Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Clin Exp Immunol ; 214(1): 1-17, 2023 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-37410892

RESUMO

Multiple sclerosis (MS) is characterized by the chronic inflammatory destruction of myelinated axons in the central nervous system. Several ideas have been put forward to clarify the roles of the peripheral immune system and neurodegenerative events in such destruction. Yet, none of the resulting models appears to be consistent with all the experimental evidence. They also do not answer the question of why MS is exclusively seen in humans, how Epstein-Barr virus contributes to its development but does not immediately trigger it, and why optic neuritis is such a frequent early manifestation in MS. Here we describe a scenario for the development of MS that unifies existing experimental evidence as well as answers the above questions. We propose that all manifestations of MS are caused by a series of unfortunate events that usually unfold over a longer period of time after a primary EBV infection and involve periodic weakening of the blood-brain barrier, antibody-mediated CNS disturbances, accumulation of the oligodendrocyte stress protein αB-crystallin and self-sustaining inflammatory damage.


Assuntos
Infecções por Vírus Epstein-Barr , Esclerose Múltipla , Humanos , Infecções por Vírus Epstein-Barr/complicações , Herpesvirus Humano 4 , Sistema Nervoso Central , Barreira Hematoencefálica/patologia
2.
J Immunol ; 202(8): 2421-2430, 2019 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-30804043

RESUMO

TLR-induced signaling potently activates cells of the innate immune system and is subject to regulation at different levels. Inflammatory conditions are associated with increased levels of extracellular adenosine, which can modulate TLR-induced production of cytokines through adenosine receptor-mediated signaling. There are four adenosine receptor subtypes that induce different signaling cascades. In this study, we demonstrate a pivotal contribution of adenosine A3 receptor (A3R)-mediated signaling to the TLR4-induced expression of IL-12 in different types of human myeloid APC. In dendritic cells, IL-12 and CCL2 responses as evoked by TLR2, 3, 4, 5, and 8, as well as IL-12 responses evoked by whole pathogens, were all reduced when A3R-mediated signaling was blocked. As a result, concomitant production of IFN-γ and IL-17 by T cells was significantly inhibited. We further show that selective inhibition of A3R-mediated signaling reduced TLR-induced phosphorylation of the transcription factor STAT1 at tyrosine 701. Next-generation sequencing revealed that A3R-mediated signaling controls the expression of metallothioneins, known inhibitors of STAT1 phosphorylation. Together our results reveal a novel regulatory layer of innate immune responses, with a central role for metallothioneins and autocrine/paracrine signaling via A3Rs.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Quimiocina CCL2/imunologia , Interleucina-12/imunologia , Células Mieloides/imunologia , Receptor A3 de Adenosina/imunologia , Transdução de Sinais/imunologia , Receptores Toll-Like/imunologia , Células Apresentadoras de Antígenos/citologia , Humanos , Interferon gama/imunologia , Interleucina-17/imunologia , Células Mieloides/citologia , Células THP-1
3.
Glia ; 65(3): 460-473, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28063173

RESUMO

The glial stress protein alpha B-crystallin (HSPB5) is an endogenous agonist for Toll-like receptor 2 in CD14+ cells. Following systemic administration, HSPB5 acts as a potent inhibitor of neuroinflammation in animal models and reduces lesion development in multiple sclerosis patients. Here, we show that systemically administered HSPB5 rapidly crosses the blood-brain barrier, implicating microglia as additional targets for HSPB5 along with peripheral monocytes and macrophages. To compare key players in the HSPB5-induced protective response of human macrophages and microglia, we applied weighted gene co-expression network analysis on transcript expression data obtained 1 and 4 h after activation. This approach identified networks of genes that are co-expressed in all datasets, thus reducing the complexity of the nonsynchronous waves of transcripts that appear after activation by HSPB5. In both cell types, HSPB5 activates a network of highly connected genes that appear to be functionally equivalent and consistent with the therapeutic effects of HSPB5 in vivo, since both networks include factors that suppress apoptosis, the production of proinflammatory factors, and the development of adaptive immunity. Yet, hub genes at the core of the network in either cell type were strikingly different. They prominently feature the well-known tolerance-promoting programmed-death ligand 1 as a key player in the macrophage response to HSPB5, and the immune-regulatory enzyme cyclooxygenase-2 (COX-2) in that of microglia. This latter finding indicates that despite its reputation as a potential target for nonsteroidal anti-inflammatory drugs, microglial COX-2 plays a central role in the therapeutic effects of HSPB5 during neuroinflammation. GLIA 2017;65:460-473.


Assuntos
Ciclo-Oxigenase 2/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Microglia/efeitos dos fármacos , Microglia/metabolismo , Cadeia B de alfa-Cristalina/farmacologia , Animais , Encéfalo/citologia , Células Cultivadas , Citocinas/metabolismo , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica/efeitos dos fármacos , Redes Reguladoras de Genes/efeitos dos fármacos , Redes Reguladoras de Genes/fisiologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Tecido Parenquimatoso/citologia , Tecido Parenquimatoso/efeitos dos fármacos , RNA Mensageiro/metabolismo , Fatores de Tempo , Fator de Necrose Tumoral alfa/metabolismo , Cadeia B de alfa-Cristalina/metabolismo
4.
Eur J Immunol ; 46(3): 701-11, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26576501

RESUMO

Pentraxin-3 (PTX3), an acute-phase protein released during inflammation, aids phagocytic clearance of pathogens and apoptotic cells, and plays diverse immunoregulatory roles in tissue injury. In neuroinflammatory diseases, like MS, resident microglia could become activated by endogenous agonists for Toll like receptors (TLRs). Previously we showed a strong TLR2-mediated induction of PTX3 in cultured human microglia and macrophages by HspB5, which accumulates in glia during MS. Given the anti-inflammatory effects of HspB5, we examined the contribution of PTX3 to these effects in MS and its animal model EAE. Our data indicate that TLR engagement effectively induces PTX3 expression in human microglia, and that such expression is readily detectable in MS lesions. Enhanced PTX3 expression is prominently expressed in microglia in preactive MS lesions, and in microglia/macrophages engaged in myelin phagocytosis in actively demyelinating lesions. Yet, we did not detect PTX3 in cerebrospinal fluid of MS patients. PTX3 expression is also elevated in spinal cords during chronic relapsing EAE in Biozzi ABH mice, but the EAE severity and time course in PTX3-deficient mice did not differ from WT mice. Moreover, systemic PTX3 administration did not alter the disease onset or severity. Our findings reveal local functions of PTX3 during neuroinflammation in facilitating myelin phagocytosis, but do not point to a role for PTX3 in controlling the development of autoimmune neuroinflammation.


Assuntos
Encéfalo/imunologia , Proteína C-Reativa/administração & dosagem , Proteína C-Reativa/genética , Encefalomielite Autoimune Experimental/imunologia , Esclerose Múltipla/imunologia , Componente Amiloide P Sérico/administração & dosagem , Componente Amiloide P Sérico/genética , Coluna Vertebral/imunologia , Animais , Encéfalo/patologia , Proteína C-Reativa/líquido cefalorraquidiano , Proteína C-Reativa/imunologia , Modelos Animais de Doenças , Humanos , Inflamação/imunologia , Macrófagos/imunologia , Camundongos , Camundongos Biozzi , Microglia/imunologia , Esclerose Múltipla/patologia , Bainha de Mielina/metabolismo , Fagocitose , Componente Amiloide P Sérico/líquido cefalorraquidiano , Componente Amiloide P Sérico/imunologia , Coluna Vertebral/patologia , Receptores Toll-Like/imunologia , Regulação para Cima
5.
Immunology ; 149(2): 146-56, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27388634

RESUMO

Current therapies for multiple sclerosis (MS) reduce the frequency of relapses by modulating adaptive immune responses but fail to limit the irreversible neurodegeneration driving progressive disability. Experimental autoimmune encephalomyelitis (EAE) in Biozzi ABH mice recapitulates clinical features of MS including relapsing-remitting episodes and secondary-progressive disability. To address the contribution of recurrent inflammatory events and ageing as factors that amplify progressive neurological disease, we examined EAE in 8- to 12-week-old and 12-month-old ABH mice. Compared with the relapsing-remitting (RREAE) and secondary progressive (SPEAE) EAE observed in young mice, old mice developed progressive disease from onset (PEAE) associated with pronounced axonal damage and increased numbers of CD3(+) T cells and microglia/macrophages, but not B cells. Whereas the clinical neurological features of PEAE and SPEAE were comparable, the pathology was distinct. SPEAE was associated with significantly reduced perivascular infiltrates and T-cell numbers in the central nervous system (CNS) compared with PEAE and the acute phase of RREAE. In contrast to perivascular infiltrates that declined during progression from RREAE into SPEAE, the numbers of microglia clusters remained constant. Similar to what is observed during MS, the microglia clusters emerging during EAE were associated with axonal damage and oligodendrocytes expressing heat-shock protein B5, but not lymphocytes. Taken together, our data reveal that the course of EAE is dependent on the age of the mice. Younger mice show a relapsing-remitting phase followed by progressive disease, whereas old mice immediately show progression. This indicates that recurrent episodes of inflammation in the CNS, as well as age, contribute to progressive neurological disease.


Assuntos
Envelhecimento/imunologia , Encefalomielite Autoimune Experimental/imunologia , Esclerose Múltipla/imunologia , Inflamação Neurogênica/imunologia , Oligodendroglia/imunologia , Linfócitos T/imunologia , Cadeia B de alfa-Cristalina/metabolismo , Animais , Apoptose , Células Cultivadas , Progressão da Doença , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos , Estresse Oxidativo , Regulação para Cima , Cadeia B de alfa-Cristalina/genética
6.
Immunology ; 141(3): 302-13, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23981039

RESUMO

Communication between the immune system and the central nervous system (CNS) is exemplified by cross-talk between glia and neurons shown to be essential for maintaining homeostasis. While microglia are actively modulated by neurons in the healthy brain, little is known about the cross-talk between oligodendrocytes and microglia. Oligodendrocytes, the myelin-forming cells in the CNS, are essential for the propagation of action potentials along axons, and additionally serve to support neurons by producing neurotrophic factors. In demyelinating diseases such as multiple sclerosis, oligodendrocytes are thought to be the victims. Here, we review evidence that oligodendrocytes also have strong immune functions, express a wide variety of innate immune receptors, and produce and respond to chemokines and cytokines that modulate immune responses in the CNS. We also review evidence that during stress events in the brain, oligodendrocytes can trigger a cascade of protective and regenerative responses, in addition to responses that elicit progressive neurodegeneration. Knowledge of the cross-talk between microglia and oligodendrocytes may continue to uncover novel pathways of immune regulation in the brain that could be further exploited to control neuroinflammation and degeneration.


Assuntos
Encéfalo/imunologia , Comunicação Celular , Microglia/imunologia , Oligodendroglia/imunologia , Transdução de Sinais , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Encéfalo/fisiopatologia , Citocinas/metabolismo , Doenças Desmielinizantes/imunologia , Doenças Desmielinizantes/metabolismo , Doenças Desmielinizantes/patologia , Exossomos/metabolismo , Proteínas de Choque Térmico/metabolismo , Humanos , Microglia/metabolismo , Microglia/patologia , Bainha de Mielina/metabolismo , Oligodendroglia/metabolismo , Oligodendroglia/patologia , Estresse Fisiológico
7.
Immunology ; 142(2): 151-66, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24329535

RESUMO

Neurodegeneration, the progressive dysfunction and loss of neurons in the central nervous system (CNS), is the major cause of cognitive and motor dysfunction. While neuronal degeneration is well-known in Alzheimer's and Parkinson's diseases, it is also observed in neurotrophic infections, traumatic brain and spinal cord injury, stroke, neoplastic disorders, prion diseases, multiple sclerosis and amyotrophic lateral sclerosis, as well as neuropsychiatric disorders and genetic disorders. A common link between these diseases is chronic activation of innate immune responses including those mediated by microglia, the resident CNS macrophages. Such activation can trigger neurotoxic pathways leading to progressive degeneration. Yet, microglia are also crucial for controlling inflammatory processes, and repair and regeneration. The adaptive immune response is implicated in neurodegenerative diseases contributing to tissue damage, but also plays important roles in resolving inflammation and mediating neuroprotection and repair. The growing awareness that the immune system is inextricably involved in mediating damage as well as regeneration and repair in neurodegenerative disorders, has prompted novel approaches to modulate the immune system, although it remains whether these approaches can be used in humans. Additional factors in humans include ageing and exposure to environmental factors such as systemic infections that provide additional clues that may be human specific and therefore difficult to translate from animal models. Nevertheless, a better understanding of how immune responses are involved in neuronal damage and regeneration, as reviewed here, will be essential to develop effective therapies to improve quality of life, and mitigate the personal, economic and social impact of these diseases.


Assuntos
Doenças Neurodegenerativas/imunologia , Humanos , Inflamação/imunologia , Doenças Neurodegenerativas/terapia
8.
Acta Neuropathol ; 128(2): 215-29, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24997049

RESUMO

Activated microglia and macrophages play a key role in driving demyelination during multiple sclerosis (MS), but the factors responsible for their activation remain poorly understood. Here, we present evidence for a dual-trigger role of IFN-γ and alpha B-crystallin (HSPB5) in this context. In MS-affected brain tissue, accumulation of the molecular chaperone HSPB5 by stressed oligodendrocytes is a frequent event. We have shown before that this triggers a TLR2-mediated protective response in surrounding microglia, the molecular signature of which is widespread in normal-appearing brain tissue during MS. Here, we show that IFN-γ, which can be released by infiltrated T cells, changes the protective response of microglia and macrophages to HSPB5 into a robust pro-inflammatory classical response. Exposure of cultured microglia and macrophages to IFN-γ abrogated subsequent IL-10 induction by HSPB5, and strongly promoted HSPB5-triggered release of TNF-α, IL-6, IL-12, IL-1ß and reactive oxygen and nitrogen species. In addition, high levels of CXCL9, CXCL10, CXL11, several guanylate-binding proteins and the ubiquitin-like protein FAT10 were induced by combined activation with IFN-γ and HSPB5. As immunohistochemical markers for microglia and macrophages exposed to both IFN-γ and HSPB5, these latter factors were found to be selectively expressed in inflammatory infiltrates in areas of demyelination during MS. In contrast, they were absent from activated microglia in normal-appearing brain tissue. Together, our data suggest that inflammatory demyelination during MS is selectively associated with IFN-γ-induced re-programming of an otherwise protective response of microglia and macrophages to the endogenous TLR2 agonist HSPB5.


Assuntos
Interferon gama/metabolismo , Macrófagos/fisiologia , Microglia/fisiologia , Esclerose Múltipla/imunologia , Cadeia B de alfa-Cristalina/metabolismo , Encéfalo/imunologia , Encéfalo/patologia , Células Cultivadas , Quimiocina CXCL10/metabolismo , Quimiocina CXCL11/metabolismo , Quimiocina CXCL9/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Humanos , Interleucina-10/metabolismo , Interleucina-12/metabolismo , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Macrófagos/patologia , Microglia/patologia , Esclerose Múltipla/patologia , Fator de Necrose Tumoral alfa/metabolismo , Ubiquitinas/metabolismo
9.
Nature ; 448(7152): 474-9, 2007 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-17568699

RESUMO

alphaB-crystallin (CRYAB) is the most abundant gene transcript present in early active multiple sclerosis lesions, whereas such transcripts are absent in normal brain tissue. This crystallin has anti-apoptotic and neuroprotective functions. CRYAB is the major target of CD4+ T-cell immunity to the myelin sheath from multiple sclerosis brain. The pathophysiological implications of this immune response were investigated here. We demonstrate that CRYAB is a potent negative regulator acting as a brake on several inflammatory pathways in both the immune system and central nervous system (CNS). Cryab-/- mice showed worse experimental autoimmune encephalomyelitis (EAE) at the acute and progressive phases, with higher Th1 and Th17 cytokine secretion from T cells and macrophages, and more intense CNS inflammation, compared with their wild-type counterparts. Furthermore, Cryab-/- astrocytes showed more cleaved caspase-3 and more TUNEL staining, indicating an anti-apoptotic function of Cryab. Antibody to CRYAB was detected in cerebrospinal fluid from multiple sclerosis patients and in sera from mice with EAE. Administration of recombinant CRYAB ameliorated EAE. Thus, the immune response against a negative regulator of inflammation, CRYAB, in multiple sclerosis, would exacerbate inflammation and demyelination. This can be countered by giving CRYAB itself for therapy of ongoing disease.


Assuntos
Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/prevenção & controle , Esclerose Múltipla/metabolismo , Esclerose Múltipla/prevenção & controle , Fármacos Neuroprotetores/metabolismo , Cadeia B de alfa-Cristalina/metabolismo , Animais , Apoptose , Astrócitos/metabolismo , Caspase 3/metabolismo , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Humanos , Inflamação/imunologia , Inflamação/patologia , Inflamação/prevenção & controle , Sistema de Sinalização das MAP Quinases , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Esclerose Múltipla/líquido cefalorraquidiano , Esclerose Múltipla/imunologia , Bainha de Mielina/imunologia , Bainha de Mielina/patologia , NF-kappa B/metabolismo , Neuroglia/patologia , Fármacos Neuroprotetores/uso terapêutico , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Auxiliares-Indutores/metabolismo , Cadeia B de alfa-Cristalina/genética , Cadeia B de alfa-Cristalina/uso terapêutico
10.
J Neurosci Res ; 90(2): 388-98, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21971760

RESUMO

Toll-like receptors (TLRs) play a key role in controlling innate immune responses to a wide variety of pathogen-associated molecules as well as endogenous signals. In addition, TLR expression within nonimmune cells has been recognized as as modulator of cell behavior. In this study we have addressed the question of whether functional TLRs are expressed on oligodendrocytes, the myelinating cells of the central nervous system. Primary cultures of rat oligodendrocytes at different maturation stages were found to express TLR2 and, to lesser extent, TLR3. Immunocytochemical analysis revealed that both TLRs were localized at the cell body and primary processes and were excluded from myelin-like membranes. Interestingly, innate immune receptor ligands were able to modulate oligodendrocyte survival, differentiation, and myelin-like membrane formation, indicating that TLRs on oligodendrocytes are functional. In highly purified oligodendrocytes cultures, the TLR2 agonist zymosan promoted survival, differentiation, and myelin-like membrane formation, whereas poly-I:C, a TLR3 ligand, was a potent inducer of apoptosis. Together, these data indicate that, in addition to other neural cell types, also oligodendrocytes express functional TLRs, which play a role in regulating various aspects of oligodendrocyte behavior.


Assuntos
Diferenciação Celular/fisiologia , Bainha de Mielina/fisiologia , Oligodendroglia/citologia , Oligodendroglia/metabolismo , Receptor 2 Toll-Like/agonistas , Receptor 2 Toll-Like/fisiologia , Receptor 3 Toll-Like/agonistas , Receptor 3 Toll-Like/fisiologia , Animais , Animais Recém-Nascidos , Diferenciação Celular/efeitos dos fármacos , Membrana Celular/efeitos dos fármacos , Membrana Celular/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Bainha de Mielina/efeitos dos fármacos , Oligodendroglia/efeitos dos fármacos , Ratos , Ratos Wistar , Zimosan/farmacologia
11.
J Immunol ; 184(12): 6929-37, 2010 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-20483774

RESUMO

TLR3 recognizes dsRNAs and is considered of key importance to antiviral host-defense responses. TLR3 also triggers neuroprotective responses in astrocytes and controls the growth of axons and neuronal progenitor cells, suggesting additional roles for TLR3-mediated signaling in the CNS. This prompted us to search for alternative, CNS-borne protein agonists for TLR3. A genome-scale functional screening of a transcript library from brain tumors revealed that the microtubule regulator stathmin is an activator of TLR3-dependent signaling in astrocytes, inducing the same set of neuroprotective factors as the known TLR3 agonist polyinosinic:polycytidylic acid. This activity of stathmin crucially depends on a long, negatively charged alpha helix in the protein. Colocalization of stathmin with TLR3 on astrocytes, microglia, and neurons in multiple sclerosis-affected human brain indicates that as an endogenous TLR3 agonist, stathmin may fulfill previously unsuspected regulatory roles during inflammation and repair in the adult CNS.


Assuntos
Encéfalo/imunologia , Estatmina/imunologia , Receptor 3 Toll-Like/imunologia , Animais , Astrócitos/imunologia , Astrócitos/metabolismo , Western Blotting , Encéfalo/metabolismo , Biblioteca Gênica , Humanos , Camundongos , Microglia/imunologia , Microglia/metabolismo , Microtúbulos/imunologia , Microtúbulos/metabolismo , Neurônios/imunologia , Neurônios/metabolismo , RNA Interferente Pequeno , Transdução de Sinais/imunologia , Estatmina/metabolismo , Receptor 3 Toll-Like/metabolismo
12.
Front Neurosci ; 14: 574, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32595446

RESUMO

Progressive neuronal death during tauopathies is associated with aggregation of modified, truncated or mutant forms of tau protein. Such aggregates are neurotoxic, promote spreading of tau aggregation, and trigger release of pro-inflammatory factors by glial cells. Counteracting such pathogenic effects of tau by simultaneously inhibiting protein aggregation as well as pro-inflammatory glial cell responses would be of significant therapeutic interest. Here, we examined the use of the small heat-shock protein HspB5 for this purpose. As a molecular chaperone, HspB5 counteracts aggregation of a wide range of abnormal proteins. As a TLR2 agonist, it selectively activates protective responses by CD14-expressing myeloid cells including microglia. We show that intracerebral infusion of HspB5 in transgenic mice with selective neuronal expression of mutant human P301S tau has significant neuroprotective effects in the superficial, frontal cortical layers. Underlying these effects at least in part, HspB5 induces several potent neuroprotective mediators in both astrocytes and microglia including neurotrophic factors and increased potential for removal of glutamate. Together, these findings highlight the potentially broad therapeutic potential of HspB5 in neurodegenerative proteinopathies.

13.
Mult Scler ; 20(8): 1030-2, 2014 07.
Artigo em Inglês | MEDLINE | ID: mdl-24368758
14.
Front Neurol ; 10: 570, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31244750

RESUMO

Sensorineural hearing loss is the most common long-term deficit after pneumococcal meningitis (PM), occurring in up to 30% of surviving patients. The infection and the following overshooting inflammatory host response damage the vulnerable sensory cells of the inner ear, resulting in loss of hair cells and spiral ganglion neurons, ultimately leading to elevated hearing thresholds. Here, we tested the oto-protective properties of the small heat shock protein alpha B-crystallin (HspB5) with previously reported anti-inflammatory, anti-apoptotic and neuroprotective functions, in an experimental model of PM-induced hearing loss. We analyzed the effect of local and systemic delivery of HspB5 in an infant rat model of PM, as well as ex vivo, using whole mount cultures. Cytokine secretion profile, hearing thresholds and inner ear damage were assessed at predefined stages of the disease up to 1 month after infection. PM was accompanied by elevated pro-inflammatory cytokine concentrations in the cerebrospinal fluid (CSF), leukocyte and neutrophil infiltration in the perilymphatic spaces of the cochlea with neutrophils extracellular trap formation during the acute phase of the disease. Elevated hearing thresholds were measured after recovery from meningitis. Intracisternal but not intraperitoneal administration of HspB5 significantly reduced the levels of TNF-α, IL-6 IFN-γ and IL-10 in the acute phase of the disease. This resulted in a greater outer hair cell survival, as well as improved hearing thresholds at later stages. These results suggest that high local concentrations of HspB5 are needed to prevent inner ear damage in acute PM. HspB5 represents a promising therapeutic option to improve the auditory outcome and counteract hearing loss after PM.

16.
Curr Opin Investig Drugs ; 8(1): 60-5, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17263186

RESUMO

Toll-like receptors (TLRs) that play key roles in inflammation are also widely expressed in the CNS. While they are well known to activate inflammatory responses to microbial products, TLRs fulfill additional roles in the absence of infection. Emerging evidence suggests that several TLRs play a role in CNS development during fetal life and in repair during adult life. This review discusses the available data on the expression and function of individual TLR family members in the CNS and clarifies why TLRs deserve close scrutiny as a novel group of therapeutic targets for CNS disorders.


Assuntos
Sistema Nervoso Central/fisiopatologia , Inflamação/fisiopatologia , Receptores Toll-Like/fisiologia , Animais , Sistema Nervoso Central/efeitos dos fármacos , Doenças do Sistema Nervoso Central/fisiopatologia , Doenças do Sistema Nervoso Central/prevenção & controle , Humanos , Inflamação/prevenção & controle , Receptores Toll-Like/agonistas
17.
Nat Biotechnol ; 21(9): 1033-9, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12910246

RESUMO

The diversity of autoimmune responses poses a formidable challenge to the development of antigen-specific tolerizing therapy. We developed 'myelin proteome' microarrays to profile the evolution of autoantibody responses in experimental autoimmune encephalomyelitis (EAE), a model for multiple sclerosis (MS). Increased diversity of autoantibody responses in acute EAE predicted a more severe clinical course. Chronic EAE was associated with previously undescribed extensive intra- and intermolecular epitope spreading of autoreactive B-cell responses. Array analysis of autoantigens targeted in acute EAE was used to guide the choice of autoantigen cDNAs to be incorporated into expression plasmids so as to generate tolerizing vaccines. Tolerizing DNA vaccines encoding a greater number of array-determined myelin targets proved superior in treating established EAE and reduced epitope spreading of autoreactive B-cell responses. Proteomic monitoring of autoantibody responses provides a useful approach to monitor autoimmune disease and to develop and tailor disease- and patient-specific tolerizing DNA vaccines.


Assuntos
Encefalomielite Autoimune Experimental/tratamento farmacológico , Encefalomielite Autoimune Experimental/imunologia , Imunoensaio/métodos , Bainha de Mielina/imunologia , Análise Serial de Proteínas/métodos , Vacinas de DNA/imunologia , Vacinas de DNA/uso terapêutico , Animais , Tolerância a Medicamentos , Encefalomielite Autoimune Experimental/diagnóstico , Camundongos , Esclerose Múltipla/diagnóstico , Esclerose Múltipla/tratamento farmacológico , Esclerose Múltipla/imunologia , Mapeamento de Interação de Proteínas/métodos , Resultado do Tratamento
18.
CNS Neurol Disord Drug Targets ; 16(3): 244-256, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27804858

RESUMO

Heat shock proteins (HSPs) are families of molecular chaperones that play important homeostatic functions in the central nervous system (CNS) by preventing protein misfolding, promoting degradation of improperly folded proteins, and protecting against apoptosis and inflammatory damage especially during hyperthermia, hypoxia, or oxidative stress. Under stress conditions, HSPs are upregulated to protect cells from damage that accumulates during ageing as well as pathological conditions. An important, yet frequently overlooked function of some HSPs is their ability to function as extracellular messengers (also termed chaperokines) that modulate immune responses within the CNS. Given the strong association between protein aggregation, innate immune cell activation and neurodegeneration, the expression and roles of HSPs in the CNS is attracting attention in many neurodegenerative disorders including inflammatory diseases such as multiple sclerosis, protein folding diseases such as Alzheimer's disease and amyotrophic lateral sclerosis, and genetic white matter diseases. This is especially so since several studies show that HSPs act therapeutically by modulating innate immune activation and may thus serve as neuroprotective agents. Here we review the evidence linking HSPs with neurodegenerative disorders in humans and the experimental animal models of these disorders. We discuss the mechanisms by which HSPs protect cells, and how the knowledge of their endogenous functions can be exploited to treat disorders of the CNS.


Assuntos
Sistema Nervoso Central/metabolismo , Proteínas de Choque Térmico/metabolismo , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Animais , Humanos
19.
Drug Discov Today ; 11(1-2): 74-80, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16478694

RESUMO

Research into human central nervous system (CNS) disorders has traditionally focused on interconnecting neurons, thought to be the most important functional elements in the CNS. Consequently, animal models have developed as the central paradigm in CNS drug development. However, evidence is accumulating that suggests glial cells play a much more important role in health and disease in the CNS than has been previously acknowledged. Brain development, neurotransmission, inflammatory and neuroprotective pathways and blood-brain barrier functions rely on glial cells. It is also the case that human glial cell cultures adequately mimic in vivo glial cell behaviour, providing a novel and valuable tool for CNS drug discovery and development.


Assuntos
Astrócitos , Doenças do Sistema Nervoso Central , Desenho de Fármacos , Microglia , Animais , Astrócitos/metabolismo , Células Cultivadas , Humanos , Inflamação , Microglia/metabolismo
20.
J Neuroimmunol ; 176(1-2): 51-62, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16844233

RESUMO

Alpha B-crystallin (alphaB) is a small heat shock protein that is strongly up-regulated in multiple sclerosis (MS) brain tissue, and can induce strong T cell responses. Assessing a potential encephalitogenic function for alphaB protein in MS and experimental autoimmune encephalomyelitis (EAE) has been challenging due to its ubiquitous expression that likely maintains central and peripheral tolerance to this protein in mice. To address this issue, we obtained alphaB-knockout (alphaB-KO) mice in H-2b background that lack immune tolerance to alphaB protein, and thus are capable of developing alphaB-specific T cells that could be tested for encephalitogenic activity after transfer into alphaB-expressing wild type (WT) mice. We found that T cell lines from spleens of alphaB protein-immunized alphaB-KO mice proliferated strongly to alphaB protein itself, and the majority of T cells were CD4+ and capable of secreting pro-inflammatory Th1 cytokines upon restimulation. However, transfer of such alphaB-reactive T cells back into WT recipients was not sufficient to induce EAE, compared to the transfer of mouse MOG-35-55 peptide-reactive T cells from the same donors that induced severe EAE in recipients. Moreover, alphaB-specific T cells failed to augment severity of actively induced EAE in WT mice that were expressing high levels of alphaB message in the CNS at the time of transfer. These results suggest that alphaB-specific T cells are immunocompetent but not encephalitogenic in 129SvEv mice, and that immune tolerance may not be the main factor that limits the encephalitogenic potential of alphaB.


Assuntos
Encefalomielite Autoimune Experimental/etiologia , Linfócitos T/imunologia , Cadeia B de alfa-Cristalina/imunologia , Sequência de Aminoácidos , Animais , Encefalomielite Autoimune Experimental/imunologia , Glicoproteínas/imunologia , Linfonodos/imunologia , Ativação Linfocitária , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Glicoproteína Mielina-Oligodendrócito , Fragmentos de Peptídeos/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Baço/imunologia , Cadeia B de alfa-Cristalina/genética , Cadeia B de alfa-Cristalina/fisiologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa