Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 12: 720733, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34484228

RESUMO

One hallmark of Guillain-Barre syndrome (GBS), a prototypic autoimmune peripheral neuropathy (APN) is infiltration of leukocytes (macrophages and T cells) into peripheral nerves, where chemokines and their receptors play major roles. In this study, we aimed to understand the potential contribution of chemokine receptors CCR2 and CX3CR1 in APN by using a well-established mouse model, B7.2 transgenic (L31) mice, which possesses a predisposed inflammatory background. We crossbred respectively CCR2KO and CX3CR1KO mice with L31 mice. The disease was initiated by partial ligation on one of the sciatic nerves. APN pathology and neurological function were evaluated on the other non-ligated sciatic nerve/limb. Our results revealed that L31/CX3CR1KO but not L31/CCR2KO mice were resistant to APN. CX3CR1 is needed for maintaining circulating monocyte and CD8+ T cell survival. While migration of a significant number of activated CD8+ T cells to peripheral nerves is essential in autoimmune response in nerve, recruitment of monocytes into PNS seems optional. Disease onset is independent of CCR2 mediated blood-derived macrophage recruitment, which can be replaced by compensatory proliferation of resident macrophages in peripheral nerve. CX3CR1 could also contribute to APN via its critical involvement in maintaining nerve macrophage phagocytic ability. We conclude that blockade of CX3CR1 signaling may represent an interesting anti-inflammatory strategy to improve therapeutic management for GBS patients.


Assuntos
Autoimunidade/genética , Receptor 1 de Quimiocina CX3C/genética , Expressão Gênica , Neurite Autoimune Experimental/etiologia , Doenças do Sistema Nervoso Periférico/etiologia , Receptores CCR2/genética , Animais , Biomarcadores , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Receptor 1 de Quimiocina CX3C/metabolismo , Modelos Animais de Doenças , Suscetibilidade a Doenças/imunologia , Imunofenotipagem , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Modelos Biológicos , Neurite Autoimune Experimental/metabolismo , Neurite Autoimune Experimental/patologia , Doenças do Sistema Nervoso Periférico/metabolismo , Receptores CCR2/metabolismo , Nervo Isquiático/imunologia , Nervo Isquiático/metabolismo , Nervo Isquiático/patologia
2.
J Biol Regul Homeost Agents ; 33(5): 1369-1376, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31637897

RESUMO

The objective of this paper was to study the effects of PYR-ARG-PRO-ARG-LEU-SER-HIS-YSGLY-PRO-MET-PRO-PHE-OH (APELIN-13) on the expression of inflammatory factors interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), and interferon-γ (IFN-γ) in rats with experimental autoimmune neuritis (EAN). A total of 30 rats were divided into a control group, an EAN group, and an APELIN-13 group. Enzyme-linked immunosorbent assay (ELISA) was used to detect the levels of IL-6, TNF-α, and IFN-γ in rat plasma. Real-time quantitative Polymerase Chain Reaction (PCR) and Western blot were used to detect the protein and mRNA expression of IL-6, TNF-α, and IFN-γ in rat lymph nodes. In the EAN group, the infiltration of various types of inflammatory cells and focal demyelination were observed near the nerve fascicles of sciatic nerves. Compared with the EAN group, the infiltration of inflammatory cells and demyelination in the APELIN-13 group decreased significantly. The levels of plasma IL-6, TNF-α, and IFN-γ in the EAN group were significantly higher than those in the control group (P < 0.05) but significantly lower than those in the APELIN-13 group (P < 0.05). Compared with the control group, the mRNA and protein expression of IL-6, TNF-α, and IFN-γ increased significantly (P < 0.05) in the EAN group but decreased significantly in the APELIN-13 group (P < 0.05). In conclusion, APELIN-13 exerted a protective effect against EAN in rats.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Interferon gama/metabolismo , Interleucina-6/metabolismo , Neurite Autoimune Experimental/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Ratos
3.
Nat Commun ; 10(1): 1467, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30931926

RESUMO

In contrast to acute peripheral nerve injury, the molecular response of Schwann cells in chronic neuropathies remains poorly understood. Onion bulb structures are a pathological hallmark of demyelinating neuropathies, but the nature of these formations is unknown. Here, we show that Schwann cells induce the expression of Neuregulin-1 type I (NRG1-I), a paracrine growth factor, in various chronic demyelinating diseases. Genetic disruption of Schwann cell-derived NRG1 signalling in a mouse model of Charcot-Marie-Tooth Disease 1A (CMT1A), suppresses hypermyelination and the formation of onion bulbs. Transgenic overexpression of NRG1-I in Schwann cells on a wildtype background is sufficient to mediate an interaction between Schwann cells via an ErbB2 receptor-MEK/ERK signaling axis, which causes onion bulb formations and results in a peripheral neuropathy reminiscent of CMT1A. We suggest that diseased Schwann cells mount a regeneration program that is beneficial in acute nerve injury, but that overstimulation of Schwann cells in chronic neuropathies is detrimental.


Assuntos
Doenças Desmielinizantes/genética , Neuregulina-1/genética , Comunicação Parácrina , Células de Schwann/metabolismo , Nervo Sural/metabolismo , Animais , Animais Geneticamente Modificados , Doença de Charcot-Marie-Tooth/genética , Doença de Charcot-Marie-Tooth/metabolismo , Doença de Charcot-Marie-Tooth/patologia , Doenças Desmielinizantes/metabolismo , Doenças Desmielinizantes/patologia , Diabetes Mellitus Tipo 1/complicações , Neuropatias Diabéticas/etiologia , Neuropatias Diabéticas/genética , Neuropatias Diabéticas/metabolismo , Neuropatias Diabéticas/patologia , Humanos , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica , Atividade Motora , Proteínas da Mielina/genética , Neuregulina-1/metabolismo , Neurite Autoimune Experimental/genética , Neurite Autoimune Experimental/metabolismo , Neurite Autoimune Experimental/patologia , Neuroglia/metabolismo , Ratos , Receptor ErbB-2/metabolismo , Células de Schwann/ultraestrutura , Nervo Isquiático/lesões , Transdução de Sinais , Nervo Sural/ultraestrutura , Nervo Tibial
4.
Mol Med Rep ; 15(4): 1565-1570, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28260057

RESUMO

Experimental autoimmune neuritis (EAN) is a cluster of differentiation 4+ T helper 1 cell-mediated inflammatory demyelinating disease of the peripheral nervous system and serves as a useful animal model for Guillain­Barré syndrome. Triggering receptor expressed on myeloid cells­1 (TREM­1) is an important receptor involved in sepsis and the innate inflammatory response. Linear plasmid 17 (LP 17) peptide is a competitive antagonist of TREM­1. To investigate the role of TREM­1 in EAN, 64 male Lewis rats were randomly divided into four groups: Normal saline, complete Freund's adjuvant, EAN and LP 17. The present study assessed the mRNA expression levels of TREM­1, tumor necrosis factor­α and interleukin­1ß in sciatic nerves and peripheral blood mononuclear cells. The results demonstrated that inhibiting TREM-1 by administering LP 17 ameliorated symptoms and reduced inflammation in EAN rats. The present study concluded that TREM­1 may be involved in the pathogenesis of EAN, and that inhibition of TREM-1 may ameliorate EAN.


Assuntos
Neurite Autoimune Experimental/metabolismo , Neurite Autoimune Experimental/patologia , Receptores Imunológicos/antagonistas & inibidores , Animais , Regulação da Expressão Gênica , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Masculino , Neurite Autoimune Experimental/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos Endogâmicos Lew , Receptores Imunológicos/metabolismo , Nervo Isquiático/patologia , Coloração e Rotulagem , Receptor Gatilho 1 Expresso em Células Mieloides , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
5.
Am J Pathol ; 187(1): 42-54, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27842213

RESUMO

The functional relevance of the innate immune system has not yet been dissected in P0106-125-induced murine experimental autoimmune neuritis. Therefore, the role of Toll-like receptor (TLR) 2, TLR4, myeloid differentiation response gene 88, and Toll-IL-1 receptor domain-containing adaptor-inducing interferon-γ (TRIF), factors critically involved in the TLR signaling pathway, was studied in experimental autoimmune neuritis. In the absence of TLR2, TLR4, myeloid differentiation response gene 88, or TRIF, the clinical course was significantly attenuated compared to wild-type mice. This could be attributed to impaired NF-κB activation, as shown by the absence of nuclear translocation of RelA with a decreased expression of IL-6, IL-12p40, and IL-17A. Remarkably, P0106-125-immunized TLR20/0 mice exhibited a delayed recovery as compared to TLR40/0 mice, which was because of an impaired T helper cell 2 polarization. Immunized TLR20/0 mice were unable to induce OX40 and OX40L by matrix metalloproteinase-2 on splenic dendritic cells. Subsequently, M2 polarization was impaired and macrophages were unable to sufficiently induce T regulatory cells (Tregs). Thus, in the recovery phase, Tregs were significantly increased in TLR40/0 mice as compared to wild-type mice, whereas Tregs in immunized TLR20/0 mice were only slightly increased. Our data highlight the relevance of innate immunity and, especially, the tight interaction between the innate and the adaptive immune system, which should be considered for therapeutic approaches of autoimmune diseases.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Neurite Autoimune Experimental/metabolismo , Neurite Autoimune Experimental/patologia , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/metabolismo , Animais , Axônios/patologia , Linfócitos T CD4-Positivos/imunologia , Complemento C1q/imunologia , Progressão da Doença , Suscetibilidade a Doenças , Imunoglobulina G/sangue , Imunoglobulina M/sangue , Interferon gama/genética , Interferon gama/metabolismo , Contagem de Linfócitos , Ativação de Macrófagos , Metaloproteinase 2 da Matriz/metabolismo , Camundongos Endogâmicos C57BL , Músculo Esquelético/inervação , Músculo Esquelético/patologia , Proteína P0 da Mielina , NF-kappa B/metabolismo , Neurite Autoimune Experimental/sangue , Neurite Autoimune Experimental/imunologia , Ligante OX40/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores OX40/metabolismo , Nervo Isquiático/metabolismo , Nervo Isquiático/patologia , Transdução de Sinais , Baço/metabolismo
6.
J Neurosci ; 36(37): 9590-603, 2016 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-27629711

RESUMO

UNLABELLED: Experimental autoimmune neuritis (EAN) is the animal model of human acute inflammatory demyelinating polyradiculoneuropathies (AIDP), an auto-immune inflammatory demyelination disease of the peripheral nervous system (PNS) and the world's leading cause of acute autoimmune neuromuscular paralysis. EAN and AIDP are characterized by self-limitation with spontaneous recovery; however, endogenous pathways that regulate inflammation resolution in EAN and AIDP remain elusive. A pathway of endogenous mediators, especially resolvins and clearance of apoptotic cells, may be involved. Here, we determined that resolvin D1 (RvD1), its synthetic enzyme, and its receptor were greatly increased in PNS during the recovery stage of EAN. Both endogenous and exogenous RvD1 increased regulatory T (Treg) cell and anti-inflammatory macrophage counts in PNS, enhanced inflammation resolution, and promoted disease recovery in EAN rats. Moreover, RvD1 upregulated the transforming growth factor-ß (TGF-ß) level and pharmacologic inhibition of TGF-ß signaling suppressed RvD1-induced Treg cell counts, but not anti-inflammatory macrophage counts, and RvD1-improved inflammation resolution and disease recovery in EAN rats. Mechanistically, the RvD1-enhanced macrophage phagocytosis of apoptotic T cells leading to reduced apoptotic T-cell accumulation in PNS induced TGF-ß production and caused Treg cells to promote inflammation resolution and disease recovery in EAN. Therefore, these data highlight the crucial role of RvD1 as an important pro-resolving molecule in EAN and suggest its potential as a therapeutic target in human neuropathies. SIGNIFICANCE STATEMENT: Experimental autoimmune neuritis (EAN) is the animal model of human acute inflammatory demyelinating polyradiculoneuropathies, an auto-immune inflammatory demyelination disease of the peripheral nervous system (PNS) and the world's leading cause of acute autoimmune neuromuscular paralysis. Here, we demonstrated that resolvin D1 (RvD1) promoted macrophage phagocytosis of apoptotic T cells in PNS, thereby upregulating transforming growth factor-ß by macrophages, increased local Treg cell counts, and finally promoted inflammation resolution and disease recovery in EAN. These data highlight the crucial role of RvD1 as an important pro-resolving molecule in EAN and suggest that it has potential as a therapeutic target in human neuritis.


Assuntos
Anti-Inflamatórios/uso terapêutico , Ácidos Docosa-Hexaenoicos/uso terapêutico , Regulação da Expressão Gênica/efeitos dos fármacos , Neurite Autoimune Experimental/tratamento farmacológico , Fator de Crescimento Transformador beta/metabolismo , Animais , Anti-Inflamatórios/farmacologia , Apoptose/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Ácidos Docosa-Hexaenoicos/metabolismo , Ectodisplasinas/metabolismo , Inibidores Enzimáticos/uso terapêutico , Fatores de Transcrição Forkhead/metabolismo , Macrófagos/efeitos dos fármacos , Masculino , Neurite Autoimune Experimental/metabolismo , Neurite Autoimune Experimental/patologia , Fagocitose/efeitos dos fármacos , Pteridinas/uso terapêutico , Ratos , Ratos Endogâmicos Lew , Receptores de Lipoxinas/antagonistas & inibidores , Receptores de Lipoxinas/metabolismo , Nervo Isquiático/patologia , Nervo Isquiático/ultraestrutura , Linfócitos T Reguladores/metabolismo , Linfócitos T Reguladores/patologia , Linfócitos T Reguladores/ultraestrutura
7.
J Neuroinflammation ; 13(1): 97, 2016 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-27142843

RESUMO

BACKGROUND: Guillain-Barré syndrome (GBS) is an acute, post-infectious, immune-mediated, demyelinating disease of peripheral nerves and nerve roots. Dimethyl fumarate (DMF), a fumaric acid ester, exhibits various biological activities, including multiple immunomodulatory and neuroprotective effects. However, the potential mechanism underlying the effect of DMF in GBS animal model experimental autoimmune neuritis (EAN) is unclear. METHODS: Using EAN, an established GBS model, we investigated the effect of DMF by assessing clinical score, histological staining and electrophysiological studies. Then, we further explored the potential mechanism by Western blot analysis, flow cytometry, fluorescence immunohistochemistry, PCR, and ELISA analysis. The Mann-Whitney U test was used to compare differences between control group and treatment groups where appropriate. RESULTS: DMF treatment reduced the neurological deficits by ameliorating inflammatory cell infiltration and demyelination of sciatic nerves. In addition, DMF treatment decreased the level of pro-inflammatory M1 macrophages while increasing the number of anti-inflammatory M2 macrophages in the spleens and sciatic nerves of EAN rats. In RAW 264.7, a shift in macrophage polarization from M1 to M2 phenotype was demonstrated to be depended on DMF application. In sciatic nerves, DMF treatment elevated the level of the antioxidant transcription factor nuclear factor erythroid-derived 2-related factor 2 (Nrf2) and its target gene hemoxygenase-1 (HO-1) which could facilitate macrophage polarization toward M2 type. Moreover, DMF improved the inflammatory milieu in spleens of EAN rats, characterized by downregulation of messenger RNA (mRNA) of IFN-γ, TNF-α, IL-6, and IL-17 and upregulation of mRNA level of IL-4 and IL-10. CONCLUSIONS: Taken together, our data demonstrate that DMF can effectively suppress EAN, and the mechanism involves altering the balance of M1/M2 macrophages and attenuating inflammation.


Assuntos
Fumarato de Dimetilo/farmacologia , Macrófagos/imunologia , Neurite Autoimune Experimental/imunologia , Neurite Autoimune Experimental/patologia , Fármacos Neuroprotetores/farmacologia , Animais , Western Blotting , Eletrofisiologia , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Imunofluorescência , Heme Oxigenase-1/metabolismo , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Masculino , Fator 2 Relacionado a NF-E2/metabolismo , Neurite Autoimune Experimental/metabolismo , Reação em Cadeia da Polimerase , Ratos , Ratos Endogâmicos Lew
8.
Exp Neurol ; 280: 106-14, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27063582

RESUMO

Guillain-Barre' syndrome (GBS) is an acute, postinfectious, immune-mediated, demyelinating disease of peripheral nerves and nerve roots. As a classical animal model of GBS, experimental autoimmune neuritis (EAN) has become well-accepted. Additionally, the potent immune modulation exerted by mammalian target of rapamycin (mTOR) inhibitors has been used to treat cancers and showed beneficial effects. Here we demonstrate that the mTOR inhibitor RAD001 (everolimus) protected rats from the symptoms of EAN, as shown by decreased paralysis, diminished inflammatory cell infiltration, reductions in demyelination of peripheral nerves and improved nerve conduction. Furthermore, RAD001 shifted macrophage polarization toward the protective M2 phenotype and modified the inflammatory milieu by downregulating the production of pro-inflammatory cytokines including IFN-γ and IL-17as well as upregulating the release of anti-inflammatory cytokines such as IL-4 and TGF-ß. Amounts of the mTOR downstream targets p-P70S6K and p-4E-BP1 in sciatic nerves decreased, whereas the level of its upstream protein p-Akt was elevated. This demonstrated that RAD001 inhibited the mTOR pathway and encouraged the expression of p-Akt, which led to M2 macrophage polarization, thus improved the outcome of EAN in rats. Consequently, RAD001 exhibits strong potential as a therapeutic strategy for ameliorating peripheral poly-neuropathy.


Assuntos
Everolimo/uso terapêutico , Imunossupressores/uso terapêutico , Neurite Autoimune Experimental/tratamento farmacológico , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo , Animais , Polaridade Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Eletromiografia , Everolimo/farmacologia , Adjuvante de Freund/toxicidade , Imunossupressores/farmacologia , Linfócitos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Masculino , Condução Nervosa/efeitos dos fármacos , Neurite Autoimune Experimental/induzido quimicamente , Neurite Autoimune Experimental/metabolismo , Ratos , Ratos Endogâmicos Lew , Fatores de Tempo
9.
J Neuroimmunol ; 293: 129-136, 2016 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-27049573

RESUMO

Accumulated evidence demonstrated that Adenosine A2A receptor (A2AR) is involved in the inflammatory diseases. In the present study, we showed that a selective A2AR agonist, CGS21680, exacerbated experimental autoimmune neuritis in Lewis rats induced with bovine peripheral myelin. The exacerbation was accompanied with reduced CD4(+)Foxp3(+) T cells, increased CD4(+)CXCR5(+) T cells, B cells, dendritic cells and antigen-specific autoantibodies, which is possibly due to the inhibition of IL-2 induced by CGS21680. Combined with previous studies, our data indicate that the effects of A2AR stimulation in vivo are variable in different diseases. Caution should be taken in the use of A2AR agonists.


Assuntos
Citocinas/metabolismo , Imunidade Humoral/fisiologia , Bainha de Mielina/imunologia , Neurite Autoimune Experimental/imunologia , Neurite Autoimune Experimental/metabolismo , Receptores A2 de Adenosina/metabolismo , Adenosina/efeitos adversos , Adenosina/análogos & derivados , Adenosina/farmacologia , Agonistas do Receptor A2 de Adenosina/farmacologia , Animais , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Bovinos , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Modelos Animais de Doenças , Feminino , Imunidade Humoral/efeitos dos fármacos , Imunoglobulina G/sangue , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/imunologia , Linfonodos/patologia , Linfócitos/efeitos dos fármacos , Linfócitos/metabolismo , Doenças do Sistema Nervoso/induzido quimicamente , Neurite Autoimune Experimental/etiologia , Neurite Autoimune Experimental/patologia , Fenetilaminas/efeitos adversos , Fenetilaminas/farmacologia , Ratos , Ratos Endogâmicos Lew , Nervo Isquiático/patologia
10.
Neurol Sci ; 37(2): 199-203, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26410085

RESUMO

Experimental autoimmune neuritis (EAN) is a well-known animal model of human demyelinating polyneuropathies. Macrophages are the major immune cells in peripheral nerves and may exert tissue-damage or tissue-protective activity during EAN. While considered to define a subpopulation of T lymphocytes, CD8 expression has been found on certain macrophages that show cytotoxic effects. Here we have studied the spatiotemporal accumulation of CD8(+) cells in sciatic nerves of EAN rats. A robust accumulation of CD8(+) cells was observed in the sciatic nerves of EAN rats, which was positively correlated with the severity of neurological signs in EAN. Moreover, double-labelling experiments showed that the major cellular sources of CD8 were reactive macrophages. Therefore, our data here suggest a pathological role of CD8(+) macrophages in EAN, which makes CD8(+) macrophage a potential therapeutic target for EAN.


Assuntos
Antígenos CD8/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Macrófagos/metabolismo , Neurite Autoimune Experimental/imunologia , Nervo Isquiático/imunologia , Animais , Linfócitos T CD8-Positivos/patologia , Macrófagos/patologia , Masculino , Neurite Autoimune Experimental/metabolismo , Neurite Autoimune Experimental/patologia , Ratos , Ratos Endogâmicos Lew , Nervo Isquiático/metabolismo , Nervo Isquiático/patologia
11.
Int J Clin Exp Pathol ; 8(7): 7849-58, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26339349

RESUMO

To explore the relationship of peripheral nerve ultrastructure and its associated protein expression in experimental autoimmune neuritis (EAN). EAN was established in Lewis rats using an emulsified mixture of P0 peptide 180-199, Mycobacterium tuberculosis, and incomplete Freund's adjuvant. Rats immunized with saline solution were used as a control group. Sciatic nerve ultrastructure and immunofluorescence histopathology were measured at the neuromuscular severity peak on day 18 post-induction. Cell-specific protein markers were used for immunofluorescence histopathology staining to characterize sciatic nerve cells: CD3 (T cell), Iba-1 (microglia), S100 (myelin), and neurofilament 200 (axon). The results showed that swelling of the myelin lamellae, vesicular disorganization, separation of the myelin lamellae, and an attenuation or disappearance of the axon were observed by transmission electron microscopy in the EAN group. CD3 and Iba-1 increased significantly in the structures characterized by separation or swelling of the myelin lamellae, and increased slightly in the structures characterized by vesicular of the myelin lamellae, S100 decreased in the structures characterized by vesicular disorganization or separation of the myelin lamellae. And neurofilament 200 decreased in the structures characterized by separation of the myelin lamellae. Furthermore, we found that Iba1 were positive in the myelin sheath, and overlapped with S100, which significantly indicated that Schwann cells played as macrophage-like cells during the disease progression of ENA. Our findings may be a significant supplement for the knowledge of EAN model, and may offer a novel sight on the treatment of Guillain-Barré syndrome.


Assuntos
Neurite Autoimune Experimental/patologia , Animais , Animais Recém-Nascidos , Adjuvante de Freund , Humanos , Imunização , Lipídeos , Macrófagos/metabolismo , Masculino , Bainha de Mielina/metabolismo , Neurite Autoimune Experimental/metabolismo , Ratos , Nervo Isquiático/metabolismo , Linfócitos T/imunologia
12.
Nat Commun ; 6: 7096, 2015 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-25963922

RESUMO

Although different autoimmune diseases show discrete clinical features, there are common molecular pathways intimately involved. Here we show that miR-125a is downregulated in peripheral CD4(+) T cells of human autoimmune diseases including systemic lupus erythematosus and Crohn's disease, and relevant autoimmune mouse models. miR-125a stabilizes both the commitment and immunoregulatory capacity of Treg cells. In miR-125a-deficient mice, the balance appears to shift from immune suppression to inflammation, and results in more severe pathogenesis of colitis and experimental autoimmune encephalomyelitis (EAE). The genome-wide target analysis reveals that miR-125a suppresses several effector T-cell factors including Stat3, Ifng and Il13. Using a chemically synthesized miR-125a analogue, we show potential to re-programme the immune homeostasis in EAE models. These findings point to miR-125a as a critical factor that controls autoimmune diseases by stabilizing Treg-mediated immune homeostasis.


Assuntos
Imunidade Celular/fisiologia , MicroRNAs/metabolismo , Linfócitos T Reguladores/fisiologia , Animais , Estudos de Casos e Controles , Colite/metabolismo , Colite/patologia , Doença de Crohn , Regulação para Baixo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Homeostase , Humanos , Lúpus Eritematoso Sistêmico , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Neurite Autoimune Experimental/metabolismo , Neurite Autoimune Experimental/patologia
13.
Am J Pathol ; 184(10): 2627-40, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25108223

RESUMO

The role of the type 2 helper T cell (Th2)-polarizing cytokines IL-4 and IL-10 has not yet been studied in P0106-125-induced murine experimental autoimmune neuritis (EAN). We, therefore, addressed the functional relevance of these cytokines and signaling via the IL-4-associated transcription factor STAT6. The clinical course of P0106-125-induced EAN in mice deficient for IL-10(0/0), IL-4(0/0), or STAT6(0/0) was significantly aggravated compared with that of wild-type control mice. In addition, treatment of P0106-125-immunized C57BL/6 mice at the onset of clinical symptoms with a monoclonal IL-10 neutralizing antibody aggravated symptoms and prolonged disease to a similar degree as in IL-10(0/0) mice. This exacerbated course was attributed to a more prominent Th1 immune response associated with a persistent M1 milieu in the sciatic nerve and in the regional and systemic lymphatic system. These data suggest a Th2-polarized milieu being required to prevent axonal damage of the sciatic nerve and to terminate the P0106-125-specific immune response in EAN. Beyond the already known role of macrophages as pathogenic effector cells in EAN, these data suggest that M2-differentiated macrophages do not damage and may even protect neural tissues in EAN. Thus, these data highlight the pathogenetic relevance of the macrophage polarization status in EAN. Therapeutic modulation of immune responses from an M1 toward an M2 milieu may be a promising novel strategy in peripheral nervous system neuritis.


Assuntos
Interleucina-10/metabolismo , Interleucina-4/metabolismo , Neurite Autoimune Experimental/imunologia , Fator de Transcrição STAT6/metabolismo , Células Th2/imunologia , Animais , Modelos Animais de Doenças , Humanos , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteína P0 da Mielina/metabolismo , Neurite Autoimune Experimental/metabolismo , Sistema Nervoso Periférico/metabolismo , Nervo Isquiático/patologia , Organismos Livres de Patógenos Específicos , Baço/patologia , Células Th2/metabolismo
14.
Scand J Immunol ; 80(3): 180-90, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24910360

RESUMO

Experimental autoimmune neuritis (EAN), an animal model of human Guillain-Barré syndrome, has long been considered as a T helper (Th) 1 cell-mediated autoimmune disorder. However, deficiency of IFN-γ, a signature Th1 cytokine, aggravated EAN, with features of elevated production of IL-17A, despite an alleviated systemic Th1 immune response. We hypothesized that Th17 cells and their cytokines might play a pathogenic role in EAN. To further clarify the roles of these Th and regulatory T cell (Treg) cytokines in the pathogenesis of EAN and their interrelationship, we investigated the expression of Th1/Th2/Th17/Treg cytokines in EAN in this study. We found that the levels of Th17 cells and IL-17A in cauda equina (CE)-infiltrating cells and splenic mononuclear cells (MNCs) as well as in serum paralleled the disease evolution, which increased progressively during the initiation stage and reached higher value at the peak of EAN. The same pattern was also noticed for the expression of IL-22. The diverse expression profiles of FoxP3, IL-17 receptors A and C were seen in CE-infiltrating cells and splenic MNCs in EAN. These findings indicate a major pro-inflammatory role of Th17 cells and IL-17A in the pathogenesis of EAN. Therapeutic interventions may be focused upon inhibiting Th17 cells and their cytokines in the early phase of EAN, so as to delay and suppress clinical signs of the disease, which has relevance for future studies on pathogenesis and treatment of GBS in humans.


Assuntos
Citocinas/imunologia , Neurite Autoimune Experimental/imunologia , Linfócitos T Reguladores/imunologia , Células Th17/imunologia , Animais , Cauda Equina/imunologia , Cauda Equina/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Citometria de Fluxo , Fatores de Transcrição Forkhead/imunologia , Fatores de Transcrição Forkhead/metabolismo , Síndrome de Guillain-Barré/imunologia , Síndrome de Guillain-Barré/metabolismo , Síndrome de Guillain-Barré/patologia , Humanos , Interferon gama/imunologia , Interferon gama/metabolismo , Interleucina-10/imunologia , Interleucina-10/metabolismo , Interleucina-12/imunologia , Interleucina-12/metabolismo , Interleucina-17/sangue , Interleucina-17/imunologia , Interleucina-17/metabolismo , Interleucina-6/imunologia , Interleucina-6/metabolismo , Interleucinas/sangue , Interleucinas/imunologia , Interleucinas/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Neurite Autoimune Experimental/metabolismo , Neurite Autoimune Experimental/patologia , Receptores de Interleucina-17/imunologia , Receptores de Interleucina-17/metabolismo , Índice de Gravidade de Doença , Baço/imunologia , Baço/metabolismo , Linfócitos T Reguladores/metabolismo , Células Th1/imunologia , Células Th1/metabolismo , Células Th17/metabolismo , Células Th2/imunologia , Células Th2/metabolismo , Fatores de Tempo , Interleucina 22
15.
J Neuropathol Exp Neurol ; 73(5): 454-66, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24709684

RESUMO

Myelin protein 0 peptide 106-125-induced murine experimental autoimmune neuritis (EAN) is a CD4-positive T cell-mediated monophasic axonal inflammatory neuropathy; interferon-γ is the key proinflammatory mediator. Experimental autoimmune neuritis is well suited for elucidating pathogenetic mechanisms underlying human acute axonal Guillain-Barré syndrome. Here, the functional role of the costimulatory molecule CD40 was defined by characterization of EAN in CD40-deficient mice. In contrast to immunized C57BL/6 mice, CD40-deficient mice were resistant to EAN owing to impaired priming of CD4-positive T-effector cells. To determine whether CD40 is a suitable candidate for the treatment of EAN, we administered monoclonal anti-CD40 antibody either before immunization or upon onset of neurologic signs. Prophylactic anti-CD40 treatment completely abolished CD4-positive T-cell priming. Therapeutic application of anti-CD40 prevented full activation of CD4-positive T cells that were in the process of priming and suppressed production of interferon-γ in peripheral lymph nodes, spleen, and serum, and of interleukin-6, interleukin-12p40, intercellular adhesion molecule-1, and vascular cell adhesion molecule-1, which are associated with activation of the nuclear factor-κB signaling pathway. This resulted in enhanced recovery by early generation of CD25-positive, Foxp3-positive, CD4-positive regulatory T cells. Thus, these experiments highlight the crucial role of CD40 as an important costimulatory molecule in EAN and suggest that it has potential as a therapeutic target in human neuritis.


Assuntos
Antígenos CD40/deficiência , Antígenos CD40/fisiologia , Proteína P0 da Mielina/toxicidade , Neurite Autoimune Experimental/imunologia , Peptídeos/toxicidade , Sequência de Aminoácidos , Animais , Axônios/imunologia , Axônios/patologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/patologia , Antígenos CD40/imunologia , Mediadores da Inflamação/toxicidade , Ativação Linfocitária/imunologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dados de Sequência Molecular , Neurite Autoimune Experimental/genética , Neurite Autoimune Experimental/metabolismo
16.
Cell Death Dis ; 4: e946, 2013 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-24309933

RESUMO

Experimental autoimmune neuritis (EAN) is an animal model of Guillain-Barré syndrome, an inflammatory demyelination disease of the peripheral nervous system. Although this disease has been extensively studied on peripheral nerves, the pathology of the central nervous system has not been fully understood. Previous studies demonstrate that expression of keratan sulfate (KS), the sugar chain of proteoglycan, is associated with activated microglia/macrophages accumulated after neuronal injuries. Unexpectedly, we found here that KS is rather diminished in rat EAN. KS was restrictively expressed in microglia in the spinal cord of normal rats. KS was positive in 50% microglia in the ventral horn and 20% in the dorsal horn. In EAN, microglia increased in number and expressed the activation marker CD68, but KS expression was abolished. Concomitantly, pro-inflammatory cytokines, i.e., interferon (IFN)-γ, interleukin (IL)-1ß, and tumor necrosis factor (TNF)-α, were increased in the spinal cord of EAN rats, whereas anti-inflammatory cytokines, such as IL-4 and IL-10, were decreased. In addition, silencing of KSGal6ST attenuated KS expression on the primary cultured microglia and upregulated expression of some activation markers (TNF-α, IL-1ß, and iNOS) under the stimulation with lipopolysaccharide and IFN-γ. This study demonstrates for the first time a close association of EAN and disappearance of KS on microglia. KS expression could be a useful marker to evaluate the status of polyneuropathy.


Assuntos
Sulfato de Queratano/metabolismo , Microglia/metabolismo , Neurite Autoimune Experimental/metabolismo , Medula Espinal/metabolismo , Animais , Western Blotting , Citometria de Fluxo , Imuno-Histoquímica , Masculino , Neurite Autoimune Experimental/genética , Ratos , Reação em Cadeia da Polimerase em Tempo Real
17.
Biochim Biophys Acta ; 1832(8): 1260-70, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23603807

RESUMO

Experimental autoimmune neuritis (EAN), an autoantigen-specific T-cell-mediated disease model for human demyelinating inflammatory disease of the peripheral nervous system, is characterized by self-limitation. Here we investigated the regulation and contribution of erythropoietin (EPO) in EAN self-limitation. In EAN sciatic nerves, hypoxia, and protein and mRNA levels of hypoxia-inducible factor 1α (HIF-1α), HIF-2α, EPO and EPO receptor (EPOR) were induced in parallel at disease peak phase but reduced at recovery periods. Further, the deactivation of HIF reduced EAN-induced EPO/EPOR upregulation in EAN, suggesting the central contribution of HIF to EPO/EPOR induction. The deactivation of EPOR signalling exacerbated EAN progression, implying that endogenous EPO contributed to EAN recovery. Exogenous EPO treatment greatly improved EAN recovery. In addition, EPO was shown to promote Schwann cell survival and myelin production. In EAN, EPO treatment inhibited lymphocyte proliferation and altered helper T cell differentiation by inducing increase of Foxp3(+)/CD4(+) regulatory T cells and decrease of IFN-γ(+)/CD4(+) Th1 cells. Furthermore, EPO inhibited inflammatory macrophage activation and promoted its phagocytic activity. In summary, our data demonstrated that EPO was induced in EAN by HIF and contributed to EAN recovery, and endogenous and exogenous EPO could effectively suppress EAN by attenuating inflammation and exerting direct cell protection, indicating that EPO contributes to the self-recovery of EAN and could be a potent candidate for treatment of autoimmune neuropathies.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/imunologia , Eritropoetina/imunologia , Eritropoetina/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/imunologia , Neurite Autoimune Experimental/imunologia , Neurite Autoimune Experimental/metabolismo , Animais , Diferenciação Celular/imunologia , Proliferação de Células , Células Cultivadas , Eritropoetina/farmacologia , Humanos , Inflamação/imunologia , Inflamação/metabolismo , Células Jurkat , Ativação Linfocitária/imunologia , Linfócitos/imunologia , Linfócitos/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Camundongos , Fagocitose/imunologia , Ratos , Ratos Endogâmicos Lew , Receptores da Eritropoetina/imunologia , Receptores da Eritropoetina/metabolismo , Células de Schwann/imunologia , Células de Schwann/metabolismo , Nervo Isquiático/imunologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Células Th1/imunologia , Células Th1/metabolismo
18.
PLoS One ; 7(5): e38157, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22666471

RESUMO

The role of tumor necrosis factor (TNF)-α and its receptors in neuroautoimmune and neuroinflammatory diseases has been controversial. On the basis of our previous studies, we hereby aimed to further clarify TNF-α's mechanism of action and to explore the potential role of TNF-α receptor (TNFR)1 as a therapeutic target in experimental autoimmune neuritis (EAN). EAN was induced by immunization with P0 peptide 180-199 in TNF-α knockout (KO) mice and anti-TNFR1 antibodies were used to treat EAN. Particularly, the effects of TNF-α deficiency and TNFR1 blockade on macrophage functions were investigated. The onset of EAN in TNF-α KO mice was markedly later than that in wild type (WT) mice. From day 14 post immunization, the clinical signs of TNF-α KO mice were significantly milder than those of their WT counterparts. Further, we showed that the clinical severity of WT mice treated with anti-TNFR1 antibodies was less severe than that of the control WT mice receiving PBS. Nevertheless, no difference with regard to the clinical signs of EAN or inflammatory infiltration in cauda equina was seen between TNF-α KO and WT mice with EAN after blockade of TNFR1. Although TNF-α deficiency did not alter the proliferation of lymphocytes in response to either antigenic or mitogenic stimuli, it down-regulated the production of interleukin (IL)-12 and nitric oxide (NO), and enhanced the production of IL-10 in macrophages. Increased ratio of regulatory T cells (Tregs) and reduced production of interferon (IFN)-γ in cauda equina infiltrating cells, and elevated levels of IgG2b antibodies against P0 peptide 180-199 in sera were found in TNF-α KO mice with EAN. In conclusion, TNF-α deficiency attenuates EAN via altering the M1/M2 balance of macrophages.


Assuntos
Macrófagos/patologia , Neurite Autoimune Experimental/imunologia , Neurite Autoimune Experimental/metabolismo , Fator de Necrose Tumoral alfa/deficiência , Sequência de Aminoácidos , Animais , Regulação para Baixo , Técnicas de Silenciamento de Genes , Imunoglobulina G/imunologia , Imunoglobulina G/metabolismo , Interferon gama/biossíntese , Interleucina-17/biossíntese , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Camundongos , Dados de Sequência Molecular , Proteína Básica da Mielina/química , Proteína Básica da Mielina/imunologia , Neurite Autoimune Experimental/genética , Fenótipo , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Linfócitos T Reguladores/imunologia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
19.
Neurobiol Aging ; 33(12): 2942-7, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22440674

RESUMO

Amyloid plaques associated with Alzheimer's disease (AD) induce inflammatory responses associated with activated microglia and reactive astrocytes, which exacerbate neurodegeneration through release of inflammatory cytokines, reactive oxygen species, and other factors. Inflammation contributes to neurodegeneration at later stages of AD, but it may also play a role in early disease pathogenesis. We found that before plaque deposition, amyloid precursor protein (APP)/presenilin 1 (PSEN1) transgenic mice (PSAPP mice), a well-characterized model of AD, exhibit evidence of cerebrovascular inflammation. Expression of the endothelial cell-specific antigen MECA-32 (mouse endothelial cell antigen-32) was upregulated in the cerebrovasculature of young PSAPP mice (3 months old) and was similar to that observed in mice with experimental autoimmune encephalomyelitis, a model of multiple sclerosis characterized by neuroinflammation. MECA-32 is normally expressed in central and peripheral vasculature throughout development, but expression in the cerebrovasculature is downregulated on establishment of the blood-brain barrier (BBB). However, CNS inflammation triggers re-expression of MECA-32 in compromised cerebrovasculature. Our study indicates that MECA-32 may be a robust marker of cerebrovascular inflammation and compromised BBB integrity, triggered by soluble amyloid-ß early in disease pathogenesis.


Assuntos
Doença de Alzheimer/complicações , Vasculite do Sistema Nervoso Central/etiologia , Vasculite do Sistema Nervoso Central/metabolismo , Doença de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Animais , Antígenos de Superfície/metabolismo , Barreira Hematoencefálica/patologia , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurite Autoimune Experimental/induzido quimicamente , Neurite Autoimune Experimental/metabolismo , Presenilina-1/genética , Regulação para Cima/fisiologia , Vasculite do Sistema Nervoso Central/patologia
20.
Brain Res Bull ; 86(3-4): 229-34, 2011 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-21784136

RESUMO

Experimental autoimmune neuritis (EAN) is a well-known animal model of human demyelinating polyneuropathies and is characterized by inflammation and demyelination in the peripheral nervous system. Tim-3 had been identified as a Th1-specific marker negatively regulating autoimmunity or inflammatory diseases. Here we have studied by immunohistochemistry the spatiotemporal accumulation of Tim-3(+) cells in sciatic nerves of EAN rats, particularly focusing on its association with alternatively activated macrophages. Our results showed that time course of Tim-3(+) cell accumulation correlated positively with disease progression of EAN; but distinct major cellular resources of Tim-3 were observed at different disease stages of EAN: during the early phase of EAN, the main cellular resource were T cells, but at the peak and during recovery phase of EAN, Tim-3 was mostly expressed on CD68(+) macrophages or CD163(+) cells. Further investigation suggested that accumulation of CD163(+) cells, particularly their relative abundances to activated macrophages at different time points, were in accordance with the recovery from EAN. Therefore, Tim3(+) cells might include a distinct macrophage population, which may be involved in anti-inflammatory effect and recovery from EAN.


Assuntos
Neurite Autoimune Experimental/metabolismo , Receptores de Superfície Celular/biossíntese , Receptores de Superfície Celular/genética , Animais , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Comportamento Animal/fisiologia , Interpretação Estatística de Dados , Progressão da Doença , Imuno-Histoquímica , Macrófagos/patologia , Masculino , Neurite Autoimune Experimental/psicologia , Ratos , Ratos Endogâmicos Lew , Receptores de Superfície Celular/metabolismo , Nervo Isquiático/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA