Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
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
2.
J Neuroinflammation ; 13: 4, 2016 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-26732432

RESUMO

BACKGROUND: Alzheimer's disease (AD) is the most common neurodegenerative disease. In addition to the occurrence of amyloid deposits and widespread tau pathology, AD is associated with a neuroinflammatory response characterized by the activation of microglia and astrocytes. Protein kinase 2 (CK2, former casein kinase II) is involved in a wide variety of cellular processes. Previous studies on CK2 in AD showed controversial results, and the involvement of CK2 in neuroinflammation in AD remains elusive. METHODS: In this study, we used immunohistochemical and immunofluorescent staining methods to investigate the localization of CK2 in the hippocampus and temporal cortex of patients with AD and non-demented controls. We compared protein levels with Western blotting analysis, and we investigated CK2 activity in human U373 astrocytoma cells and human primary adult astrocytes stimulated with IL-1ß or TNF-α. RESULTS: We report increased levels of CK2 in the hippocampus and temporal cortex of AD patients compared to non-demented controls. Immunohistochemical analysis shows CK2 immunoreactivity in astrocytes in AD and control cases. In AD, the presence of CK2 immunoreactive astrocytes is increased. CK2 immunopositive astrocytes are associated with amyloid deposits, suggesting an involvement of CK2 in the neuroinflammatory response. In U373 cells and human primary astrocytes, the selective CK2 inhibitor CX-4945 shows a dose-dependent reduction of the IL-1ß or TNF-α induced MCP-1 and IL-6 secretion. CONCLUSIONS: This data suggests that CK2 in astrocytes is involved in the neuroinflammatory response in AD. The reduction in pro-inflammatory cytokine secretion by human astrocytes using the selective CK2 inhibitor CX-4945 indicates that CK2 could be a potential target to modulate neuroinflammation in AD.


Assuntos
Doença de Alzheimer/patologia , Astrócitos/enzimologia , Encéfalo/patologia , Idoso , Idoso de 80 Anos ou mais , Amiloide/metabolismo , Astrócitos/efeitos dos fármacos , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patologia , Caseína Quinase II/metabolismo , Células Cultivadas , Citocinas/farmacologia , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Feminino , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Naftiridinas/farmacologia , Fenazinas
3.
Brain ; 137(Pt 1): 92-108, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24287115

RESUMO

Progressive multiple sclerosis is associated with metabolic failure of the axon and excitotoxicity that leads to chronic neurodegeneration. Global sodium-channel blockade causes side effects that can limit its use for neuroprotection in multiple sclerosis. Through selective targeting of drugs to lesions we aimed to improve the potential therapeutic window for treatment. This was assessed in the relapsing-progressive experimental autoimmune encephalomyelitis ABH mouse model of multiple sclerosis using conventional sodium channel blockers and a novel central nervous system-excluded sodium channel blocker (CFM6104) that was synthesized with properties that selectively target the inflammatory penumbra in experimental autoimmune encephalomyelitis lesions. Carbamazepine and oxcarbazepine were not immunosuppressive in lymphocyte-driven autoimmunity, but slowed the accumulation of disability in experimental autoimmune encephalomyelitis when administered during periods of the inflammatory penumbra after active lesion formation, and was shown to limit the development of neurodegeneration during optic neuritis in myelin-specific T cell receptor transgenic mice. CFM6104 was shown to be a state-selective, sodium channel blocker and a fluorescent p-glycoprotein substrate that was traceable. This compound was >90% excluded from the central nervous system in normal mice, but entered the central nervous system during the inflammatory phase in experimental autoimmune encephalomyelitis mice. This occurs after the focal and selective downregulation of endothelial p-glycoprotein at the blood-brain barrier that occurs in both experimental autoimmune encephalomyelitis and multiple sclerosis lesions. CFM6104 significantly slowed down the accumulation of disability and nerve loss in experimental autoimmune encephalomyelitis. Therapeutic-targeting of drugs to lesions may reduce the potential side effect profile of neuroprotective agents that can influence neurotransmission. This class of agents inhibit microglial activity and neural sodium loading, which are both thought to contribute to progressive neurodegeneration in multiple sclerosis and possibly other neurodegenerative diseases.


Assuntos
Benzamidas/uso terapêutico , Indazóis/uso terapêutico , Esclerose Múltipla/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Oxidiazóis/uso terapêutico , Bloqueadores dos Canais de Sódio/uso terapêutico , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Animais , Bancos de Espécimes Biológicos , Encéfalo/patologia , Carbamazepina/farmacologia , Proteínas de Transporte/metabolismo , Proliferação de Células/efeitos dos fármacos , Cromatografia Líquida de Alta Pressão , Sistemas de Liberação de Medicamentos , Encefalomielite Autoimune Experimental/metabolismo , Feminino , Imuno-Histoquímica , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos C57BL , Esclerose Múltipla/fisiopatologia , Neurite Óptica/fisiopatologia , Linfócitos T/efeitos dos fármacos , Uveíte/fisiopatologia , Canais de Sódio Disparados por Voltagem/metabolismo
4.
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
5.
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
6.
Acta Neuropathol ; 122(3): 313-22, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21691765

RESUMO

Hippocampal pathology was shown to be extensive in multiple sclerosis (MS) and is associated with memory impairment. In this post-mortem study, we investigated hippocampal tissue from MS and Alzheimer's disease (AD) patients and compared these to non-neurological controls. By means of biochemical assessment, (immuno)histochemistry and western blot analyses, we detected substantial alterations in the cholinergic neurotransmitter system in the MS hippocampus, which were different from those in AD hippocampus. In MS hippocampus, activity and protein expression of choline acetyltransferase (ChAT), the acetylcholine synthesizing enzyme, was decreased, while the activity and protein expression of acetylcholinesterase (AChE), the acetylcholine degrading enzyme, was found to be unaltered. In contrast, in AD hippocampus both ChAT and AChE enzyme activity and protein expression was decreased. Our findings reveal an MS-specific cholinergic imbalance in the hippocampus, which may be instrumental in terms of future treatment options for memory problems in this disease.


Assuntos
Acetilcolinesterase/metabolismo , Doença de Alzheimer/patologia , Colina O-Acetiltransferase/metabolismo , Hipocampo/enzimologia , Esclerose Múltipla/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/complicações , Feminino , Hipocampo/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Esclerose Múltipla/complicações
7.
Neurobiol Dis ; 36(3): 445-52, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19716418

RESUMO

Parkin is implicated in the pathogenesis of Parkinson's disease. Furthermore, parkin targets misfolded proteins for degradation and protects cells against various forms of cellular stress, including unfolded-protein and oxidative stress. This points towards a protective role of parkin in neurological disorders in which these stressors are implicated, including Alzheimer's disease (AD) and multiple sclerosis (MS). Here, we assessed parkin distribution in AD and MS brain tissue using immunohistochemistry. In AD brains, parkin colocalized with classic senile plaques and amyloid-laden vessels as well as astrocytes associated with both lesions. Similarly, we observed enhanced astrocytic parkin immunoreactivity in MS lesions, particularly in inflammatory lesions. Furthermore, parkin mRNA expression was increased in an astrocytoma cell line after free radical exposure. Our data indicate that parkin is upregulated in AD and MS brain tissue and might represent a defense mechanism to counteract stress-induced damage in AD and MS pathogenesis.


Assuntos
Doença de Alzheimer/metabolismo , Encéfalo/metabolismo , Esclerose Múltipla/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Doença de Alzheimer/patologia , Amiloide/metabolismo , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Astrócitos/patologia , Encéfalo/patologia , Linhagem Celular Tumoral , Feminino , Radicais Livres/toxicidade , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Esclerose Múltipla/patologia , Placa Amiloide/metabolismo , Placa Amiloide/patologia , RNA Mensageiro
9.
Acta Neuropathol Commun ; 3: 87, 2015 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-26694816

RESUMO

INTRODUCTION: The important protective role of small heat-shock proteins (HSPs) in regulating cellular survival and migration, counteracting protein aggregation, preventing apoptosis, and regulating inflammation in the central nervous system is now well-recognized. Yet, their role in the neuroinflammatory disorder multiple sclerosis (MS) is largely undocumented. With the exception of alpha B-crystallin (HSPB5), little is known about the roles of small HSPs in disease. RESULTS: Here, we examined the expression of four small HSPs during lesion development in MS, focussing on their cellular distribution, and regional differences between white matter (WM) and grey matter (GM). It is well known that MS lesions in these areas differ markedly in their pathology, with substantially more intense blood-brain barrier damage, leukocyte infiltration and microglial activation typifying WM but not GM lesions. We analysed transcript levels and protein distribution profiles for HSPB1, HSPB6, HSPB8 and HSPB11 in MS lesions at different stages, comparing them with normal-appearing brain tissue from MS patients and non-neurological controls. During active stages of demyelination in WM, and especially the centre of chronic active MS lesions, we found significantly increased expression of HSPB1, HSPB6 and HSPB8, but not HSPB11. When induced, small HSPs were exclusively found in astrocytes but not in oligodendrocytes, microglia or neurons. Surprisingly, while the numbers of astrocytes displaying high expression of small HSPs were markedly increased in actively demyelinating lesions in WM, no such induction was observed in GM lesions. This difference was particularly obvious in leukocortical lesions covering both WM and GM areas. CONCLUSIONS: Since induction of small HSPs in astrocytes is apparently a secondary response to damage, their differential expression between WM and GM likely reflects differences in mediators that accompany demyelination in either WM or GM during MS. Our findings also suggest that during MS, cortical structures fail to benefit from the protective actions of small HSPs.


Assuntos
Regulação da Expressão Gênica/fisiologia , Substância Cinzenta/metabolismo , Proteínas de Choque Térmico Pequenas/metabolismo , Esclerose Múltipla/patologia , Substância Branca/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Estudos de Casos e Controles , Sistema Nervoso Central/patologia , Feminino , Proteína Glial Fibrilar Ácida/metabolismo , Antígenos HLA-DR/metabolismo , Proteínas de Choque Térmico Pequenas/genética , Humanos , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Proteína Proteolipídica de Mielina/metabolismo , RNA Mensageiro/metabolismo , Estatísticas não Paramétricas
10.
J Neuropathol Exp Neurol ; 74(1): 48-63, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25470347

RESUMO

Similar to macrophages, microglia adopt diverse activation states and contribute to repair and tissue damage in multiple sclerosis. Using reverse transcription-quantitative polymerase chain reaction and immunohistochemistry, we show that in vitro M1-polarized (proinflammatory) human adult microglia express the distinctive markers CD74, CD40, CD86, and CCR7, whereas M2 (anti-inflammatory) microglia express mannose receptor and the anti-inflammatory cytokine CCL22. The expression of these markers was assessed in clusters of activated microglia in normal-appearing white matter (preactive lesions) and areas of remyelination, representing reparative multiple sclerosis lesions. We show that activated microglia in preactive and remyelinating lesions express CD74, CD40, CD86, and the M2 markers CCL22 and CD209, but not mannose receptor. To examine whether this intermediate microglia profile is static or dynamic and thus susceptible to changes in the microenvironment, we polarized microglia into M1 or M2 phenotype in vitro and then subsequently treated them with the opposing polarization regimen. These studies revealed that expression of CD40, CXCL10, and mannose receptor is dynamic and that microglia, like macrophages, can switch between M1 and M2 phenotypic profiles. Taken together, our data define the differential activation states of microglia during lesion development in multiple sclerosis-affected CNS tissues and underscore the plasticity of human adult microglia in vitro.


Assuntos
Encéfalo/patologia , Antígenos de Histocompatibilidade Classe II/metabolismo , Microglia/patologia , Esclerose Múltipla/patologia , Proteína Proteolipídica de Mielina/metabolismo , Idoso , Idoso de 80 Anos ou mais , Antígenos CD/genética , Antígenos CD/metabolismo , Diferenciação Celular/fisiologia , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Feminino , Citometria de Fluxo , Humanos , Macrófagos/patologia , Masculino , Microglia/metabolismo , Pessoa de Meia-Idade , Proteína Proteolipídica de Mielina/genética , RNA Mensageiro/metabolismo , Estatísticas não Paramétricas , Transcriptoma
11.
J Neuropathol Exp Neurol ; 72(10): 970-9, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24042199

RESUMO

Microglial nodules are frequently observed in the normal-appearing white matter of multiple sclerosis (MS) patients. Previously, we have shown that these clusters, which we call "preactive MS lesions," are closely associated with stressed oligodendrocytes and myelin sheaths that contain markedly elevated levels of the small stress protein alpha-B-crystallin (HspB5). Here, we show that microglia in these lesions express the recently identified receptors for HspB5, that is, CD14, Toll-like receptor family 1 and 2 (TLR1 and TLR2), and several molecular markers of the microglial response to HspB5. These markers were identified by genome-wide transcript profiling of 12 primary human microglial cultures at 2 time points after exposure to HspB5. These data indicate that HspB5 activates production by microglia of an array of chemokines, immune-regulatory mediators, and a striking number of antiviral genes that are generally inducible by type I interferons. Together, our data suggest that preactive MS lesions are at least in part driven by HspB5 derived from stressed oligodendrocytes and may reflect a local attempt to restore tissue homeostasis.


Assuntos
Encéfalo/efeitos dos fármacos , Microglia/efeitos dos fármacos , Esclerose Múltipla/metabolismo , Fibras Nervosas Mielinizadas/efeitos dos fármacos , Cadeia B de alfa-Cristalina/farmacologia , Idoso , Idoso de 80 Anos ou mais , Axônios/efeitos dos fármacos , Axônios/metabolismo , Axônios/patologia , Encéfalo/metabolismo , Encéfalo/patologia , Feminino , Humanos , Receptores de Lipopolissacarídeos/genética , Receptores de Lipopolissacarídeos/metabolismo , Masculino , Microglia/imunologia , Microglia/metabolismo , Microglia/patologia , Pessoa de Meia-Idade , Esclerose Múltipla/imunologia , Esclerose Múltipla/patologia , Fibras Nervosas Mielinizadas/metabolismo , Fibras Nervosas Mielinizadas/patologia , Receptor 1 Toll-Like/genética , Receptor 1 Toll-Like/metabolismo , Receptor 2 Toll-Like/genética , Receptor 2 Toll-Like/metabolismo , Cadeia B de alfa-Cristalina/metabolismo
12.
Biomaterials ; 34(3): 831-40, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23117214

RESUMO

As an extracellular protein, the small heat-shock protein alpha B-crystallin (HSPB5) has anti-inflammatory effects in several mouse models of inflammation. Here, we show that these effects are associated with the ability of HSPB5 to activate an immune-regulatory response in macrophages via endosomal/phagosomal CD14 and Toll-like receptors 1 and 2. Humans, however, possess natural antibodies against HSPB5 that block receptor binding. To protect it from these antibodies, we encapsulated HSPB5 in porous PLGA microparticles. We document here size, morphology, protein loading and release characteristics of such microparticles. Apart from effectively protecting HSPB5 from neutralization, PLGA microparticles also strongly promoted macrophage targeting of HSPB via phagocytosis. As a result, HSPB5 in porous PLGA microparticles was more than 100-fold more effective in activating macrophages than free soluble protein. Yet, the immune-regulatory nature of the macrophage response, as documented here by microarray transcript profiling, remained the same. In mice developing cigarette smoke-induced COPD, HSPB5-loaded PLGA microparticles were selectively taken up by alveolar macrophages upon intratracheal administration, and significantly suppressed lung infiltration by lymphocytes and neutrophils. In contrast, 30-fold higher doses of free soluble HSPB5 remained ineffective. Our data indicate that porous HSPB5-PLGA microparticles hold considerable promise as an anti-inflammatory biomaterial for humans.


Assuntos
Anti-Inflamatórios/administração & dosagem , Pulmão/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Pneumonia/complicações , Pneumonia/tratamento farmacológico , Doença Pulmonar Obstrutiva Crônica/complicações , Cadeia B de alfa-Cristalina/administração & dosagem , Animais , Anti-Inflamatórios/imunologia , Anti-Inflamatórios/uso terapêutico , Linhagem Celular , Portadores de Fármacos/química , Proteínas de Choque Térmico Pequenas/administração & dosagem , Proteínas de Choque Térmico Pequenas/imunologia , Proteínas de Choque Térmico Pequenas/uso terapêutico , Humanos , Ácido Láctico/química , Receptores de Lipopolissacarídeos/imunologia , Pulmão/imunologia , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Pneumonia/imunologia , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Receptor 1 Toll-Like/imunologia , Receptor 2 Toll-Like/imunologia , Cadeia B de alfa-Cristalina/imunologia , Cadeia B de alfa-Cristalina/uso terapêutico
13.
Int J Biochem Cell Biol ; 44(10): 1670-9, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22233974

RESUMO

There is now compelling evidence that members of the family of small heat shock proteins (HSP) can be secreted by a variety of different types of cells. Secretion of small HSP may at times represent altruistic delivery of supporting and stabilizing factors from one cell to another. A probably more general effect of extracellular small HSP, however, is exerted by their ability to activate macrophages and macrophage-like cells. When doing so, small HSP induce an immune-regulatory state of activation, stimulating macrophages to suppress inflammation. For this reason, small HSP deserve consideration as broadly applicable therapeutic agents for inflammatory disorders. In one particular case, however, adaptive immune responses to the small HSP itself may subvert the protective quality of the innate immune response it triggers. This situation only applies to alpha B-crystallin, and is unique for humans as well. In this special case, local concentrations of alpha B-crystallin determine the balance between protective innate responses and destructive adaptive responses, the latter of which are held responsible for the development of multiple sclerosis lesions. This article is part of a Directed Issue entitled: Small HSPs in physiology and pathology.


Assuntos
Proteínas de Choque Térmico Pequenas/fisiologia , Imunidade Inata , Mediadores da Inflamação/fisiologia , Esclerose Múltipla/metabolismo , Imunidade Adaptativa , Animais , Exossomos/metabolismo , Proteínas de Choque Térmico Pequenas/metabolismo , Humanos , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/fisiologia , Esclerose Múltipla/imunologia
14.
Brain Pathol ; 21(1): 44-54, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20731658

RESUMO

Multiple Sclerosis (MS) is a neuroinflammatory disease mainly affecting young adults. A major pathological hallmark of MS is the presence of demyelinated lesions in the central nervous system. In the active phase of the disease, astrocytes become activated, migrate and contribute to local tissue remodeling that ultimately can result in an astroglial scar. This process is facilitated by extracellular matrix proteins, including fibronectin. Tissue Transglutaminase (TG2) is a multifunctional enzyme with a ubiquitous tissue distribution and it has been shown that inflammatory cytokines can induce TG2 activity. In addition, TG2 is known to mediate cell adhesion and migration. We therefore hypothesized that TG2 is present in MS lesions and plays a role in cell adhesion and/or migration. Our studies showed that TG2 immunoreactivity appeared in astrocytes in active and chronic active MS lesions. These TG2 positive astrocytes partly co-localized with fibronectin. Additional in vitro studies showed that TG2 mediated astrocytoma adhesion to and migration on the extracellular matrix protein fibronectin. We therefore speculate that TG2 mediates the enhanced interaction of astrocytes with fibronectin in the extracellular matrix of MS lesions, thereby contributing to astrocyte adhesion and migration, and thus in tissue remodeling and possibly glial scarring.


Assuntos
Astrócitos/metabolismo , Adesão Celular/fisiologia , Movimento Celular/fisiologia , Esclerose Múltipla/enzimologia , Bainha de Mielina/metabolismo , Transglutaminases/metabolismo , Idoso , Idoso de 80 Anos ou mais , Análise de Variância , Astrócitos/patologia , Western Blotting , Linhagem Celular Tumoral , Células Cultivadas , Matriz Extracelular/metabolismo , Matriz Extracelular/patologia , Feminino , Fibronectinas/metabolismo , Proteínas de Ligação ao GTP , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Esclerose Múltipla/patologia , Bainha de Mielina/patologia , Proteína 2 Glutamina gama-Glutamiltransferase , RNA Interferente Pequeno
15.
J Neuropathol Exp Neurol ; 69(7): 694-703, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20535035

RESUMO

We present the first comparative analysis of serum immunoglobulin G reactivity profiles against the full spectrum of human myelin-associated proteins in multiple sclerosis patients and healthy control subjects. In both groups, serum antibodies display a consistent and prominent reaction to alphaB-crystallin (CRYAB) versus other myelin proteins. As an apparently major target for the adaptive immune system in humans, CRYAB selectively accumulates in oligodendrocytes, but not in astrocytes, or axons in so-called preactive multiple sclerosis lesions. These are clusters of activated HLA-DR-expressing microglia in myelinated normal-appearing white matter with no obvious leukocyte infiltration. They are found in most multiple sclerosis patients at all stages of disease. In these lesion areas, CRYAB in oligodendrocytes may come directly in contact with activated HLA-DR+ microglia. We demonstrate that CRYAB activates innate responses by microglia by stimulating the secretion of leukocyte-recruiting factors, including tumor necrosis factor, interleukin 17, CCL5, and CCL1, and immune-regulatory cytokines such as interleukin 10, transforming growth factor-beta, and interleukin 13. Together, these data suggest that CRYAB accumulation in preactive lesions may be part of a reversible reparative local response that involves both oligodendrocytes and microglia. At the same time, however, accumulated CRYAB may represent a major target for adaptive immune responses that could contribute to progression of preactive lesions to a stage of demyelination.


Assuntos
Imunidade Adaptativa/fisiologia , Esclerose Múltipla/imunologia , Esclerose Múltipla/fisiopatologia , alfa-Cristalinas/metabolismo , Adulto , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Citocinas/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , Antígenos HLA-DR/metabolismo , Humanos , Espectrometria de Massas/métodos , Microglia/metabolismo , Pessoa de Meia-Idade , Esclerose Múltipla/patologia , Proteína Proteolipídica de Mielina/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas de Neurofilamentos/metabolismo , Fator de Transcrição 2 de Oligodendrócitos , Oligodendroglia/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA