Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Int J Mol Sci ; 25(2)2024 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-38255863

RESUMO

Multiple sclerosis (MS) is an autoimmune disease of the central nervous system (CNS) of still unclear etiology. In recent years, the search for biomarkers facilitating its diagnosis, prognosis, therapy response, and other parameters has gained increasing attention. In this regard, in a previous meta-analysis comprising 22 studies, we found that MS is associated with higher nitrite/nitrate (NOx) levels in the cerebrospinal fluid (CSF) compared to patients with non-inflammatory other neurological diseases (NIOND). However, many of the included studies did not distinguish between the different clinical subtypes of MS, included pre-treated patients, and inclusion criteria varied. As a follow-up to our meta-analysis, we therefore aimed to analyze the serum and CSF NOx levels in clinically well-defined cohorts of treatment-naïve MS patients compared to patients with somatic symptom disorder. To this end, we analyzed the serum and CSF levels of NOx in 117 patients (71 relapsing-remitting (RR) MS, 16 primary progressive (PP) MS, and 30 somatic symptom disorder). We found that RRMS and PPMS patients had higher serum NOx levels compared to somatic symptom disorder patients. This difference remained significant in the subgroup of MRZ-negative RRMS patients. In conclusion, the measurement of NOx in the serum might indeed be a valuable tool in supporting MS diagnosis.


Assuntos
Doenças Autoimunes , Sintomas Inexplicáveis , Esclerose Múltipla Recidivante-Remitente , Esclerose Múltipla , Humanos , Esclerose Múltipla/diagnóstico , Estresse Nitrosativo , Sistema Nervoso Central
2.
Curr Opin Neurol ; 35(3): 307-312, 2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35674073

RESUMO

PURPOSE OF REVIEW: The introduction some 30 years ago of ß-interferon, followed by a panel of immunomodulators and immunosuppressants has led to a remarkable improvement in the management of multiple sclerosis (MS) patients. Despite these noticeable progresses, which lower the number of relapses and thereby ameliorate patients' quality of life, preventing long-term progression of disability is still an unmet need, highlighting the necessity to develop therapeutic strategies aimed at repairing demyelinated lesions and protecting axons from degeneration. The capacity of human brain to self-regenerate demyelinated lesion has opened a field of research aimed at fostering this endogenous potential. RECENT FINDINGS: The pioneer electron microscopic evidence by Périer and Grégoire [Périer O, Grégoire A. Electron microscopic features of multiple sclerosis lesions. Brain 1965; 88:937-952] suggesting the capacity of human brain to self-regenerate demyelinated lesion has opened a field of research aimed at fostering this endogenous potential. Here we review some recently identified mechanisms involved in the remyelination process, focusing on the role of electrical activity and the involvement of innate immune cells. We then provide an update on current strategies promoting endogenous myelin repair. SUMMARY: Identification of therapeutic targets for remyelination has opened an active therapeutic field in MS. Although still in early phase trials, with heterogenous efficacy, the door for myelin regeneration in MS is now opened.


Assuntos
Esclerose Múltipla , Remielinização , Humanos , Esclerose Múltipla/tratamento farmacológico , Bainha de Mielina/patologia , Oligodendroglia/patologia , Qualidade de Vida
3.
J Neuroinflammation ; 19(1): 270, 2022 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-36348455

RESUMO

BACKGROUND: Cladribine is a synthetic purine analogue that interferes with DNA synthesis and repair next to disrupting cellular proliferation in actively dividing lymphocytes. The compound is approved for the treatment of multiple sclerosis (MS). Cladribine can cross the blood-brain barrier, suggesting a potential effect on central nervous system (CNS) resident cells. Here, we explored compartment-specific immunosuppressive as well as potential direct neuroprotective effects of oral cladribine treatment in experimental autoimmune encephalomyelitis (EAE) mice. METHODS: In the current study, we compare immune cell frequencies and phenotypes in the periphery and CNS of EAE mice with distinct grey and white matter lesions (combined active and focal EAE) either orally treated with cladribine or vehicle, using flow cytometry. To evaluate potential direct neuroprotective effects, we assessed the integrity of the primary auditory cortex neuronal network by studying neuronal activity and spontaneous synaptic activity with electrophysiological techniques ex vivo. RESULTS: Oral cladribine treatment significantly attenuated clinical deficits in EAE mice. Ex vivo flow cytometry showed that cladribine administration led to peripheral immune cell depletion in a compartment-specific manner and reduced immune cell infiltration into the CNS. Histological evaluations revealed no significant differences for inflammatory lesion load following cladribine treatment compared to vehicle control. Single cell electrophysiology in acute brain slices was performed and showed an impact of cladribine treatment on intrinsic cellular firing patterns and spontaneous synaptic transmission in neurons of the primary auditory cortex. Here, cladribine administration in vivo partially restored cortical neuronal network function, reducing action potential firing. Both, the effect on immune cells and neuronal activity were transient. CONCLUSIONS: Our results indicate that cladribine exerts a neuroprotective effect after crossing the blood-brain barrier independently of its peripheral immunosuppressant action.


Assuntos
Encefalomielite Autoimune Experimental , Encefalomielite , Fármacos Neuroprotetores , Camundongos , Animais , Encefalomielite Autoimune Experimental/patologia , Cladribina/uso terapêutico , Fármacos Neuroprotetores/farmacologia , Camundongos Endogâmicos C57BL , Modelos Animais de Doenças , Imunossupressores/uso terapêutico
4.
J Neurol Neurosurg Psychiatry ; 93(9): 978-985, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35193952

RESUMO

BACKGROUND: Vaccination has proven to be effective in preventing SARS-CoV-2 transmission and severe disease courses. However, immunocompromised patients have not been included in clinical trials and real-world clinical data point to an attenuated immune response to SARS-CoV-2 vaccines among patients with multiple sclerosis (MS) receiving immunomodulatory therapies. METHODS: We performed a retrospective study including 59 ocrelizumab (OCR)-treated patients with MS who received SARS-CoV-2 vaccination. Anti-SARS-CoV-2-antibody titres, routine blood parameters and peripheral immune cell profiles were measured prior to the first (baseline) and at a median of 4 weeks after the second vaccine dose (follow-up). Moreover, the SARS-CoV-2-specific T cell response and peripheral B cell subsets were analysed at follow-up. Finally, vaccination-related adverse events were assessed. RESULTS: After vaccination, we found anti-SARS-CoV-2(S) antibodies in 27.1% and a SARS-CoV-2-specific T cell response in 92.7% of MS cases. T cell-mediated interferon (IFN)-γ release was more pronounced in patients without anti-SARS-CoV-2(S) antibodies. Antibody titres positively correlated with peripheral B cell counts, time since last infusion and total IgM levels. They negatively correlated with the number of previous infusion cycles. Peripheral plasma cells were increased in antibody-positive patients. A positive correlation between T cell response and peripheral lymphocyte counts was observed. Moreover, IFN-γ release was negatively correlated with the time since the last infusion. CONCLUSION: In OCR-treated patients with MS, the humoral immune response to SARS-CoV-2 vaccination is attenuated while the T cell response is preserved. However, it is still unclear whether T or B cell-mediated immunity is required for effective clinical protection. Nonetheless, given the long-lasting clinical effects of OCR, monitoring of peripheral B cell counts could facilitate individualised treatment regimens and might be used to identify the optimal time to vaccinate.


Assuntos
COVID-19 , Esclerose Múltipla , Vacinas Virais , Anticorpos Monoclonais Humanizados , Anticorpos Antivirais , COVID-19/prevenção & controle , Vacinas contra COVID-19/uso terapêutico , Humanos , Imunidade , Esclerose Múltipla/tratamento farmacológico , Estudos Retrospectivos , SARS-CoV-2 , Vacinação
5.
Mult Scler ; 28(3): 429-440, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-34240656

RESUMO

BACKGROUND: The envelope protein of human endogenous retrovirus W (HERV-W-Env) is expressed by macrophages and microglia, mediating axonal damage in chronic active MS lesions. OBJECTIVE AND METHODS: This phase 2, double-blind, 48-week trial in relapsing-remitting MS with 48-week extension phase assessed the efficacy and safety of temelimab; a monoclonal antibody neutralizing HERV-W-Env. The primary endpoint was the reduction of cumulative gadolinium-enhancing T1-lesions in brain magnetic resonance imaging (MRI) scans at week 24. Additional endpoints included numbers of T2 and T1-hypointense lesions, magnetization transfer ratio, and brain atrophy. In total, 270 participants were randomized to receive monthly intravenous temelimab (6, 12, or 18 mg/kg) or placebo for 24 weeks; at week 24 placebo-treated participants were re-randomized to treatment groups. RESULTS: The primary endpoint was not met. At week 48, participants treated with 18 mg/kg temelimab had fewer new T1-hypointense lesions (p = 0.014) and showed consistent, however statistically non-significant, reductions in brain atrophy and magnetization transfer ratio decrease, as compared with the placebo/comparator group. These latter two trends were sustained over 96 weeks. No safety issues emerged. CONCLUSION: Temelimab failed to show an effect on features of acute inflammation but demonstrated preliminary radiological signs of possible anti-neurodegenerative effects. Current data support the development of temelimab for progressive MS. TRIAL REGISTRATION: CHANGE-MS: ClinicalTrials.gov: NCT02782858, EudraCT: 2015-004059-29; ANGEL-MS: ClinicalTrials.gov: NCT03239860, EudraCT: 2016-004935-18.


Assuntos
Anticorpos Monoclonais Humanizados , Esclerose Múltipla Recidivante-Remitente , Esclerose Múltipla , Anticorpos Monoclonais Humanizados/efeitos adversos , Anticorpos Monoclonais Humanizados/uso terapêutico , Método Duplo-Cego , Produtos do Gene env/uso terapêutico , Humanos , Imageamento por Ressonância Magnética , Esclerose Múltipla/tratamento farmacológico , Esclerose Múltipla Recidivante-Remitente/diagnóstico por imagem , Esclerose Múltipla Recidivante-Remitente/tratamento farmacológico , Esclerose Múltipla Recidivante-Remitente/patologia , Resultado do Tratamento
6.
Proc Natl Acad Sci U S A ; 116(30): 15216-15225, 2019 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-31213545

RESUMO

Axonal degeneration is central to clinical disability and disease progression in multiple sclerosis (MS). Myeloid cells such as brain-resident microglia and blood-borne monocytes are thought to be critically involved in this degenerative process. However, the exact underlying mechanisms have still not been clarified. We have previously demonstrated that human endogenous retrovirus type W (HERV-W) negatively affects oligodendroglial precursor cell (OPC) differentiation and remyelination via its envelope protein pathogenic HERV-W (pHERV-W) ENV (formerly MS-associated retrovirus [MSRV]-ENV). In this current study, we investigated whether pHERV-W ENV also plays a role in axonal injury in MS. We found that in MS lesions, pHERV-W ENV is present in myeloid cells associated with axons. Focusing on progressive disease stages, we could then demonstrate that pHERV-W ENV induces a degenerative phenotype in microglial cells, driving them toward a close spatial association with myelinated axons. Moreover, in pHERV-W ENV-stimulated myelinated cocultures, microglia were found to structurally damage myelinated axons. Taken together, our data suggest that pHERV-W ENV-mediated microglial polarization contributes to neurodegeneration in MS. Thus, this analysis provides a neurobiological rationale for a recently completed clinical study in MS patients showing that antibody-mediated neutralization of pHERV-W ENV exerts neuroprotective effects.


Assuntos
Axônios/virologia , Retrovirus Endógenos/metabolismo , Microglia/virologia , Esclerose Múltipla/genética , Neurônios/virologia , Proteínas do Envelope Viral/genética , Animais , Axônios/metabolismo , Axônios/ultraestrutura , Diferenciação Celular , Ensaios Clínicos Fase II como Assunto , Técnicas de Cocultura , Retrovirus Endógenos/genética , Retrovirus Endógenos/patogenicidade , Feminino , Expressão Gênica , Humanos , Masculino , Microglia/metabolismo , Microglia/ultraestrutura , Esclerose Múltipla/metabolismo , Esclerose Múltipla/patologia , Esclerose Múltipla/virologia , Bainha de Mielina/metabolismo , Bainha de Mielina/ultraestrutura , Bainha de Mielina/virologia , Neurônios/metabolismo , Neurônios/ultraestrutura , Ratos , Ratos Wistar , Proteínas do Envelope Viral/metabolismo
7.
Int J Mol Sci ; 23(21)2022 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-36362063

RESUMO

Siponimod (Mayzent®), a sphingosine 1-phosphate receptor (S1PR) modulator which prevents lymphocyte egress from lymphoid tissues, is approved for the treatment of relapsing-remitting and active secondary progressive multiple sclerosis. It can cross the blood-brain barrier (BBB) and selectively binds to S1PR1 and S1PR5 expressed by several cell populations of the central nervous system (CNS) including microglia. In multiple sclerosis, microglia are a key CNS cell population moving back and forth in a continuum of beneficial and deleterious states. On the one hand, they can contribute to neurorepair by clearing myelin debris, which is a prerequisite for remyelination and neuroprotection. On the other hand, they also participate in autoimmune inflammation and axonal degeneration by producing pro-inflammatory cytokines and molecules. In this study, we demonstrate that siponimod can modulate the microglial reaction to lipopolysaccharide-induced pro-inflammatory activation.


Assuntos
Azetidinas , Esclerose Múltipla , Humanos , Microglia/metabolismo , Compostos de Benzil/farmacologia , Azetidinas/farmacologia , Azetidinas/metabolismo , Esclerose Múltipla/metabolismo
8.
BMC Neurol ; 20(1): 158, 2020 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-32340606

RESUMO

BACKGROUND: Fingolimod (Gilenya®), a first-in-class sphingosine-1-phosphate receptor modulator is approved for the treatment of relapsing-remitting multiple sclerosis. Fingolimod-induced selective immunosuppression leads to an increased risk of opportunistic infections such as cryptococcosis. So far, a total of 8 cases of fingolimod-related cryptococcal meningoencephalitis have been published. CASE PRESENTATION: A 49-year-old female with relapsing-remitting multiple sclerosis presented with cephalgia, fever, confusion and generalized weakness. She had been on fingolimod therapy for the past 5.5 years. Clinical examination suggested meningoencephalitis and laboratory findings showed an IgG2 deficiency. Initially no pathogen could be detected, but after 4 days Cryptococcus neoformans was found in the patient's blood cultures leading to the diagnosis of cryptococcal meningoencephalitis. After antimycotic therapy, her symptoms improved and the patient was discharged. CONCLUSION: MS patients on immunomodulatory  therapy are at constant risk for opportunistic infections. Cephalgia, fever and generalized weakness in combination with fingolimod-induced lymphopenia should be considered a red flag for cryptococcosis.


Assuntos
Criptococose/diagnóstico , Cloridrato de Fingolimode/efeitos adversos , Meningoencefalite/diagnóstico , Esclerose Múltipla Recidivante-Remitente/tratamento farmacológico , Feminino , Cloridrato de Fingolimode/administração & dosagem , Humanos , Imunoglobulina G/imunologia , Imunossupressores/uso terapêutico , Linfopenia/induzido quimicamente , Pessoa de Meia-Idade , Infecções Oportunistas/diagnóstico
9.
Int J Mol Sci ; 21(12)2020 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-32570968

RESUMO

Mesenchymal stem cell (MSC)-secreted factors have been shown to significantly promote oligodendrogenesis from cultured primary adult neural stem cells (aNSCs) and oligodendroglial precursor cells (OPCs). Revealing underlying mechanisms of how aNSCs can be fostered to differentiate into a specific cell lineage could provide important insights for the establishment of novel neuroregenerative treatment approaches aiming at myelin repair. However, the nature of MSC-derived differentiation and maturation factors acting on the oligodendroglial lineage has not been identified thus far. In addition to missing information on active ingredients, the degree to which MSC-dependent lineage instruction is functional in vivo also remains to be established. We here demonstrate that MSC-derived factors can indeed stimulate oligodendrogenesis and myelin sheath generation of aNSCs transplanted into different rodent central nervous system (CNS) regions, and furthermore, we provide insights into the underlying mechanism on the basis of a comparative mass spectrometry secretome analysis. We identified a number of secreted proteins known to act on oligodendroglia lineage differentiation. Among them, the tissue inhibitor of metalloproteinase type 1 (TIMP-1) was revealed to be an active component of the MSC-conditioned medium, thus validating our chosen secretome approach.


Assuntos
Células-Tronco Mesenquimais/citologia , Células-Tronco Neurais/citologia , Oligodendroglia/citologia , Inibidor Tecidual de Metaloproteinase-1/metabolismo , Células-Tronco Adultas/citologia , Animais , Diferenciação Celular , Células Cultivadas , Meios de Cultivo Condicionados/química , Feminino , Células-Tronco Mesenquimais/metabolismo , Cultura Primária de Células , Proteômica , Ratos , Transplante de Células-Tronco
10.
Glia ; 67(1): 160-170, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30430656

RESUMO

Remyelination in the adult CNS depends on activation, differentiation, and functional integration of resident oligodendroglial precursor cells (OPCs) and constitutes the only spontaneous neuroregenerative process able to compensate for functional deficits upon loss of oligodendrocytes and myelin sheaths as it is observed in multiple sclerosis. The proteins encoded by p57kip2- and by human endogenous retrovirus type W (pHERV-W) envelope genes were previously identified as negative regulators of OPC maturation. We here focused on the activity of the ENV protein and investigated how it can be neutralized for an improved myelin repair. We could demonstrate that myelination in vitro is severely affected by this protein but that application of an anti-ENV neutralizing antibody, currently investigated in clinical trials, can rescue the generation of internodes. We then compared p57kip2 and ENV dependent inhibitory mechanisms and found that a dominant negative version of the p57kip2 protein can equally save OPCs from myelination failure in response to ENV-mediated TLR4 activation. Additional experiments addressing p57kip2's underlying mode of action revealed a direct interaction with ATP6v1d, a central component of a vascular ATPase. Its pharmacological blocking was then shown to exert an analogous myelination rescue effect in presence of the ENV protein. Therefore, our study provides mechanistic insights into oligodendroglial inhibition processes and presents three different means to counteract the anti-myelination effect of the ENV protein. These observations are therefore of interest in light of understanding the complexity of the numerous oligodendroglial inhibitors and might promote the establishment of novel regenerative therapies.


Assuntos
Diferenciação Celular/fisiologia , Retrovirus Endógenos , Produtos do Gene env/toxicidade , Bainha de Mielina/fisiologia , Oligodendroglia/fisiologia , Proteínas da Gravidez/toxicidade , Animais , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura , Inibidor de Quinase Dependente de Ciclina p57/farmacologia , Feminino , Humanos , Masculino , Bainha de Mielina/efeitos dos fármacos , Oligodendroglia/efeitos dos fármacos , Ratos , Ratos Wistar
11.
Curr Opin Neurol ; 32(3): 378-384, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30865009

RESUMO

PURPOSE OF REVIEW: Medications for relapsing multiple sclerosis (MS) effectively reduce relapse rate, mitigate disability progression and improve MRI measures of inflammation. However, they have virtually no impact on remyelination which is the major mechanism preventing MS-associated neurodegeneration. Stimulating the generation of myelin-(re)producing cells is therefore a central focus of current MS research and a yet unmet clinical need. Here, we present and evaluate key scientific studies from the field of (therapeutic) remyelination research covering the past 1.5 years. RECENT FINDINGS: On the one hand, recent research in the field of remyelination has strongly focused on repurposing drugs that are already approved for other indications by the Food and Drug Administration or the European Medicines Agency. On the other hand, emerging agents such as the mAbs opicinumab and GNbAC1 target entirely new and unconventional pathways. Some of them have already been tested in clinical trials in which they were found to exert beneficial effects on remyelination as well as on neuroregeneration/neuroprotection. SUMMARY: Several of the agents discussed in this review have shown a high potential as future neuroregenerative drugs. However, future trials with more sensitive clinical and paraclinical primary endpoints will be necessary to prove their effectiveness in MS.


Assuntos
Imunossupressores/uso terapêutico , Esclerose Múltipla/tratamento farmacológico , Bainha de Mielina/efeitos dos fármacos , Regeneração Nervosa/efeitos dos fármacos , Fármacos Neuroprotetores/uso terapêutico , Remielinização/efeitos dos fármacos , Substância Branca/efeitos dos fármacos , Anticorpos Monoclonais , Humanos , Imunossupressores/farmacologia , Imageamento por Ressonância Magnética , Esclerose Múltipla/diagnóstico por imagem , Fármacos Neuroprotetores/farmacologia , Substância Branca/diagnóstico por imagem
12.
Mult Scler ; 25(1): 7-14, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30270725

RESUMO

Current multiple sclerosis (MS) therapies are effective in reducing relapse rate, short-term measures of disability, and magnetic resonance imaging (MRI) measures of inflammation in relapsing remitting MS (RRMS), whereas in progressive/degenerative disease phases these medications are of little or no benefit. Therefore, the development of new therapies aimed at reversing neurodegeneration is of great interest. Remyelination, which is usually a spontaneous endogenous process, is achieved when myelin-producing oligodendrocytes are generated from oligodendrocyte precursor cells (OPCs). Even though these precursor cells are abundant in MS brains, their regeneration capacity is limited. Enhancing the generation of myelin-producing cells is therefore a major focus of MS research. Here we present an overview of the different advancements in the field of remyelination, including suitable animal models for testing remyelination therapies, approved medications with a proposed role in regeneration, myelin repair treatments under investigation in clinical trials, as well as future therapeutics aimed at facilitating myelin repair.


Assuntos
Esclerose Múltipla/tratamento farmacológico , Oligodendroglia/efeitos dos fármacos , Remielinização/efeitos dos fármacos , Animais , Humanos
13.
J Neuroinflammation ; 15(1): 76, 2018 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-29534752

RESUMO

BACKGROUND: Multiple sclerosis (MS) is a neuroinflammatory autoimmune disease of the central nervous system (CNS) which in most cases initially presents with episodes of transient functional deficits (relapsing-remitting MS; RRMS) and eventually develops into a secondary progressive form (SPMS). Aside from neuroimmunological activities, MS is also characterized by neurodegenerative and regenerative processes. The latter involve the restoration of myelin sheaths-electrically insulating structures which are the primary targets of autoimmune attacks. Spontaneous endogenous remyelination takes place even in the adult CNS and is primarily mediated by activation, recruitment, and differentiation of resident oligodendroglial precursor cells (OPCs). However, the overall efficiency of remyelination is limited and further declines with disease duration and progression. From a therapeutic standpoint, it is therefore key to understand how oligodendroglial maturation can be modulated pharmacologically. Teriflunomide has been approved as a first-line treatment for RRMS in the USA and the European Union. As the active metabolite of leflunomide, an established disease-modifying anti-rheumatic drug, it mainly acts via an inhibition of de novo pyrimidine synthesis exerting a cytostatic effect on proliferating B and T cells. METHODS: We investigated teriflunomide-dependent effects on primary rat oligodendroglial homeostasis, proliferation, and differentiation related to cellular processes important for myelin repair hence CNS regeneration in vitro. To this end, several cellular parameters, including specific oligodendroglial maturation markers, in vitro myelination, and p53 family member signaling, were examined by means of gene/protein expression analyses. The rate of myelination was determined using neuron-oligodendrocyte co-cultures. RESULTS: Low teriflunomide concentrations resulted in cell cycle exit while higher doses led to decreased cell survival. Short-term teriflunomide pulses can efficiently promote oligodendroglial cell differentiation suggesting that young, immature cells could benefit from such stimulation. In vitro myelination can be boosted by means of an early stimulation window with teriflunomide. p73 signaling is functionally involved in promoting OPC differentiation and myelination. CONCLUSION: Our findings indicate a critical window of opportunity during which regenerative oligodendroglial activities including myelination of CNS axons can be stimulated by teriflunomide.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Crotonatos/farmacologia , Bainha de Mielina/efeitos dos fármacos , Bainha de Mielina/metabolismo , Oligodendroglia/efeitos dos fármacos , Toluidinas/farmacologia , 2',3'-Nucleotídeo Cíclico Fosfodiesterases/genética , 2',3'-Nucleotídeo Cíclico Fosfodiesterases/metabolismo , Animais , Animais Recém-Nascidos , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Células Cultivadas , Córtex Cerebral/citologia , Técnicas de Cocultura , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Hidroxibutiratos , Carioferinas/genética , Carioferinas/metabolismo , Masculino , Proteína Básica da Mielina/genética , Proteína Básica da Mielina/metabolismo , Fator de Crescimento Neural/farmacologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Nitrilas , Células Precursoras de Oligodendrócitos/efeitos dos fármacos , Ratos , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Proteína Exportina 1
14.
Mult Scler ; 24(6): 696-700, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29629605

RESUMO

BACKGROUND: While there is now a multitude of potent medications for relapsing-remitting multiple sclerosis (RRMS), effective therapies targeting neurodegeneration in progressive multiple sclerosis types are still lacking. Stimulation of neurorepair in this disease remains a pathogenetically defined treatment goal. However, therapeutic progress is slowed by the still inadequate tool set to capture "regeneration/repair" in MS and to define appropriate outcomes in clinical trials. OBJECTIVES: In this review, we discuss studies investigating promising regenerative agents for progressive MS which were recently presented during the European Committee for Treatment and Research in Multiple Sclerosis (ECTRIMS)/Americas Committee for Treatment and Research in Multiple Sclerosis (ACTRIMS) 2017 meeting in Paris.


Assuntos
Esclerose Múltipla Crônica Progressiva/tratamento farmacológico , Regeneração Nervosa/efeitos dos fármacos , Feminino , Humanos , Masculino
16.
J Neurosci ; 35(3): 906-19, 2015 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-25609610

RESUMO

Multiple sclerosis is an autoimmune disease of the CNS resulting in degeneration of myelin sheaths and loss of oligodendrocytes, which means that protection and electrical insulation of axons and rapid signal propagation are impaired, leading to axonal damage and permanent disabilities. Partial replacement of lost oligodendrocytes and remyelination can occur as a result of activation and recruitment of resident oligodendroglial precursor cells. However, the overall remyelination capacity remains inefficient because precursor cells often fail to generate new oligodendrocytes. Increasing evidence points to the existence of several molecular inhibitors that act on these cells and interfere with their cellular maturation. The p57kip2 gene encodes one such potent inhibitor of oligodendroglial differentiation and this study sheds light on the underlying mode of action. We found that subcellular distribution of the p57kip2 protein changed during differentiation of rat, mouse, and human oligodendroglial cells both in vivo and in vitro. Nuclear export of p57kip2 was correlated with promoted myelin expression, higher morphological phenotypes, and enhanced myelination in vitro. In contrast, nuclear accumulation of p57kip2 resulted in blocked oligodendroglial differentiation. Experimental evidence suggests that the inhibitory role of p57kip2 depends on specific interactions with binding proteins such as LIMK-1, CDK2, Mash1, and Hes5 either by controlling their site of action or their activity. Because functional restoration in demyelinating diseases critically depends on the successful generation of oligodendroglial cells, a therapeutic need that is currently unmet, the regulatory mechanism described here might be of particular interest for identifying suitable drug targets and devising novel therapeutic approaches.


Assuntos
Diferenciação Celular/fisiologia , Núcleo Celular/metabolismo , Inibidor de Quinase Dependente de Ciclina p57/metabolismo , Oligodendroglia/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Células Cultivadas , Córtex Cerebelar/metabolismo , Quinase 2 Dependente de Ciclina/metabolismo , Humanos , Quinases Lim/metabolismo , Camundongos , Esclerose Múltipla/metabolismo , Bainha de Mielina/metabolismo , Oligodendroglia/citologia , Transporte Proteico/fisiologia , Ratos , Ratos Wistar , Proteínas Repressoras/metabolismo
17.
Mult Scler ; 21(5): 541-9, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25623245

RESUMO

Myelin destruction due to inflammatory oligodendrocyte cell damage or death in conjunction with axonal degeneration are among the major histopathological hallmarks of multiple sclerosis (MS). The majority of available immunomodulatory medications for MS are approved for relapsing-remitting (RR) MS, for which they reduce relapse rate, MRI measures of inflammation, and the accumulation of disability. These medications are, however, of little benefit during progressive MS where axonal degeneration following demyelination outweighs inflammation. This has sparked great interest in the development of new remyelination therapies aimed at reversing the neurodegenerative damage observed in this disease. Remyelination as a result of oligodendrocyte production from oligodendrocyte precursor cells (OPCs) is considered a promising potential target for the treatment of all stages of MS. In this review we present an overview of a) approved medications (some of them FDA-and EMA-approved for other diseases) with a proposed role in regeneration, b) regenerative treatments under investigation in clinical trials, and c) promising future therapeutic approaches aiming specifically at facilitating endogenous repair.


Assuntos
Esclerose Múltipla/tratamento farmacológico , Esclerose Múltipla/patologia , Bainha de Mielina/efeitos dos fármacos , Humanos , Bainha de Mielina/patologia , Regeneração Nervosa/efeitos dos fármacos , Oligodendroglia
18.
Mult Scler ; 21(9): 1200-3, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25480862

RESUMO

BACKGROUND: The envelope protein (ENV) of the human endogenous retrovirus type W is implicated in inflammatory reactions in multiple sclerosis (MS) but also interferes with oligodendroglial maturation. A neutralizing antibody GNbAC1 has been developed and successfully been tested in clinical trials. OBJECTIVES AND METHODS: We stimulated primary oligodendroglial cells with ENV upon preincubation with GNbAC1 and assessed for nitrosative stress and myelin expression. RESULTS: Neutralization of ENV by GNbAC1 reduces its ability to induce stress reactions resulting in a rescue of myelin expression. CONCLUSIONS: Beyond immune cell modulation, this monoclonal antibody may therefore help to overcome the oligodendroglial differentiation blockade in MS.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Anticorpos Neutralizantes/farmacologia , Diferenciação Celular/efeitos dos fármacos , Oligodendroglia/efeitos dos fármacos , Linhagem Celular , Retrovirus Endógenos , Humanos , Oligodendroglia/citologia , Oligodendroglia/virologia , Proteínas do Envelope Viral
19.
Ann Neurol ; 74(5): 721-32, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23836485

RESUMO

OBJECTIVE: Differentiation of oligodendroglial precursor cells is crucial for central nervous system remyelination and is influenced by both extrinsic and intrinsic factors. Recent studies showed that human endogenous retrovirus type W (HERV-W) contributes significantly to brain damage. In particular, its envelope protein ENV can mediate injury to specific cell types of the brain and immune system. Here, we investigated whether ENV protein affects oligodendroglial differentiation. METHODS: Immunostaining and gene expression analyses were performed to establish the expression and regulation of the known ENV receptor, Toll-like receptor 4 (TLR4), on oligodendroglial precursor cells in human brain tissue and in culture. Cultured primary oligodendroglial precursor cells were stimulated with ENV protein to determine the effects of this ligand/receptor interaction. RESULTS: We demonstrated that the ENV protein is present in close proximity to TLR4-expressing oligodendroglial precursor cells adjacent to multiple sclerosis lesions. Human and rat oligodendroglial precursor cells expressed TLR4, and the ENV-mediated activation of TLR4 led to the induction of proinflammatory cytokines and inducible nitric oxide synthase as well as the formation of nitrotyrosine groups and a subsequent reduction in myelin protein expression. INTERPRETATION: Our findings suggest that ENV-mediated induction of nitrosative stress via activation of TLR4 results in an overall reduction of the oligodendroglial differentiation capacity, thereby contributing to remyelination failure. Therefore, pharmacological or antibody-mediated inhibition of ENV may prevent the blockade of myelin repair in the diseased or injured central nervous system.


Assuntos
Diferenciação Celular , Produtos do Gene env/metabolismo , Bainha de Mielina/metabolismo , Células-Tronco Neurais/citologia , Oligodendroglia/citologia , Proteínas da Gravidez/metabolismo , Receptor 4 Toll-Like/metabolismo , Animais , Células Cultivadas , Produtos do Gene env/genética , Humanos , Bainha de Mielina/genética , Células-Tronco Neurais/metabolismo , Oligodendroglia/metabolismo , Proteínas da Gravidez/genética , Ratos , Receptor 4 Toll-Like/genética
20.
Glia ; 61(8): 1250-60, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23828667

RESUMO

Impaired remyelination in multiple sclerosis (MS) might be due to the failure of oligodendrocyte precursor cells (OPC) to differentiate into myelinating oligodendrocytes. Animal experimental data have shown that p57kip2 inhibits oligodendroglial differentiation, indicating that this factor could contribute to remyelination failure. This study investigates oligodendroglial p57kip2 expression and its association with remyelination in MS lesions. To analyze the potential association of p57kip2 expression with human oligodendroglial maturation, double immunofluorescence staining was performed on brain tissue from 30 MS patients and 20 controls. Anti-p57kip2 antibody was combined with either anti-Nogo-A to label mature oligodendrocytes or anti-Olig2 antibodies to identify immature OPCs. We evaluated MS lesions with or without remyelination, the periplaque white matter (PPWM) as well as control white matter (WM). p57kip2-expressing cells were assessed and correlated with the extent of remyelination. Most Nogo-A-positive oligodendrocytes (range, 87-98%) and all Olig2strong-positive OPCs expressed p57kip2 in MS lesions, in the PPWM and in control WM. p57kip2 expression in oligodendrocytes and OPCs were similar in MS lesions with remyelination compared to MS lesions lacking remyelination. Interestingly, all oligodendroglial lineage cells showed nuclear p57kip2 expression only, with mature oligodendrocytes expressing p57kip2 at low or intermediate levels and OPCs featuring strong expression levels, indicating that this factor may be dynamically expressed during maturation processes. Therefore, p57kip2 appears to be widely expressed in the human oligodendroglial lineage, and potential beneficial effects on remyelination in the MS brain are not based on subcellular p57kip2 localization shifts, as suggested by previous animal experiments.


Assuntos
Inibidor de Quinase Dependente de Ciclina p57/biossíntese , Regulação da Expressão Gênica , Esclerose Múltipla/metabolismo , Esclerose Múltipla/patologia , Oligodendroglia/metabolismo , Oligodendroglia/patologia , Adolescente , Adulto , Idoso , Linhagem da Célula/genética , Criança , Pré-Escolar , Inibidor de Quinase Dependente de Ciclina p57/genética , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Esclerose Múltipla/genética , Bainha de Mielina/genética , Bainha de Mielina/metabolismo , Bainha de Mielina/patologia , Frações Subcelulares/metabolismo , Frações Subcelulares/patologia , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA