Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
CNS Neurosci Ther ; 26(2): 260-269, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31418518

RESUMEN

In spinal cord injured adult mammals, neutralizing the neurite growth inhibitor Nogo-A with antibodies promotes axonal regeneration and functional recovery, although axonal regeneration is limited in length. Neurotrophic factors such as BDNF stimulate neurite outgrowth and protect axotomized neurons. Can the effects obtained by neutralizing Nogo-A, inducing an environment favorable for axonal sprouting, be strengthened by adding BDNF? A unilateral incomplete hemicord lesion at C7 level interrupted the main corticospinal component in three groups of adult macaque monkeys: control monkeys (n = 6), anti-Nogo-A antibody-treated monkeys (n = 7), and anti-Nogo-A antibody and BDNF-treated monkeys (n = 5). The functional recovery of manual dexterity was significantly different between the 3 groups of monkeys, the lowest in the control group. Whereas the anti-Nogo-A antibody-treated animals returned to manual dexterity performances close to prelesion ones, irrespective of lesion size, both the control and the anti-Nogo-A/BDNF animals presented a limited functional recovery. In the control group, the limited spontaneous functional recovery depended on lesion size, a dependence absent in the combined treatment group (anti-Nogo-A antibody and BDNF). The functional recovery in the latter group was significantly lower than in anti-Nogo-A antibody-treated monkeys, although the lesion was larger in three out of the five monkeys in the combined treatment group.


Asunto(s)
Anticuerpos Bloqueadores/uso terapéutico , Factor Neurotrófico Derivado del Encéfalo/uso terapéutico , Trastornos del Movimiento/tratamiento farmacológico , Proteínas Nogo/antagonistas & inhibidores , Traumatismos de la Médula Espinal/tratamiento farmacológico , Animales , Axones , Médula Cervical/lesiones , Mano , Macaca fascicularis , Masculino , Destreza Motora , Trastornos del Movimiento/etiología , Regeneración Nerviosa , Desempeño Psicomotor/efectos de los fármacos , Recuperación de la Función , Traumatismos de la Médula Espinal/complicaciones
2.
Neurorehabil Neural Repair ; 32(6-7): 578-589, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29869587

RESUMEN

BACKGROUND: Neutralization of central nervous system neurite growth inhibitory factors, for example, Nogo-A, is a promising approach to improving recovery following spinal cord injury (SCI). In animal SCI models, intrathecal delivery of anti-Nogo-A antibodies promoted regenerative neurite growth and functional recovery. OBJECTIVE: This first-in-man study assessed the feasibility, safety, tolerability, pharmacokinetics, and preliminary efficacy of the human anti-Nogo-A antibody ATI355 following intrathecal administration in patients with acute, complete traumatic paraplegia and tetraplegia. METHODS: Patients (N = 52) started treatment 4 to 60 days postinjury. Four consecutive dose-escalation cohorts received 5 to 30 mg/2.5 mL/day continuous intrathecal ATI355 infusion over 24 hours to 28 days. Following pharmacokinetic evaluation, 2 further cohorts received a bolus regimen (6 intrathecal injections of 22.5 and 45 mg/3 mL, respectively, over 4 weeks). RESULTS: ATI355 was well tolerated up to 1-year follow-up. All patients experienced ≥1 adverse events (AEs). The 581 reported AEs were mostly mild and to be expected following acute SCI. Fifteen patients reported 16 serious AEs, none related to ATI355; one bacterial meningitis case was considered related to intrathecal administration. ATI355 serum levels showed dose-dependency, and intersubject cerebrospinal fluid levels were highly variable after infusion and bolus injection. In 1 paraplegic patient, motor scores improved by 8 points. In tetraplegic patients, mean total motor scores increased, with 3/19 gaining >10 points, and 1/19 27 points at Week 48. Conversion from complete to incomplete SCI occurred in 7/19 patients with tetraplegia. CONCLUSIONS: ATI335 was well tolerated in humans; efficacy trials using intrathecal antibody administration may be considered in acute SCI.


Asunto(s)
Inmunoglobulina G/uso terapéutico , Regeneración Nerviosa/efectos de los fármacos , Neuritas/efectos de los fármacos , Proteínas Nogo/inmunología , Paraplejía/tratamiento farmacológico , Cuadriplejía/tratamiento farmacológico , Traumatismos de la Médula Espinal/tratamiento farmacológico , Adolescente , Adulto , Relación Dosis-Respuesta a Droga , Estudios de Factibilidad , Femenino , Humanos , Inmunoglobulina G/administración & dosificación , Inyecciones Espinales , Masculino , Persona de Mediana Edad , Paraplejía/etiología , Cuadriplejía/etiología , Recuperación de la Función/efectos de los fármacos , Resultado del Tratamiento , Adulto Joven
3.
J Neurotrauma ; 35(23): 2837-2849, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-29690837

RESUMEN

Traumatic brain injury (TBI) commonly results in injury to the components of the white matter tracts, causing post-injury cognitive deficits. The myelin-producing oligodendrocytes (OLs) are vulnerable to TBI, although may potentially be replaced by proliferating oligodendrocyte progenitor cells (OPCs). The cytokine interleukin-1ß (IL-1ß) is a key mediator of the complex inflammatory response, and when neutralized in experimental TBI, behavioral outcome was improved. To evaluate the role of IL-1ß on oligodendrocyte cell death and OPC proliferation, 116 adult male mice subjected to sham injury or the central fluid percussion injury (cFPI) model of traumatic axonal injury, were analyzed at two, seven, and 14 days post-injury. At 30 min post-injury, mice were randomly administered an IL-1ß neutralizing or a control antibody. OPC proliferation (5-ethynyl 2'- deoxyuridine (EdU)/Olig2 co-labeling) and mature oligodendrocyte cell loss was evaluated in injured white matter tracts. Microglia/macrophages immunohistochemistry and ramification using Sholl analysis were also evaluated. Neutralizing IL-1ß resulted in attenuated cell death, indicated by cleaved caspase-3 expression, and attenuated loss of mature OLs from two to seven days post-injury in brain-injured animals. IL-1ß neutralization also attenuated the early, two day post-injury increase of microglia/macrophage immunoreactivity and altered their ramification. The proliferation of OPCs in brain-injured animals was not altered, however. Our data suggest that IL-1ß is involved in the TBI-induced loss of OLs and early microglia/macrophage activation, although not the OPC proliferation. Attenuated oligodendrocyte cell loss may contribute to the improved behavioral outcome observed by IL-1ß neutralization in this mouse model of diffuse TBI.


Asunto(s)
Traumatismos Difusos del Encéfalo/patología , Lesiones Traumáticas del Encéfalo/patología , Interleucina-1beta/antagonistas & inhibidores , Oligodendroglía/patología , Animales , Traumatismos Difusos del Encéfalo/inmunología , Traumatismos Difusos del Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/inmunología , Lesiones Traumáticas del Encéfalo/metabolismo , Interleucina-1beta/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Distribución Aleatoria
4.
Neurosci Lett ; 660: 109-114, 2017 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-28923481

RESUMEN

Triggering receptor expressed in myeloid cells (TREM2) is a member of the immunoglobulin superfamily and is expressed in macrophages, dendritic cells, microglia, and osteoclasts. TREM2 plays a role in phagocytosis, regulates release of cytokine, contributes to microglia maintenance, and its ectodomain is shed from the cell surface. Here, the question was addressed at which position sheddases cleave TREM2 and what are the proteases involved in this process. Using both pharmacological and genetic approaches we report that the main protease contributing to the release of TREM2 ectodomain is ADAM17, (a disintegrin and metalloproteinase domain containing protein, also called TACE, TNFα converting enzyme) while ADAM10 plays a minor role. Complementary biochemical experiments reveal that cleavage occurs between histidine 157 and serine 158. Shedding is not altered for the R47H-mutated TREM2 protein that confers an increased risk for the development of Alzheimers disease. These findings reveal a link between shedding of TREM2 and its regulation during inflammatory conditions or chronic neurodegenerative disease like AD in which activity or expression of sheddases might be altered.


Asunto(s)
Proteína ADAM17/metabolismo , Histidina/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores Inmunológicos/metabolismo , Proteína ADAM10/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Células CHO , Línea Celular , Cricetulus , Humanos , Proteínas de la Membrana/metabolismo
5.
J Neuroinflammation ; 13(1): 189, 2016 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-27549131

RESUMEN

BACKGROUND: The fractalkine (CX3CR1) ligand is expressed in astrocytes and reported to be neuroprotective. When cleaved from the membrane, soluble fractalkine (sCX3CL1) activates the receptor CX3CR1. Although somewhat controversial, CX3CR1 is reported to be expressed in neurons and microglia. The membrane-bound form of CX3CL1 additionally acts as an adhesion molecule for microglia and infiltrating white blood cells. Much research has been done on the role of fractalkine in neuronal cells; however, little is known about the regulation of the CX3CL1 ligand in astrocytes. METHODS: The mechanisms involved in the up-regulation and cleavage of CX3CL1 from human astrocytes were investigated using immunocytochemistry, Q-PCR and ELISA. All statistical analysis was performed using GraphPad Prism 5. RESULTS: A combination of ADAM17 (TACE) and ADAM10 protease inhibitors was found to attenuate IL-1ß-, TNF-α- and IFN-γ-induced sCX3CL1 levels in astrocytes. A specific ADAM10 (but not ADAM17) inhibitor also attenuated these effects, suggesting ADAM10 proteases induce release of sCX3CL1 from stimulated human astrocytes. A p38 MAPK inhibitor also attenuated the levels of sCX3CL1 upon treatment with IL-1ß, TNF-α or IFN-γ. In addition, an IKKß inhibitor significantly reduced the levels of sCX3CL1 induced by IL-1ß or TNF-α in a concentration-dependent manner, suggesting a role for the NF-kB pathway. CONCLUSIONS: In conclusion, this study shows that the release of soluble astrocytic fractalkine is regulated by ADAM10 proteases with p38 MAPK also playing a role in the fractalkine shedding event. These findings are important for understanding the role of CX3CL1 in healthy and stimulated astrocytes and may benefit our understanding of this pathway in neuro-inflammatory and neurodegenerative diseases.


Asunto(s)
Proteína ADAM10/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Quimiocina CX3CL1/metabolismo , Citocinas/farmacología , Proteínas de la Membrana/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Células Cultivadas , Quimiocina CX3CL1/genética , Medio de Cultivo Libre de Suero/farmacología , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Humanos , Metaloproteinasas de la Matriz/metabolismo , ARN Mensajero/metabolismo , Factores de Tiempo
6.
Eur J Neurosci ; 43(8): 1016-33, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27091435

RESUMEN

Widespread traumatic axonal injury (TAI) results in brain network dysfunction, which commonly leads to persisting cognitive and behavioural impairments following traumatic brain injury (TBI). TBI induces a complex neuroinflammatory response, frequently located at sites of axonal pathology. The role of the pro-inflammatory cytokine interleukin (IL)-1ß has not been established in TAI. An IL-1ß-neutralizing or a control antibody was administered intraperitoneally at 30 min following central fluid percussion injury (cFPI), a mouse model of widespread TAI. Mice subjected to moderate cFPI (n = 41) were compared with sham-injured controls (n = 20) and untreated, naive mice (n = 9). The anti-IL-1ß antibody reached the target brain regions in adequate therapeutic concentrations (up to ~30 µg/brain tissue) at 24 h post-injury in both cFPI (n = 5) and sham-injured (n = 3) mice, with lower concentrations at 72 h post-injury (up to ~18 µg/g brain tissue in three cFPI mice). Functional outcome was analysed with the multivariate concentric square field (MCSF) test at 2 and 9 days post-injury, and the Morris water maze (MWM) at 14-21 days post-injury. Following TAI, the IL-1ß-neutralizing antibody resulted in an improved behavioural outcome, including normalized behavioural profiles in the MCSF test. The performance in the MWM probe (memory) trial was improved, although not in the learning trials. The IL-1ß-neutralizing treatment did not influence cerebral ventricle size or the number of microglia/macrophages. These findings support the hypothesis that IL-1ß is an important contributor to the processes causing complex cognitive and behavioural disturbances following TAI.


Asunto(s)
Axones/metabolismo , Lesiones Traumáticas del Encéfalo/metabolismo , Interleucina-1beta/metabolismo , Aprendizaje por Laberinto , Animales , Anticuerpos Neutralizantes/inmunología , Axones/patología , Cognición , Interleucina-1beta/inmunología , Locomoción , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/metabolismo , Microglía/patología
7.
Curr Drug Targets ; 17(16): 1882-1893, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26953244

RESUMEN

Multiple sclerosis (MS) is an immune mediated demyelinating disease of the central nervous system (CNS). The importance of immune cells to MS pathology is supported by clinical data linking the depletion of T and B cells, or the prevention of their migration into the brain with significant reduction in relapses and development of new lesions. In vitro studies, preclinical animal models and encouraging data with the anti-IL-17A antibody secukinumab in a small proof of concept study in man, indicate that IL-17A, a key interleukin associated with many inflammatory and autoimmune diseases, may be involved in MS. Not only cells involved in adaptive immune responses such as Th17 cells and cytotoxic T cells, or innate immune responses such as mucosa-associated invariant T (MAIT) cells and γδT cells, but also CNS resident cells such as astrocytes and oligodendrocytes might contribute to the local production of IL-17A. IL-17A synergizes with other proinflammatory cytokines, by inducing the release of additional cytokines, mediators of tissue damage and chemokines, that recruit new inflammatory cells. IL-17A adversely affects the functions of microglia, astrocytes, oligodendrocytes, neurons, neural precursor cells and endothelial cells. Blockade of IL-17A might be beneficial to MS patients not only by inhibiting inflammation and tissue destruction, but also by enhancing repair processes.


Asunto(s)
Interleucina-17/metabolismo , Esclerosis Múltiple/inmunología , Neuroglía/inmunología , Linfocitos T/inmunología , Animales , Astrocitos/inmunología , Citocinas/metabolismo , Evaluación Preclínica de Medicamentos , Humanos , Interleucina-17/inmunología , Terapia Molecular Dirigida , Esclerosis Múltiple/patología , Esclerosis Múltiple/terapia , Oligodendroglía/inmunología , Transducción de Señal
8.
J Neuroinflammation ; 13: 31, 2016 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-26856814

RESUMEN

BACKGROUND: BAF312 (Siponimod) is a dual agonist at the sphingosine-1 phosphate receptors, S1PR1 and S1PR5. This drug is currently undergoing clinical trials for the treatment of secondary progressive multiple sclerosis (MS). Here, we investigated the effects of BAF312 on isolated astrocyte and microglia cultures as well as in slice culture models of demyelination. METHODS: Mouse and human astrocytes were treated with S1PR modulators and changes in the levels of pERK, pAkt, and calcium signalling as well as S1PR1 internalization and cytokine levels was investigated using Western blotting, immunochemistry, ELISA and confocal microscopy. Organotypic slice cultures were prepared from the cerebellum of 10-day-old mice and treated with lysophosphatidylcholine (LPC), psychosine and/or S1PR modulators, and changes in myelination states were measured by fluorescence of myelin basic protein and neurofilament H. RESULTS: BAF312 treatment of human and mouse astrocytes activated pERK, pAKT and Ca(2+) signalling as well as inducing S1PR1 internalization. Notably, activation of S1PR1 increased pERK and pAKT in mouse astrocytes while both S1PR1 and S1PR3 equally increased pERK and pAKT in human astrocytes, suggesting that the coupling of S1PR1 and S1PR3 to pERK and pAKT differ in mouse and human astrocytes. We also observed that BAF312 moderately attenuated lipopolysaccharide (LPS)- or TNFα/IL17-induced levels of IL6 in both astrocyte and microglia cell cultures. In organotypic slice cultures, BAF312 reduced LPC-induced levels of IL6 and attenuated LPC-mediated demyelination. We have shown previously that the toxic lipid metabolite psychosine induces demyelination in organotypic slice cultures, without altering the levels of cytokines, such as IL6. Importantly, psychosine-induced demyelination was also attenuated by BAF312. CONCLUSIONS: Overall, this study suggests that BAF312 can modulate glial cell function and attenuate demyelination, highlighting this drug as a further potential therapy in demyelinating disorders, beyond MS.


Asunto(s)
Antiinflamatorios/farmacología , Astrocitos/efectos de los fármacos , Azetidinas/farmacología , Compuestos de Bencilo/farmacología , Cerebelo/citología , Enfermedades Desmielinizantes/tratamiento farmacológico , Animales , Animales Recién Nacidos , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/genética , Humanos , Inmunosupresores/farmacología , Técnicas In Vitro , Indanos/farmacología , Interleucina-6/metabolismo , Lisofosfatidilcolinas/farmacología , Ratones , Proteína Básica de Mielina/metabolismo , Técnicas de Cultivo de Órganos , Oxadiazoles/farmacología , Transporte de Proteínas/efectos de los fármacos , Receptores de Lisoesfingolípidos/agonistas , Receptores de Lisoesfingolípidos/antagonistas & inhibidores , Receptores de Lisoesfingolípidos/metabolismo , Tiofenos/farmacología , Factores de Tiempo , beta-Alanina/análogos & derivados , beta-Alanina/farmacología , eIF-2 Quinasa/metabolismo
9.
PLoS One ; 9(6): e99444, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24911000

RESUMEN

The family of sphingosine-1-phosphate receptors (S1PRs) is G-protein-coupled, comprised of subtypes S1PR1-S1PR5 and activated by the endogenous ligand S1P. The phosphorylated version of Fingolimod (pFTY720), an oral therapy for multiple sclerosis (MS), induces S1PR1 internalisation in T cells, subsequent insensitivity to S1P gradients and sequestering of these cells within lymphoid organs, thus limiting immune response. S1PRs are also expressed in neuronal and glial cells where pFTY720 is suggested to directly protect against lysolecithin-induced deficits in myelination state in organotypic cerebellar slices. Of note, the effect of pFTY720 on immune cells already migrated into the CNS, prior to treatment, has not been well established. We have previously found that organotypic slice cultures do contain immune cells, which, in principle, could also be regulated by pFTY720 to maintain levels of myelin. Here, a mouse organotypic cerebellar slice and splenocyte co-culture model was thus used to investigate the effects of pFTY720 on splenocyte-induced demyelination. Spleen cells isolated from myelin oligodendrocyte glycoprotein immunised mice (MOG-splenocytes) or from 2D2 transgenic mice (2D2-splenocytes) both induced demyelination when co-cultured with mouse organotypic cerebellar slices, to a similar extent as lysolecithin. As expected, in vivo treatment of MOG-immunised mice with FTY720 inhibited demyelination induced by MOG-splenocytes. Importantly, in vitro treatment of MOG- and 2D2-splenocytes with pFTY720 also attenuated demyelination caused by these cells. In addition, while in vitro treatment of 2D2-splenocytes with pFTY720 did not alter cell phenotype, pFTY720 inhibited the release of the pro-inflammatory cytokines such as interferon gamma (IFNγ) and interleukin 6 (IL6) from these cells. This work suggests that treatment of splenocytes by pFTY720 attenuates demyelination and reduces pro-inflammatory cytokine release, which likely contributes to enhanced myelination state induced by pFTY720 in organotypic cerebellar slices.


Asunto(s)
Cerebelo/inmunología , Cerebelo/patología , Enfermedades Desmielinizantes/inmunología , Inmunosupresores/farmacología , Glicoles de Propileno/farmacología , Esfingosina/análogos & derivados , Bazo/citología , Bazo/inmunología , Animales , Técnicas de Cocultivo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Clorhidrato de Fingolimod , Mediadores de Inflamación/metabolismo , Ratones , Ratones Transgénicos , Glicoproteína Mielina-Oligodendrócito/inmunología , Fenotipo , Esfingosina/farmacología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Técnicas de Cultivo de Tejidos
10.
Glia ; 62(5): 725-35, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24677511

RESUMEN

The family of interleukin 17 receptors (IL17Rs), subtypes IL17RA-IL17RE, is targeted by the group of pro-inflammatory IL17 cytokines (IL17A-F) and moreover the newly developed anti-IL17A antibody secukinumab (AIN457) has shown promise in Phase II trials in multiple sclerosis. Here, we show that human astrocytes, isolated from a fetal cerebral cortex, express IL17RA and IL17RC and in vitro treatment with IL17A increases protein levels of IL6 in human astrocytes, which is enhanced in the presence of TNFα, as determined by homogeneous time resolved fluorescence. Studies on acutely isolated mouse astrocytes are comparable to human astrocytes although the protein levels of IL6 are lower in mouse astrocytes, which also show a lower response to IL17F and IL1ß in promoting IL6 levels. In human astrocytes, IL17A and TNFα also induce mRNA expression of IL6, IL8 and the Th17 cytokines CXCL1, CXCL2, and CCL20, with little effect on Th1 cytokines CXCL9, CXCL10, and CXCL11. The effects of IL17A are associated with nuclear translocation of the NF-κB transcription factor, as determined by immunocytochemistry, where treatment of human astrocytes with the inhibitors of the NF-κB pathway and with secukinumab inhibits the IL17A and IL17A/TNFα-induced increase in nuclear translocation of NF-κB and levels of IL6. Taken together the data shows that IL17A signaling plays a key role in regulating the levels of cytokines, such as IL6, in human astrocytes via a mechanism that involves NF-κB signaling and that selective inhibition of IL17A signaling attenuates levels of pro-inflammatory molecules in astrocytes.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Interleucina-17/antagonistas & inhibidores , Interleucina-17/farmacología , Interleucina-6/biosíntesis , Animales , Animales Recién Nacidos , Anticuerpos Monoclonales Humanizados , Células Cultivadas , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL
11.
Restor Neurol Neurosci ; 31(6): 733-60, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24064551

RESUMEN

PURPOSE: How are motor maps modified within and in the immediate vicinity of a damaged zone in the motor cortex of non-human primates? METHODS: In eight adult macaque monkeys subjected to a restricted chemical lesion of the hand area in the primary motor cortex (M1), motor maps were established using intracortical micro-stimulation (ICMS) techniques. The monkeys were subdivided into five animals without treatment, whereas three monkeys received an anti-Nogo-A antibody treatment. RESULTS: Following permanent M1 injury, the lesion territory became largely non micro-excitable several months post-lesion, in spite of some recovery of hand function. Few sites within the lesion territory remained excitable, though irrespective to the degree of functional recovery. Around the lesion in M1, there was no reallocation of proximal shoulder/arm territories into distal hand functions. However, ICMS delivered at supra-threshold intensities in these proximal territories elicited digit movements. Post-lesion ICMS thresholds to elicit movements of forelimb muscle territories increased, independently from the degree of functional recovery. Further behavioural evidence for an enhancement of functional recovery promoted by the anti-Nogo-A antibody treatment is provided. CONCLUSION: The degree of functional recovery is not related to a reorganization of motor maps within, and in the vicinity of, a M1 lesion.


Asunto(s)
Lesiones Encefálicas/rehabilitación , Mapeo Encefálico/métodos , Mano/fisiología , Corteza Motora/fisiología , Recuperación de la Función/fisiología , Animales , Estimulación Eléctrica , Electrodos Implantados , Femenino , Lateralidad Funcional/fisiología , Mano/inervación , Bombas de Infusión Implantables , Macaca fascicularis , Masculino , Corteza Motora/lesiones , Destreza Motora/fisiología , Proteínas de la Mielina/metabolismo , Proteínas Nogo , Desempeño Psicomotor/fisiología , Técnicas Estereotáxicas
12.
Front Neurol ; 4: 101, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23885254

RESUMEN

In relation to mechanisms involved in functional recovery of manual dexterity from cervical cord injury or from motor cortical injury, our goal was to determine whether the movements that characterize post-lesion functional recovery are comparable to original movement patterns or do monkeys adopt distinct strategies to compensate the deficits depending on the type of lesion? To this aim, data derived from earlier studies, using a skilled finger task (the modified Brinkman board from which pellets are retrieved from vertical or horizontal slots), in spinal cord and motor cortex injured monkeys were analyzed and compared. Twelve adult macaque monkeys were subjected to a hemi-section of the cervical cord (n = 6) or to a unilateral excitotoxic lesion of the hand representation in the primary motor cortex (n = 6). In addition, in each subgroup, one half of monkeys (n = 3) were treated for 30 days with a function blocking antibody against the neurite growth inhibitory protein Nogo-A, while the other half (n = 3) represented control animals. The motor deficits, and the extent and time course of functional recovery were assessed. For some of the parameters investigated (wrist angle for horizontal slots and movement types distribution for vertical slots after cervical injury; movement types distribution for horizontal slots after motor cortex lesion), post-lesion restoration of the original movement patterns ("true" recovery) led to a quantitatively better functional recovery. In the motor cortex lesion groups, pharmacological reversible inactivation experiments showed that the peri-lesion territory of the primary motor cortex or re-arranged, spared domain of the lesion zone, played a major role in the functional recovery, together with the ipsilesional intact premotor cortex.

13.
Exp Brain Res ; 223(3): 321-40, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22990293

RESUMEN

Following unilateral lesion of the primary motor cortex, the reorganization of callosal projections from the intact hemisphere to the ipsilesional premotor cortex (PM) was investigated in 7 adult macaque monkeys, in absence of treatment (control; n = 4) or treated with function blocking antibodies against the neurite growth inhibitory protein Nogo-A (n = 3). After functional recovery, though incomplete, the tracer biotinylated dextran amine (BDA) was injected in the ipsilesional PM. Retrogradely labelled neurons were plotted in the intact hemisphere and their number was normalized with respect to the volume of the core of BDA injection sites. (1) The callosal projections to PM in the controls originate mainly from homotypic PM areas and, but to a somewhat lesser extent, from the mesial cortex (cingulate and supplementary motor areas). (2) In the lesioned anti-Nogo-A antibody-treated monkeys, the normalized number of callosal retrogradely labelled neurons was up to several folds higher than in controls, especially in the homotypic PM areas. (3) Except one control with a small lesion and a limited, transient deficit, the anti-Nogo-A antibody-treated monkeys recovered to nearly baseline levels of performance (73-90 %), in contrast to persistent deficits in the control monkeys. These results are consistent with a sprouting and/or sparing of callosal axons promoted by the anti-Nogo-A antibody treatment after lesion of the primary motor cortex, as compared to untreated monkeys.


Asunto(s)
Anticuerpos Bloqueadores/administración & dosificación , Encefalopatías/inmunología , Cuerpo Calloso/inmunología , Corteza Motora/inmunología , Proteínas de la Mielina/inmunología , Plasticidad Neuronal/inmunología , Animales , Anticuerpos Bloqueadores/farmacología , Encefalopatías/patología , Encefalopatías/fisiopatología , Cuerpo Calloso/patología , Macaca fascicularis , Corteza Motora/patología , Plasticidad Neuronal/efectos de los fármacos , Proteínas Nogo , Corteza Somatosensorial/citología , Corteza Somatosensorial/inmunología
14.
J Neurotrauma ; 29(3): 567-78, 2012 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-21815784

RESUMEN

Blocking the function of the myelin protein Nogo-A or its signaling pathway is a promising method to overcome an important neurite growth inhibitory factor of the adult central nervous system (CNS), and to enhance axonal regeneration and plasticity after brain or spinal cord injuries. Several studies have shown increased axonal regeneration and enhanced compensatory sprouting, along with substantially improved functional recovery after treatment with anti-Nogo-A antibodies, Nogo-receptor antagonists, or inhibition of the downstream mediator RhoA/ROCK in adult rodents. Proof-of-concept studies in spinal cord-injured macaque monkeys with anti-Nogo-A antibodies have replicated these findings; recently, clinical trials in spinal cord-injured patients have begun. However, the optimal time window for successful Nogo-A function blocking treatments has not yet been determined. We studied the effect of acute as well as 1- or 2-weeks delayed intrathecal anti-Nogo-A antibody infusions on the regeneration of corticospinal tract (CST) axons and the recovery of motor function after large but anatomically incomplete thoracic spinal cord injuries in adult rats. We found that lesioned CST fibers regenerated over several millimeters after acute or 1-week-delayed treatments, but not when the antibody treatment was started with a delay of 2 weeks. Swimming and narrow beam crossing recovered well in rats treated acutely or with a 1-week delay with anti-Nogo-A antibodies, but not in the 2-week-delayed group. These results show that the time frame for treatment of spinal cord lesions with anti-Nogo-A antibodies is restricted to less than 2 weeks in adult rodents.


Asunto(s)
Anticuerpos Bloqueadores/farmacología , Proteínas de la Mielina/antagonistas & inhibidores , Traumatismos de la Médula Espinal/tratamiento farmacológico , Animales , Anticuerpos Bloqueadores/líquido cefalorraquídeo , Proteoglicanos Tipo Condroitín Sulfato/metabolismo , Progresión de la Enfermedad , Femenino , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Locomoción/fisiología , Regeneración Nerviosa/efectos de los fármacos , Proteínas Nogo , Equilibrio Postural/fisiología , Tractos Piramidales/crecimiento & desarrollo , Tractos Piramidales/lesiones , Ratas , Ratas Endogámicas Lew , Recuperación de la Función , Traumatismos de la Médula Espinal/líquido cefalorraquídeo , Traumatismos de la Médula Espinal/patología , Natación/fisiología
15.
J Neurosci ; 31(25): 9323-31, 2011 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-21697382

RESUMEN

Immunization against amyloid-ß (Aß) can reduce amyloid accumulation in vivo and is considered a potential therapeutic approach for Alzheimer's disease. However, it has been associated with meningoencephalitis thought to be mediated by inflammatory T-cells. With the aim of producing an immunogenic vaccine without this side effect, we designed CAD106 comprising Aß1-6 coupled to the virus-like particle Qß. Immunization with this vaccine did not activate Aß-specific T-cells. In APP transgenic mice, CAD106 induced efficacious Aß antibody titers of different IgG subclasses mainly recognizing the Aß3-6 epitope. CAD106 reduced brain amyloid accumulation in two APP transgenic mouse lines. Plaque number was a more sensitive readout than plaque area, followed by Aß42 and Aß40 levels. Studies with very strong overall amyloid reduction showed an increase in vascular Aß, which atypically was nonfibrillar. The efficacy of Aß immunotherapy depended on the Aß levels and thus differed between animal models, brain regions, and stage of amyloid deposition. Therefore, animal studies may not quantitatively predict the effect in human Alzheimer's disease. Our studies provided no evidence for increased microhemorrhages or inflammatory reactions in amyloid-containing brain. In rhesus monkeys, CAD106 induced a similar antibody response as in mice. The antibodies stained amyloid deposits on tissue sections of mouse and human brain but did not label cellular structures containing APP. They reacted with Aß monomers and oligomers and blocked Aß toxicity in cell culture. We conclude that CAD106 immunization is suited to interfere with Aß aggregation and its downstream detrimental effects.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/uso terapéutico , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/inmunología , Inmunoterapia/métodos , Enfermedad de Alzheimer/inmunología , Péptidos beta-Amiloides/efectos adversos , Animales , Células Cultivadas , Ratones , Ratones Transgénicos , Resultado del Tratamiento
16.
Eur J Neurosci ; 34(1): 110-23, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21623956

RESUMEN

Increasing evidence suggests that interleukin-1ß (IL-1ß) is a key mediator of the inflammatory response following traumatic brain injury (TBI). Recently, we showed that intracerebroventricular administration of an IL-1ß-neutralizing antibody was neuroprotective following TBI in mice. In the present study, an anti-IL-1ß antibody or control antibody was administered intraperitoneally following controlled cortical injury (CCI) TBI or sham injury in 105 mice and we extended our histological, immunological and behavioral analysis. First, we demonstrated that the treatment antibody reached target brain regions of brain-injured animals in high concentrations (> 11 nm) remaining up to 8 days post-TBI. At 48 h post-injury, the anti-IL-1ß treatment attenuated the TBI-induced hemispheric edema (P < 0.05) but not the memory deficits evaluated using the Morris water maze (MWM). Neutralization of IL-1ß did not influence the TBI-induced increases (P < 0.05) in the gene expression of the Ccl3 and Ccr2 chemokines, IL-6 or Gfap. Up to 20 days post-injury, neutralization of IL-1ß was associated with improved visuospatial learning in the MWM, reduced loss of hemispheric tissue and attenuation of the microglial activation caused by TBI (P < 0.05). Motor function using the rotarod and cylinder tests was not affected by the anti-IL-1ß treatment. Our results suggest an important negative role for IL-1ß in TBI. The improved histological and behavioral outcome following anti-IL-1ß treatment also implies that further exploration of IL-1ß-neutralizing compounds as a treatment option for TBI patients is warranted.


Asunto(s)
Edema Encefálico/etiología , Edema Encefálico/patología , Lesiones Encefálicas/complicaciones , Lesiones Encefálicas/patología , Trastornos del Conocimiento/etiología , Interleucina-1beta/metabolismo , Animales , Anticuerpos/uso terapéutico , Conducta Animal/fisiología , Edema Encefálico/fisiopatología , Lesiones Encefálicas/fisiopatología , Quimiocinas/genética , Quimiocinas/metabolismo , Trastornos del Conocimiento/fisiopatología , Modelos Animales de Enfermedad , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Aprendizaje/fisiología , Masculino , Memoria/fisiología , Ratones , Ratones Endogámicos C57BL , Microglía/citología , Microglía/metabolismo , Pruebas Neuropsicológicas , Resultado del Tratamiento
18.
Mol Cell Neurosci ; 45(4): 363-9, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20659559

RESUMEN

LINGO-1 is a potent negative regulator of oligodendrocyte differentiation and hence may play a pivotal restrictive role during remyelination in demyelinating diseases such as multiple sclerosis. However, little is known as to which stages of oligodendrocyte differentiation are inhibited by LINGO-1, which domains of the protein are involved and whether accessory proteins are required. Here, we show that LINGO-1 expression in the human oligodendroglial cell line MO3.13 inhibited process extension and this was reversed by an anti-LINGO-1 antibody or the antagonist LINGO-1-Fc. LINGO-1 expression was also found to inhibit myelin basic protein transcription in the rat oligodendroglial cell line CG4. Both of these inhibitory actions of LINGO-1 were abrogated by deletion of the entire ectodomain or cytoplasmic domains but, surprisingly, were unaffected by deletion of the leucine-rich repeats (LRRs). As in neurons, LINGO-1 physically associated with endogenous p75(NTR) in MO3.13 cells and, correspondingly, its inhibition of process extension was reversed by antagonists of p75(NTR). Thus, LINGO-1 inhibits multiple aspects of oligodendrocyte differentiation independently of the LRRs via a process that requires p75(NTR) signalling.


Asunto(s)
Diferenciación Celular/fisiología , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Oligodendroglía/citología , Oligodendroglía/metabolismo , Receptor de Factor de Crecimiento Nervioso/metabolismo , Animales , Western Blotting , Línea Celular , Separación Celular , Citometría de Flujo , Humanos , Inmunoprecipitación , Proteína Básica de Mielina/metabolismo , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transcripción Genética
19.
Eur J Neurosci ; 30(3): 385-96, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19614750

RESUMEN

Interleukin-1beta (IL-1beta) may play a central role in the inflammatory response following traumatic brain injury (TBI). We subjected 91 mice to controlled cortical impact (CCI) brain injury or sham injury. Beginning 5 min post-injury, the IL-1beta neutralizing antibody IgG2a/k (1.5 microg/mL) or control antibody was infused at a rate of 0.25 microL/h into the contralateral ventricle for up to 14 days using osmotic minipumps. Neutrophil and T-cell infiltration and microglial activation was evaluated at days 1-7 post-injury. Cognition was assessed using Morris water maze, and motor function using rotarod and cylinder tests. Lesion volume and hemispheric tissue loss were evaluated at 18 days post-injury. Using this treatment strategy, cortical and hippocampal tissue levels of IgG2a/k reached 50 ng/mL, sufficient to effectively inhibit IL-1betain vitro. IL-1beta neutralization attenuated the CCI-induced cortical and hippocampal microglial activation (P < 0.05 at post-injury days 3 and 7), and cortical infiltration of neutrophils (P < 0.05 at post-injury day 7). There was only a minimal cortical infiltration of activated T-cells, attenuated by IL-1beta neutralization (P < 0.05 at post-injury day 7). CCI induced a significant deficit in neurological motor and cognitive function, and caused a loss of hemispheric tissue (P < 0.05). In brain-injured animals, IL-1beta neutralizing treatment resulted in reduced lesion volume, hemispheric tissue loss and attenuated cognitive deficits (P < 0.05) without influencing neurological motor function. Our results indicate that IL-1beta is a central component in the post-injury inflammatory response that, in view of the observed positive neuroprotective and cognitive effects, may be a suitable pharmacological target for the treatment of TBI.


Asunto(s)
Lesiones Encefálicas/inmunología , Inflamación/metabolismo , Interleucina-1beta/inmunología , Animales , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/fisiopatología , Cognición/fisiología , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Inflamación/fisiopatología , Interleucina-1beta/metabolismo , Masculino , Aprendizaje por Laberinto , Ratones , Ratones Endogámicos C57BL , Microglía/inmunología , Infiltración Neutrófila/inmunología , Prueba de Desempeño de Rotación con Aceleración Constante , Linfocitos T/inmunología
20.
Eur J Neurosci ; 29(5): 983-96, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19291225

RESUMEN

In rodents and nonhuman primates subjected to spinal cord lesion, neutralizing the neurite growth inhibitor Nogo-A has been shown to promote regenerative axonal sprouting and functional recovery. The goal of the present report was to re-examine the data on the recovery of the primate manual dexterity using refined behavioral analyses and further statistical assessments, representing secondary outcome measures from the same manual dexterity test. Thirteen adult monkeys were studied; seven received an anti-Nogo-A antibody whereas a control antibody was infused into the other monkeys. Monkeys were trained to perform the modified Brinkman board task requiring opposition of index finger and thumb to grasp food pellets placed in vertically and horizontally oriented slots. Two parameters were quantified before and following spinal cord injury: (i) the standard 'score' as defined by the number of pellets retrieved within 30 s from the two types of slots; (ii) the newly introduced 'contact time' as defined by the duration of digit contact with the food pellet before successful retrieval. After lesion the hand was severely impaired in all monkeys; this was followed by progressive functional recovery. Remarkably, anti-Nogo-A antibody-treated monkeys recovered faster and significantly better than control antibody-treated monkeys, considering both the score for vertical and horizontal slots (Mann-Whitney test: P = 0.05 and 0.035, respectively) and the contact time (P = 0.008 and 0.005, respectively). Detailed analysis of the lesions excluded the possibility that this conclusion may have been caused by differences in lesion properties between the two groups of monkeys.


Asunto(s)
Anticuerpos/uso terapéutico , Lateralidad Funcional/efectos de los fármacos , Proteínas de la Mielina/inmunología , Recuperación de la Función/efectos de los fármacos , Traumatismos de la Médula Espinal/tratamiento farmacológico , Traumatismos de la Médula Espinal/fisiopatología , Animales , Conducta Animal/efectos de los fármacos , Vértebras Cervicales/patología , Femenino , Lateralidad Funcional/fisiología , Macaca fascicularis , Macaca mulatta , Masculino , Proteínas Nogo , Desempeño Psicomotor/efectos de los fármacos , Recuperación de la Función/fisiología , Traumatismos de la Médula Espinal/patología , Estadísticas no Paramétricas , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...