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

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Eur J Neurosci ; 43(8): 1016-33, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27091435

RESUMO

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.


Assuntos
Axônios/metabolismo , Lesões Encefálicas Traumáticas/metabolismo , Interleucina-1beta/metabolismo , Aprendizagem em Labirinto , Animais , Anticorpos Neutralizantes/imunologia , Axônios/patologia , Cognição , Interleucina-1beta/imunologia , Locomoção , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Microglia/patologia
2.
J Neuroinflammation ; 13: 31, 2016 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-26856814

RESUMO

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.


Assuntos
Anti-Inflamatórios/farmacologia , Astrócitos/efeitos dos fármacos , Azetidinas/farmacologia , Compostos de Benzil/farmacologia , Cerebelo/citologia , Doenças Desmielinizantes/tratamento farmacológico , Animais , Animais Recém-Nascidos , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/genética , Humanos , Imunossupressores/farmacologia , Técnicas In Vitro , Indanos/farmacologia , Interleucina-6/metabolismo , Lisofosfatidilcolinas/farmacologia , Camundongos , Proteína Básica da Mielina/metabolismo , Técnicas de Cultura de Órgãos , Oxidiazóis/farmacologia , Transporte Proteico/efeitos dos fármacos , Receptores de Lisoesfingolipídeo/agonistas , Receptores de Lisoesfingolipídeo/antagonistas & inibidores , Receptores de Lisoesfingolipídeo/metabolismo , Tiofenos/farmacologia , Fatores de Tempo , beta-Alanina/análogos & derivados , beta-Alanina/farmacologia , eIF-2 Quinase/metabolismo
3.
J Neuroinflammation ; 13(1): 189, 2016 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-27549131

RESUMO

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.


Assuntos
Proteína ADAM10/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Quimiocina CX3CL1/metabolismo , Citocinas/farmacologia , Proteínas de Membrana/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Células Cultivadas , Quimiocina CX3CL1/genética , Meios de Cultura Livres de Soro/farmacologia , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Humanos , Metaloproteinases da Matriz/metabolismo , RNA Mensageiro/metabolismo , Fatores de Tempo
4.
Glia ; 62(5): 725-35, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24677511

RESUMO

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.


Assuntos
Anticorpos Monoclonais/farmacologia , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Interleucina-17/antagonistas & inibidores , Interleucina-17/farmacologia , Interleucina-6/biossíntese , Animais , Animais Recém-Nascidos , Anticorpos Monoclonais Humanizados , Células Cultivadas , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL
5.
Nat Med ; 12(7): 790-2, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16819551

RESUMO

In rodents, after spinal lesion, neutralizing the neurite growth inhibitor Nogo-A promotes axonal sprouting and functional recovery. To evaluate this treatment in primates, 12 monkeys were subjected to cervical lesion. Recovery of manual dexterity and sprouting of corticospinal axons were enhanced in monkeys treated with Nogo-A-specific antibody as compared to monkeys treated with control antibody.


Assuntos
Anticorpos/uso terapêutico , Atividade Motora/efeitos dos fármacos , Proteínas da Mielina/imunologia , Doenças do Colo do Útero/imunologia , Animais , Colo do Útero/fisiopatologia , Modelos Animais de Doenças , Feminino , Macaca , Proteínas Nogo
6.
J Neurosci ; 31(25): 9323-31, 2011 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-21697382

RESUMO

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.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Peptídeos beta-Amiloides/uso terapêutico , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/imunologia , Imunoterapia/métodos , Doença de Alzheimer/imunologia , Peptídeos beta-Amiloides/efeitos adversos , Animais , Células Cultivadas , Camundongos , Camundongos Transgênicos , Resultado do Tratamento
7.
Exp Brain Res ; 223(3): 321-40, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22990293

RESUMO

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.


Assuntos
Anticorpos Bloqueadores/administração & dosagem , Encefalopatias/imunologia , Corpo Caloso/imunologia , Córtex Motor/imunologia , Proteínas da Mielina/imunologia , Plasticidade Neuronal/imunologia , Animais , Anticorpos Bloqueadores/farmacologia , Encefalopatias/patologia , Encefalopatias/fisiopatologia , Corpo Caloso/patologia , Macaca fascicularis , Córtex Motor/patologia , Plasticidade Neuronal/efeitos dos fármacos , Proteínas Nogo , Córtex Somatossensorial/citologia , Córtex Somatossensorial/imunologia
8.
Eur J Neurosci ; 34(1): 110-23, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21623956

RESUMO

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.


Assuntos
Edema Encefálico/etiologia , Edema Encefálico/patologia , Lesões Encefálicas/complicações , Lesões Encefálicas/patologia , Transtornos Cognitivos/etiologia , Interleucina-1beta/metabolismo , Animais , Anticorpos/uso terapêutico , Comportamento Animal/fisiologia , Edema Encefálico/fisiopatologia , Lesões Encefálicas/fisiopatologia , Quimiocinas/genética , Quimiocinas/metabolismo , Transtornos Cognitivos/fisiopatologia , Modelos Animais de Doenças , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Aprendizagem/fisiologia , Masculino , Memória/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Microglia/citologia , Microglia/metabolismo , Testes Neuropsicológicos , Resultado do Tratamento
9.
Mol Cell Neurosci ; 45(4): 363-9, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20659559

RESUMO

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.


Assuntos
Diferenciação Celular/fisiologia , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Oligodendroglia/citologia , Oligodendroglia/metabolismo , Receptor de Fator de Crescimento Neural/metabolismo , Animais , Western Blotting , Linhagem Celular , Separação Celular , Citometria de Fluxo , Humanos , Imunoprecipitação , Proteína Básica da Mielina/metabolismo , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transcrição Gênica
10.
CNS Neurosci Ther ; 26(2): 260-269, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31418518

RESUMO

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.


Assuntos
Anticorpos Bloqueadores/uso terapêutico , Fator Neurotrófico Derivado do Encéfalo/uso terapêutico , Transtornos dos Movimentos/tratamento farmacológico , Proteínas Nogo/antagonistas & inibidores , Traumatismos da Medula Espinal/tratamento farmacológico , Animais , Axônios , Medula Cervical/lesões , Mãos , Macaca fascicularis , Masculino , Destreza Motora , Transtornos dos Movimentos/etiologia , Regeneração Nervosa , Desempenho Psicomotor/efeitos dos fármacos , Recuperação de Função Fisiológica , Traumatismos da Medula Espinal/complicações
11.
Eur J Neurosci ; 29(5): 983-96, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19291225

RESUMO

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.


Assuntos
Anticorpos/uso terapêutico , Lateralidade Funcional/efeitos dos fármacos , Proteínas da Mielina/imunologia , Recuperação de Função Fisiológica/efeitos dos fármacos , Traumatismos da Medula Espinal/tratamento farmacológico , Traumatismos da Medula Espinal/fisiopatologia , Animais , Comportamento Animal/efeitos dos fármacos , Vértebras Cervicais/patologia , Feminino , Lateralidade Funcional/fisiologia , Macaca fascicularis , Macaca mulatta , Masculino , Proteínas Nogo , Desempenho Psicomotor/efeitos dos fármacos , Recuperação de Função Fisiológica/fisiologia , Traumatismos da Medula Espinal/patologia , Estatísticas não Paramétricas , Fatores de Tempo
12.
Eur J Neurosci ; 30(3): 385-96, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19614750

RESUMO

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.


Assuntos
Lesões Encefálicas/imunologia , Inflamação/metabolismo , Interleucina-1beta/imunologia , Animais , Lesões Encefálicas/metabolismo , Lesões Encefálicas/fisiopatologia , Cognição/fisiologia , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Inflamação/fisiopatologia , Interleucina-1beta/metabolismo , Masculino , Aprendizagem em Labirinto , Camundongos , Camundongos Endogâmicos C57BL , Microglia/imunologia , Infiltração de Neutrófilos/imunologia , Teste de Desempenho do Rota-Rod , Linfócitos T/imunologia
13.
Pharmacol Ther ; 117(1): 77-93, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17961662

RESUMO

Multiple sclerosis (MS) is an autoimmune, neurological disability with unknown etiology. The current therapies available for MS work by an immunomodulatory action, preventing T-cell- and macrophage-mediated destruction of brain-resident oligodendrocytes and axonal loss. Recently, FTY720 (fingolimod) was shown to significantly reduce relapse rates in MS patients and is currently in Phase III clinical trials. This drug attenuates trafficking of harmful T cells entering the brain by regulating sphingosine-1-phosphate (S1P) receptors. Here, we outline the direct roles that S1P receptors play in the central nervous system (CNS) and discuss additional modalities by which FTY720 may provide direct neuroprotection in MS.


Assuntos
Imunossupressores/farmacologia , Propilenoglicóis/farmacologia , Receptores de Lisoesfingolipídeo/metabolismo , Esfingosina/análogos & derivados , Animais , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Sistemas de Liberação de Medicamentos , Cloridrato de Fingolimode , Humanos , Imunossupressores/uso terapêutico , Esclerose Múltipla/tratamento farmacológico , Esclerose Múltipla/fisiopatologia , Propilenoglicóis/uso terapêutico , Receptores de Lisoesfingolipídeo/efeitos dos fármacos , Esfingosina/farmacologia , Esfingosina/uso terapêutico
14.
BMC Neurosci ; 9: 5, 2008 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-18194520

RESUMO

BACKGROUND: After unilateral cervical cord lesion at the C7/C8 border interrupting the dorsolateral funiculus in adult monkeys, neutralization of Nogo-A using a specific monoclonal antibody promoted sprouting of corticospinal (CS) axons rostral and caudal to the lesion and, in parallel, improved functional recovery. In monkeys lesioned but not treated with the anti-Nogo-A antibody, the CS neurons in the contralesional primary motor cortex (M1) survived to the axotomy, but their soma shrank. Because the anti-Nogo-A treatment induces regeneration and/or sprouting of CS axons, it may improve access to neurotrophic factors. The question therefore arises as to whether anti-Nogo-A treatment prevents the soma shrinkage observed in the contralesional M1? RESULTS: Using the marker SMI-32, a quantitative and qualitative anatomical assessment of the pyramidal neurons in the layer V (thus including the CS cells) in M1 was performed and compared across three groups of animals: intact monkeys (n = 5); monkeys subjected to the cervical cord lesion and treated with a control antibody (n = 4); monkeys with the cervical lesion and treated with anti-Nogo-A antibody (n = 5). SMI-32 positive neurons on the side contralateral to the lesion were generally less well stained than those on the ipsilesional hemisphere, suggesting that they expressed less neurofilaments. Nevertheless, in all three groups of monkeys, the amount of SMI-32 positive neurons in both hemispheres was generally comparable, confirming the notion that most axotomized CS neurons survived. However, shrinkage of CS cell body area was observed in the contralesional hemisphere in the two groups of lesioned monkeys. The cell surface shrinkage was found to be of the same magnitude in the monkeys treated with the anti-Nogo-A antibody as in the control antibody treated monkeys. CONCLUSION: The anti-Nogo-A antibody treatment did not preserve the axotomized CS cells from soma shrinkage, indicating that the anti-Nogo-A antibody treatment affects morphologically the axotomized CS neurons mainly at distal levels, especially the axon collateralization in the cervical cord, and little or not at all at the level of their soma.


Assuntos
Tamanho Celular/efeitos dos fármacos , Vértebras Cervicais , Soros Imunes/farmacologia , Proteínas da Mielina/antagonistas & inibidores , Proteínas da Mielina/imunologia , Traumatismos da Medula Espinal/tratamento farmacológico , Traumatismos da Medula Espinal/patologia , Animais , Haplorrinos , Soros Imunes/administração & dosagem , Macaca fascicularis , Macaca mulatta , Córtex Motor , Neurônios/citologia , Neurônios/efeitos dos fármacos , Proteínas Nogo
15.
Behav Brain Res ; 187(2): 262-72, 2008 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-17963852

RESUMO

Neglect is a complex human cognitive spatial disorder typically induced by damage to prefrontal or posterior parietal association cortices. Behavioral treatments for neglect rarely generalize outside of the therapeutic context or across tasks within the same therapeutic context. Recovery, when it occurs, is spontaneous over the course of weeks to months, but often it is incomplete. A number of studies have indicated that anti-Nogo-A antibodies can be used to enhance plasticity and behavioral recovery following damage to motor cortex, and spinal cord. In the present studies the anti-Nogo-A antibodies IN-1, 7B12, or 11C7 were applied intraventricularly to adult rats demonstrating severe neglect produced by unilateral medial agranular cortex lesions in rats. The three separate anti-Nogo-A antibody groups were treated immediately following the medial agranular cortex lesions. Each of the three antibodies induced dramatic significant behavioral recovery from neglect relative to controls. Severing the corpus callosum to destroy inputs from the contralesional hemisphere resulted in reinstatement of severe neglect, pointing to a possible role of interhemispheric mechanisms in behavioral recovery from neglect.


Assuntos
Córtex Cerebral/fisiologia , Proteínas da Mielina/fisiologia , Plasticidade Neuronal/fisiologia , Transtornos da Percepção/fisiopatologia , Recuperação de Função Fisiológica/fisiologia , Análise de Variância , Animais , Corpo Caloso/fisiologia , Lateralidade Funcional/fisiologia , Atividade Motora/fisiologia , Proteínas Nogo , Distribuição Aleatória , Ratos , Estatísticas não Paramétricas
16.
Brain Res ; 1233: 27-34, 2008 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-18691563

RESUMO

Medial agranular cortex (AGm) has a prominent bilateral projection to the dorsocentral striatum (DCS). We wished to develop a normal baseline by which to assess neuronal plasticity in this corticostriatal system in rats with neglect resulting from a unilateral lesion in AGm, followed by treatment with agents that promote sprouting and functional recovery in other systems. Injections of biotinylated dextran amine were made into AGm in normal rats, and unbiased sampling was used to quantify the density of axons and axonal varicosities present in DCS (the latter represent presynaptic profiles). Labeling density in contralateral DCS is approximately half of that seen in ipsilateral DCS (this ratio is 0.50 for axons, 0.55 for varicosities). The ratio of varicosities is stable over a greater than seven-fold range of absolute densities. There is no consistent relationship between the absolute density of axons and axon varicosities; however, the ratio measures are strongly correlated. We conclude that changes in the contralateral/ipsilateral ratio of axon density after experimental treatments do reflect changes in synaptic density, but axon varicosities are likely to be the most sensitive anatomical parameter by which to assess plasticity at the light microscopic level.


Assuntos
Axônios/fisiologia , Córtex Cerebral/citologia , Vias Neurais/citologia , Plasticidade Neuronal/fisiologia , Sinapses/fisiologia , Animais , Axônios/classificação , Córtex Cerebral/fisiologia , Neostriado/citologia , Neostriado/fisiologia , Vias Neurais/fisiologia , Ratos , Ratos Long-Evans , Valores de Referência , Coloração e Rotulagem , Sinapses/classificação
17.
J Neurotrauma ; 35(23): 2837-2849, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-29690837

RESUMO

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.


Assuntos
Lesões Encefálicas Difusas/patologia , Lesões Encefálicas Traumáticas/patologia , Interleucina-1beta/antagonistas & inibidores , Oligodendroglia/patologia , Animais , Lesões Encefálicas Difusas/imunologia , Lesões Encefálicas Difusas/metabolismo , Lesões Encefálicas Traumáticas/imunologia , Lesões Encefálicas Traumáticas/metabolismo , Interleucina-1beta/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Distribuição Aleatória
18.
Neurorehabil Neural Repair ; 32(6-7): 578-589, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29869587

RESUMO

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.


Assuntos
Imunoglobulina G/uso terapêutico , Regeneração Nervosa/efeitos dos fármacos , Neuritos/efeitos dos fármacos , Proteínas Nogo/imunologia , Paraplegia/tratamento farmacológico , Quadriplegia/tratamento farmacológico , Traumatismos da Medula Espinal/tratamento farmacológico , Adolescente , Adulto , Relação Dose-Resposta a Droga , Estudos de Viabilidade , Feminino , Humanos , Imunoglobulina G/administração & dosagem , Injeções Espinhais , Masculino , Pessoa de Meia-Idade , Paraplegia/etiologia , Quadriplegia/etiologia , Recuperação de Função Fisiológica/efeitos dos fármacos , Resultado do Tratamento , Adulto Jovem
19.
J Comp Neurol ; 502(4): 644-59, 2007 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-17394135

RESUMO

After injury, regrowth of axons in mammalian adult central nervous system is highly limited. However, in monkeys subjected to unilateral cervical lesion (C7-C8 level), neutralization of an important neurite outgrowth inhibitor, Nogo-A, stimulated axonal sprouting caudal to the lesion, accompanied by enhanced functional recovery of manual dexterity, compared with lesioned monkeys treated with a control antibody (Freund et al. [2006] Nat. Med. 12:790-792). The present study aimed at comparing the same two groups of monkeys for axonal sprouting rostral to the cervical lesion. The corticospinal tract was labeled by injecting the anterograde tracer biotinylated dextran amine into the contralesional motor cortex. The corticospinal axons were interrupted at the level of the lesion, accompanied by retrograde axonal degeneration (axon dieback), reflected by the presence of terminal retraction bulbs. The number of terminal retraction bulbs was lower in anti-Nogo-A antibody treated monkeys, and, when present, they were found closer to the lesion than in control-antibody treated monkeys. Compared with control antibody treated monkeys, the anti-Nogo-A antibody treated monkeys exhibited an increased cumulated axon arbor length and a higher number of axon arbors going in the medial direction from the white to the gray matter. Higher in the cervical cord (at C5 level), the anti-Nogo-A treatment enhanced the number of corticospinal fibers crossing the midline, suggesting axonal sprouting. Thus, the anti-Nogo-A antibody treatment enhanced axonal sprouting rostral to the cervical lesion; some of these fibers grew around the lesion and into the caudal spinal segments. These processes paralleled the observed improved functional recovery.


Assuntos
Cones de Crescimento/efeitos dos fármacos , Proteínas da Mielina/antagonistas & inibidores , Regeneração Nervosa/efeitos dos fármacos , Tratos Piramidais/efeitos dos fármacos , Traumatismos da Medula Espinal/tratamento farmacológico , Animais , Anticorpos/farmacologia , Anticorpos/uso terapêutico , Biotina/análogos & derivados , Contagem de Células , Tamanho Celular/efeitos dos fármacos , Dextranos , Feminino , Lateralidade Funcional/fisiologia , Cones de Crescimento/imunologia , Cones de Crescimento/metabolismo , Macaca fascicularis , Macaca mulatta , Masculino , Proteínas da Mielina/metabolismo , Degeneração Neural/tratamento farmacológico , Degeneração Neural/imunologia , Degeneração Neural/fisiopatologia , Regeneração Nervosa/imunologia , Proteínas Nogo , Tratos Piramidais/imunologia , Tratos Piramidais/fisiopatologia , Recuperação de Função Fisiológica/efeitos dos fármacos , Recuperação de Função Fisiológica/imunologia , Traumatismos da Medula Espinal/imunologia , Traumatismos da Medula Espinal/fisiopatologia , Resultado do Tratamento
20.
J Neurosurg ; 107(4): 844-53, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17937233

RESUMO

OBJECT: Central nervous system axons regenerate poorly after traumatic brain injury (TBI), partly due to inhibitors such as the protein Nogo-A present in myelin. The authors evaluated the efficacy of anti-Nogo-A monoclonal antibody (mAb) 7B12 administration on the neurobehavioral and cognitive outcome of rats following lateral fluid-percussion brain injury, characterized the penetration of the 7B12 or control antibodies into target brain regions, and evaluated the effects of Nogo-A inhibition on hemispheric tissue loss and sprouting of uninjured motor tracts in the cervical cord. To elucidate a potential molecular response to Nogo-A inhibition, we evaluated the effects of 7B12 on hippocampal GAP-43 expression. METHODS: Beginning 24 hours after lateral fluid-percussion brain injury or sham injury in rats, the mAb 7B12 or control antibody was infused intracerebroventricularly over 14 days, and behavior was assessed over 4 weeks. RESULTS: Immunoreactivity for 7B12 or immunoglobulin G was detected in widespread brain regions at 1 and 3 weeks postinjury. The brain-injured animals treated with 7B12 showed improvement in cognitive function (p < 0.05) at 4 weeks but no improvement in neurological motor function from 1 to 4 weeks postinjury compared with brain-injured, vehicle-treated controls. The enhanced cognitive function following inhibition of Nogo-A was correlated with an attenuated postinjury downregulation of hippocampal GAP-43 expression (p < 0.05). CONCLUSIONS: Increased GAP-43 expression may be a novel molecular mechanism of the enhanced cognitive recovery mediated by Nogo-A inhibition after TBI in rats.


Assuntos
Anticorpos Monoclonais/farmacologia , Lesões Encefálicas/tratamento farmacológico , Proteína GAP-43/metabolismo , Hipocampo/metabolismo , Proteínas da Mielina/antagonistas & inibidores , Animais , Comportamento Animal , Lesões Encefálicas/metabolismo , Lesões Encefálicas/patologia , Córtex Cerebral/lesões , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Cognição , Regulação para Baixo/fisiologia , Imunoglobulina G/farmacologia , Masculino , Fibras Nervosas Mielinizadas/metabolismo , Fibras Nervosas Mielinizadas/patologia , Regeneração Nervosa/efeitos dos fármacos , Proteínas Nogo , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA