Microdisección con Láser , Ratones , Ratones Endogámicos DBA , Actividad Motora/efectos de los fármacos , Análisis de Secuencia por Matrices de Oligonucleótidos , Psicosis Inducidas por Sustancias/metabolismo , Psicosis Inducidas por Sustancias/fisiopatología , Reacción en Cadena en Tiempo Real de la Polimerasa
12.
Psychiatr Genet ; 21(3): 125-31, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21317683

RESUMEN

OBJECTIVE: An interaction between predisposing genes and environmental stressors is thought to underlie the neurodevelopmental disorder schizophrenia. In a targeted gene screening, we previously found that the minor allele of the single nucleotide polymorphism (SNP) rs6336 in the neurotrophic tyrosine kinase receptor 1 (NTRK1/TRKA) gene is associated with schizophrenia as a risk factor. METHODS: We genotyped the TRKA SNP in a total of eight independent Caucasian schizophrenia case-control groups. RESULT: Remarkably, although in five of the groups a higher frequency of the risk allele was indeed found in the patients compared with the controls, in the three other groups the SNP acted as a protective factor. CONCLUSION: An intriguing possibility is that this dual character of the TRKA SNP is caused by its interaction with endophenotypic and/or epistatic factors.


Asunto(s)
Polimorfismo de Nucleótido Simple , Receptor trkA/genética , Esquizofrenia/genética , Adulto , Secuencia de Bases , Estudios de Casos y Controles , Cartilla de ADN , Europa (Continente) , Femenino , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa , Estados Unidos
13.
J Clin Invest ; 121(2): 715-25, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21245577

RESUMEN

Post-mortem analyses of brains from patients with Parkinson disease who received fetal mesencephalic transplants show that α-synuclein-containing (α-syn-containing) Lewy bodies gradually appear in grafted neurons. Here, we explored whether intercellular transfer of α-syn from host to graft, followed by seeding of α-syn aggregation in recipient neurons, can contribute to this phenomenon. We assessed α-syn cell-to-cell transfer using microscopy, flow cytometry, and high-content screening in several coculture model systems. Coculturing cells engineered to express either GFP- or DsRed-tagged α-syn resulted in a gradual increase in double-labeled cells. Importantly, α-syn-GFP derived from 1 neuroblastoma cell line localized to red fluorescent aggregates in other cells expressing DsRed-α-syn, suggesting a seeding effect of transmitted α-syn. Extracellular α-syn was taken up by cells through endocytosis and interacted with intracellular α-syn. Next, following intracortical injection of recombinant α-syn in rats, we found neuronal uptake was attenuated by coinjection of an endocytosis inhibitor. Finally, we demonstrated in vivo transfer of α-syn between host cells and grafted dopaminergic neurons in mice overexpressing human α-syn. In summary, intercellularly transferred α-syn interacts with cytoplasmic α-syn and can propagate α-syn pathology. These results suggest that α-syn propagation is a key element in the progression of Parkinson disease pathology.


Asunto(s)
Encéfalo/metabolismo , Trasplante de Células , Dopamina/metabolismo , Neuronas/metabolismo , alfa-Sinucleína/metabolismo , Animales , Células Cultivadas , Técnicas de Cocultivo , Medios de Cultivo Condicionados , Células HEK293 , Humanos , Cuerpos de Lewy/metabolismo , Ratones , Neuronas/citología , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Ratas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , alfa-Sinucleína/genética
14.
Psychiatr Genet ; 20(3): 93-101, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20410851

RESUMEN

OBJECTIVE: Linkage and association studies of bipolar affective disorder (BAD) point out chromosome 12q24 as a region of interest. METHODS: To investigate this region further, we conducted an association study of 22 DNA markers within a 1.14 Mb region in a Danish sample of 166 patients with BAD and 311 control individuals. Two-hundred and four Danish patients with schizophrenia were also included in the study. RESULTS: We observed highly significant allelic and genotypic association between BAD and two highly correlated markers. The risk allele of both markers considered separately conferred an odds ratio of 2 to an individual carrying one risk allele and an odds ratio of 4 for individuals carrying both risk alleles assuming an additive genetic model. These findings were supported by the haplotype analysis. In addition, we obtained a replication of four markers associated with BAD in an earlier UK study. The most significantly associated marker was also analyzed in a Scottish case-control sample and was earlier associated with BAD in the UK cohort. The association of that particular marker was strongly associated with BAD in a meta-analysis of the Danish, Scottish and UK sample (P=0.0003). The chromosome region confined by our most distant markers is gene-poor and harbours only a few predicted genes. This study implicates the Slynar locus. We confirmed one annotated Slynar transcript and identified a novel transcript in human brain cDNA. CONCLUSION: This study confirms 12q24.3 as a region of functional importance in the pathogenesis of BAD and highlights the importance of focused genotyping.


Asunto(s)
Trastorno Bipolar/genética , Cromosomas Humanos Par 12/genética , Sitios Genéticos/genética , Predisposición Genética a la Enfermedad , Alelos , Encéfalo/metabolismo , Encéfalo/patología , Dinamarca , Estudios de Asociación Genética , Marcadores Genéticos , Haplotipos/genética , Humanos , Desequilibrio de Ligamiento/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Esquizofrenia/genética
15.
BMC Neurosci ; 11: 9, 2010 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-20105316

RESUMEN

BACKGROUND: The amygdala-kindled rat is a model for human temporal lobe epilepsy and activity-dependent synaptic plasticity. Hippocampal RNA isolated from amygdala-kindled rats at different kindling stages was analyzed to identify kindling-induced genes. Furthermore, effects of the anti-epileptic drug levetiracetam on kindling-induced gene expression were examined. RESULTS: Cyclooxygenase-2 (Cox-2), Protocadherin-8 (Pcdh8) and TGF-beta-inducible early response gene-1 (TIEG1) were identified and verified as differentially expressed transcripts in the hippocampus of kindled rats by in situ hybridization and quantitative RT-PCR. In addition, we identified a panel of 16 additional transcripts which included Arc, Egr3/Pilot, Homer1a, Ania-3, MMP9, Narp, c-fos, NGF, BDNF, NT-3, Synaptopodin, Pim1 kinase, TNF-alpha, RGS2, Egr2/krox-20 and beta-A activin that were differentially expressed in the hippocampus of amygdala-kindled rats. The list consists of many synaptic plasticity-related immediate early genes (IEGs) as well as some late response genes encoding transcription factors, neurotrophic factors and proteins that are known to regulate synaptic remodelling. In the hippocampus, induction of IEG expression was dependent on the afterdischarge (AD) duration. Levetiracetam, 40 mg/kg, suppressed the development of kindling measured as severity of seizures and AD duration. In addition, single animal profiling also showed that levetiracetam attenuated the observed kindling-induced IEG expression; an effect that paralleled the anti-epileptic effect of the drug on AD duration. CONCLUSIONS: The present study provides mRNA expression data that suggest that levetiracetam attenuates expression of genes known to regulate synaptic remodelling. In the kindled rat, levetiracetam does so by shortening the AD duration thereby reducing the seizure-induced changes in mRNA expression in the hippocampus.


Asunto(s)
Amígdala del Cerebelo/fisiopatología , Anticonvulsivantes/farmacología , Expresión Génica/efectos de los fármacos , Genes Inmediatos-Precoces/efectos de los fármacos , Hipocampo/efectos de los fármacos , Piracetam/análogos & derivados , Amígdala del Cerebelo/efectos de los fármacos , Animales , Anticonvulsivantes/administración & dosificación , Anticonvulsivantes/sangre , Modelos Animales de Enfermedad , Estimulación Eléctrica , Epilepsia del Lóbulo Temporal/tratamiento farmacológico , Epilepsia del Lóbulo Temporal/genética , Epilepsia del Lóbulo Temporal/fisiopatología , Hipocampo/fisiopatología , Levetiracetam , Masculino , Plasticidad Neuronal/efectos de los fármacos , Plasticidad Neuronal/genética , Plasticidad Neuronal/fisiología , Piracetam/administración & dosificación , Piracetam/sangre , Piracetam/farmacología , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Convulsiones/tratamiento farmacológico , Convulsiones/genética , Convulsiones/fisiopatología , Sinapsis/efectos de los fármacos , Sinapsis/fisiología , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/genética , Transmisión Sináptica/fisiología , Factores de Tiempo
16.
J Psychiatr Res ; 43(15): 1195-9, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19435634

RESUMEN

Schizophrenia is a complex neurodevelopmental disorder that is thought to be induced by an interaction between predisposing genes and environmental stressors. To identify predisposing genetic factors, we performed a targeted (mostly neurodevelopmental) gene approach involving the screening of 396 selected non-synonymous single-nucleotide polymorphisms (SNPs) in three independent Caucasian schizophrenia case-control cohorts (USA, Denmark and Norway). A meta-analysis revealed ten non-synonymous SNPs that were nominally associated with schizophrenia, nine of which have not been previously linked to the disorder. Risk alleles are in TRKA (rs6336), BARD1 (rs28997576), LAMA5 (rs3810548), DKK2 (rs7037102), NOD2 (rs2066844) and RELN (rs2229860), whereas protective alleles are in NOD2 (rs2066845), NRG1 (rs10503929), ADAM7 (rs13259668) and TNR (rs859427). Following correction for multiple testing, the most attractive candidate for further study concerns SNP rs6336 (q=0.12) that causes the substitution of an evolutionarily highly conserved amino acid residue in the kinase domain of the neurodevelopmentally important receptor TRKA. Thus, TRKA signaling may represent a novel susceptibility pathway for schizophrenia.


Asunto(s)
Predisposición Genética a la Enfermedad , Polimorfismo de Nucleótido Simple/genética , Receptor trkA/genética , Esquizofrenia/genética , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Casos y Controles , Distribución de Chi-Cuadrado , Estudios de Cohortes , Dinamarca , Femenino , Frecuencia de los Genes , Estudio de Asociación del Genoma Completo , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Proteína Reelina , Estados Unidos , Población Blanca
17.
Hum Mol Genet ; 18(8): 1405-14, 2009 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-19218269

RESUMEN

The molecular mechanisms that lead to the cognitive defects characteristic of Down syndrome (DS), the most frequent cause of mental retardation, have remained elusive. Here we use a transgenic DS mouse model (152F7 line) to show that DYRK1A gene dosage imbalance deregulates chromosomal clusters of genes located near neuron-restrictive silencer factor (REST/NRSF) binding sites. We found that Dyrk1a binds the SWI/SNF complex known to interact with REST/NRSF. The mutation of a REST/NRSF binding site in the promoter of the REST/NRSF target gene L1cam modifies the transcriptional effect of Dyrk1a-dosage imbalance on L1cam. Dyrk1a dosage imbalance perturbs Rest/Nrsf levels with decreased Rest/Nrsf expression in embryonic neurons and increased expression in adult neurons. Using transcriptome analysis of embryonic brain subregions of transgenic 152F7 mouse line, we identified a coordinated deregulation of multiple genes that are responsible for dendritic growth impairment present in DS. Similarly, Dyrk1a overexpression in primary mouse cortical neurons induced severe reduction of the dendritic growth and dendritic complexity. We propose that DYRK1A overexpression-related neuronal gene deregulation via disturbance of REST/NRSF levels, and the REST/NRSF-SWI/SNF chromatin remodelling complex, significantly contributes to the neural phenotypic changes that characterize DS.


Asunto(s)
Ensamble y Desensamble de Cromatina , Proteínas Cromosómicas no Histona/metabolismo , Síndrome de Down/genética , Síndrome de Down/fisiopatología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Animales , Encéfalo/citología , Encéfalo/metabolismo , Células Cultivadas , Dendritas/fisiología , Ratones , Neuronas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Tirosina Quinasas/genética , Transfección , Quinasas DyrK
18.
Mol Cell Neurosci ; 39(4): 586-91, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18790059

RESUMEN

LRRK2, alpha-synuclein, UCH-L1 and DJ-1 are implicated in the etiology of Parkinson's disease. We show for the first time that increase in striatal alpha-synuclein levels induce increased Lrrk2 mRNA levels while Dj-1 and Uch-L1 are unchanged. We also demonstrate that a mouse strain lacking the dopamine signaling molecule DARPP-32 has significantly reduced levels of both Lrrk2 and alpha-synuclein, while mice carrying a disabling mutation of the DARPP-32 phosphorylation site T34A or lack alpha-synuclein do not show any changes. To test if striatal dopamine depletion influences Lrrk2 or alpha-synuclein expression, we used the neurotoxin 6-hydroxydopamine in rats and MitoPark mice in which there is progressive degeneration of dopamine neurons. Because striatal Lrrk2 and alpha-synuclein levels were not changed by dopamine depletion, we conclude that Lrrk2 and alpha-synuclein mRNA levels are possibly co-regulated, but they are not influenced by striatal dopamine levels.


Asunto(s)
Cuerpo Estriado/metabolismo , Enfermedad de Parkinson/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , alfa-Sinucleína/metabolismo , Animales , Cuerpo Estriado/anatomía & histología , Dopamina/metabolismo , Fosfoproteína 32 Regulada por Dopamina y AMPc/genética , Fosfoproteína 32 Regulada por Dopamina y AMPc/metabolismo , Femenino , Humanos , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Neuronas/citología , Neuronas/metabolismo , Oxidopamina/metabolismo , Enfermedad de Parkinson/etiología , Proteínas Serina-Treonina Quinasas/genética , Ratas , Ratas Sprague-Dawley , alfa-Sinucleína/genética
19.
Science ; 305(5681): 239-42, 2004 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-15247477

RESUMEN

Erythropoietin (EPO) is both hematopoietic and tissue protective, putatively through interaction with different receptors. We generated receptor subtype-selective ligands allowing the separation of EPO's bioactivities at the cellular level and in animals. Carbamylated EPO (CEPO) or certain EPO mutants did not bind to the classical EPO receptor (EPOR) and did not show any hematopoietic activity in human cell signaling assays or upon chronic dosing in different animal species. Nevertheless, CEPO and various nonhematopoietic mutants were cytoprotective in vitro and conferred neuroprotection against stroke, spinal cord compression, diabetic neuropathy, and experimental autoimmune encephalomyelitis at a potency and efficacy comparable to EPO.


Asunto(s)
Eritropoyetina/análogos & derivados , Eritropoyetina/uso terapéutico , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Animales , Apoptosis , Sitios de Unión , Células Cultivadas , Neuropatías Diabéticas/tratamiento farmacológico , Diseño de Fármacos , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Eritropoyesis , Eritropoyetina/química , Eritropoyetina/genética , Eritropoyetina/metabolismo , Eritropoyetina/farmacología , Femenino , Hematócrito , Humanos , Ligandos , Ratones , Ratones Endogámicos C3H , Mutagénesis , Neuronas/metabolismo , Fármacos Neuroprotectores/química , Fármacos Neuroprotectores/metabolismo , Fármacos Neuroprotectores/farmacología , Ratas , Ratas Sprague-Dawley , Receptores de Eritropoyetina/metabolismo , Proteínas Recombinantes , Transducción de Señal , Compresión de la Médula Espinal/tratamiento farmacológico , Accidente Cerebrovascular/tratamiento farmacológico , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...