Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
J Neuroinflammation ; 20(1): 139, 2023 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-37296476

RESUMO

BACKGROUND: Autoimmune neuropathies can result in long-term disability and incomplete recovery, despite adequate first-line therapy. Kinesin-5 inhibition was shown to accelerate neurite outgrowth in different preclinical studies. Here, we evaluated the potential neuro-regenerative effects of the small molecule kinesin-5 inhibitor monastrol in a rodent model of acute autoimmune neuropathies, experimental autoimmune neuritis. METHODS: Experimental autoimmune neuritis was induced in Lewis rats with the neurogenic P2-peptide. At the beginning of the recovery phase at day 18, the animals were treated with 1 mg/kg monastrol or sham and observed until day 30 post-immunisation. Electrophysiological and histological analysis for markers of inflammation and remyelination of the sciatic nerve were performed. Neuromuscular junctions of the tibialis anterior muscles were analysed for reinnervation. We further treated human induced pluripotent stem cells-derived secondary motor neurons with monastrol in different concentrations and performed a neurite outgrowth assay. RESULTS: Treatment with monastrol enhanced functional and histological recovery in experimental autoimmune neuritis. Motor nerve conduction velocity at day 30 in the treated animals was comparable to pre-neuritis values. Monastrol-treated animals showed partially reinnervated or intact neuromuscular junctions. A significant and dose-dependent accelerated neurite outgrowth was observed after kinesin-5 inhibition as a possible mode of action. CONCLUSION: Pharmacological kinesin-5 inhibition improves the functional outcome in experimental autoimmune neuritis through accelerated motor neurite outgrowth and histological recovery. This approach could be of interest to improve the outcome of autoimmune neuropathy patients.


Assuntos
Células-Tronco Pluripotentes Induzidas , Neurite Autoimune Experimental , Ratos , Animais , Humanos , Neurite Autoimune Experimental/tratamento farmacológico , Neurite Autoimune Experimental/patologia , Cinesinas/uso terapêutico , Ratos Endogâmicos Lew , Células-Tronco Pluripotentes Induzidas/patologia
2.
J Neuroinflammation ; 20(1): 122, 2023 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-37217991

RESUMO

BACKGROUND: Autoimmune neuropathies are common peripheral nervous system (PNS) disorders. Environmental influences and dietary components are known to affect the course of autoimmune diseases. Intestinal microorganisms can be dynamically regulated through diet, and this study combines intestinal microorganisms with diseases to open up new therapeutic ideas. METHODS: In Lewis rats, a model of EAN was established with P0 peptide, Lactobacillus were used as treatment, serum T-cell ratio, inflammatory factors, sciatic neuropathological changes, and pathological inflammatory effects on intestinal mucosa were detected, and fecal metabolomics and 16 s microbiome analysis were performed to further explore the mechanism. RESULTS: In the EAN rat model, Lactobacillus paracasei L9 (LP) could dynamically regulate the CD4+/CD8+T balance in serum, reduce serum IL-1, IL-6 and TNF-α expression levels, improve sciatic nerve demyelination and inflammatory infiltration, and reduce nervous system score. In the rat model of EAN, intestinal mucosa was damaged. Occludin and ZO-1 were downregulated. IL-1, TNF-α and Reg3γ were upregulated. LP gavage induced intestinal mucosa recovery; occludin and ZO-1 upregulation; IL-1, TNF-α and Reg3γ downregulation. Finally, metabolomics and 16 s microbiome analysis were performed, and differential metabolites were enriched with an important metabolic pathway, arginine and proline metabolism. CONCLUSION: LP improved EAN in rats by influencing intestinal community and the lysine and proline metabolism.


Assuntos
Microbioma Gastrointestinal , Lacticaseibacillus paracasei , Neurite Autoimune Experimental , Ratos , Animais , Neurite Autoimune Experimental/patologia , Fator de Necrose Tumoral alfa/metabolismo , Ocludina/metabolismo , Ratos Endogâmicos Lew , Nervo Isquiático/patologia , Progressão da Doença , Interleucina-1/metabolismo , Prolina/metabolismo , Prolina/farmacologia , Prolina/uso terapêutico
3.
J Neuroinflammation ; 20(1): 35, 2023 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-36788526

RESUMO

BACKGROUND: Guillain-Barré syndrome (GBS) and chronic inflammatory demyelinating polyneuropathy (CIDP) are human autoimmune peripheral neuropathy. Besides humoral immunity, cellular immunity is also believed to contribute to these pathologies, especially CIDP. Sphingosine-1-phosphate receptor 1 (S1PR1) regulates the maturation, migration, and trafficking of lymphocytes. As of date, the therapeutic effect of sphingosine-1-phosphate receptor (S1PR) agonists on patients with GBS or CIDP remains unclear. METHODS: To evaluate the effect of siponimod, an agonist of S1PR1 and S1PR5, on experimental autoimmune neuritis (EAN), an animal model of autoimmune peripheral neuropathy, was used. Lewis rats were immunized with 125 µg of synthetic peptide from bovine P2 protein. Rats in the siponimod group were orally administered 1.0 mg/kg siponimod and those in the EAN group were administrated the vehicle on days 5-27 post-immunization (p.i.) daily. The symptom severity was recorded daily. The changes in the expression of cytokines and transcription factors in the lymph nodes and cauda equina (CE) which correlate with the pathogenesis of EAN and recovery of injured nerve were measured using reverse transcription quantitative PCR. Histological study of CE was also performed. RESULTS: Flaccid paralysis developed on day 11 p.i. in both groups. Siponimod relieved the symptom severity and decreased the expression of interferon-gamma and IL-10 mRNAs in lymph nodes and CE compared with that in the EAN group. The expression of Jun proto-oncogene (c-Jun) mRNA increased from the peak to the recovery phase and that of Sonic hedgehog signaling molecule (Shh) and Glial cell line-derived neurotrophic factor (Gdnf) increased prior to increase in c-Jun with no difference observed between the two groups. Histologically, siponimod also reduced demyelinating lesions and inflammatory cell invasion in CE. CONCLUSIONS: Siponimod has a potential to ameliorate EAN. Shh and Gdnf, as well as C-Jun played a significant role during the recovery of injured nerves.


Assuntos
Síndrome de Guillain-Barré , Neurite Autoimune Experimental , Polirradiculoneuropatia Desmielinizante Inflamatória Crônica , Humanos , Ratos , Animais , Bovinos , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Nervo Isquiático/patologia , Proteínas Hedgehog/metabolismo , Ratos Endogâmicos Lew , Síndrome de Guillain-Barré/tratamento farmacológico
4.
BMC Neurol ; 23(1): 48, 2023 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-36709251

RESUMO

BACKGROUND: Guillain-Barré syndrome (GBS) is a rare, autoimmune disease. B.infantis is reported to be effective in alleviating GBS by regulating abnormal function of T helper (Th) cells. OBJECTIVES: In this study, T cells were isolated from healthy and GBS patients. The therapeutic effect of Bifidobacterium infantis (B.infantis) and whether it is achieved by PD-1 was examined at cellular and animal models. METHODS: We used CCK-8, flow cytometry and real-time PCR to determine the differentiation of T cell subsets at cellular level. Then, an experimental autoimmune neuritis (EAN) animal model using six-week SD rats (n = 30, male) weighing 180-200 g was established to support the role of B. infantis in GBS through PD-1. RESULTS: B. infantis inhibited the proliferation and promoted apoptosis of T cells from GBS. At the same time, the expression levels of PD-1 increased, which was correlated with decreased T-bet (Th1) and ROR-γt (Th17) and increased Foxp3 (Treg) expression. Moreover, B. infantis alleviated the symptoms of GBS. Th1 and Th17 cells decreased while Treg cells increased after B. infantis treatment, which could be partly abrogated by PD-1 inhibitor. CONCLUSIONS: We concluded from this study that B.infantis alleviated GBS partly through PD-1.


Assuntos
Síndrome de Guillain-Barré , Masculino , Ratos , Animais , Bifidobacterium longum subspecies infantis , Receptor de Morte Celular Programada 1/uso terapêutico , Ratos Sprague-Dawley , Imunoterapia
5.
J Autoimmun ; 133: 102945, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36356552

RESUMO

Monocytes in peripheral blood and sciatic nerves play vital roles in immune-mediated neuropathies such as Guillain-Barré syndrome (GBS). Different subpopulations of monocytes, including classical and non-classical, exhibit distinct functions as well as phenotypic conversion potentials. However, the mechanisms underlying their development during immune-mediated neuropathy remain unclear. Notch signaling participates in monocyte differentiation and function. In this study, we used a myeloid-specific Notch signaling activation transgenic mouse (NICcA) and investigated the role of Notch signaling in monocytes during experimental autoimmune neuritis (EAN) in a mouse model of GBS. Clinical score assessment and histopathological examination revealed that sciatic nerve injury was attenuated in NICcA EAN mice compared to that in control mice. Flow cytometry and immunofluorescence staining suggested that increasing Ly6Clo monocytes in the peripheral blood and nerve tissue might contribute to the alleviation of neuritis in NICcA mice. Meanwhile, an in vitro study suggested that bone marrow-derived monocytes from NICcA mice are more inclined toward Ly6Clo cells than Ly6Chi cells. Differential expression of monocyte development-associated genes was detected in NICcA and wild-type mice using RNA sequencing. The expression of Nr4a1 is upregulated remarkably when Notch signaling is activated. Treatment with Nr4a1 antagonist on NICcA mice-derived monocytes compromise their Ly6Clo tendency. Consistently, a relationship between monocyte conversion and disease severity was observed in blood samples from patients with GBS. In conclusion, our current study showed that monocyte conversion modulated by Notch signaling plays an essential role in the EAN mouse model.


Assuntos
Monócitos , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares , Camundongos , Animais , Análise de Sequência de RNA
6.
J Neuroinflammation ; 18(1): 293, 2021 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-34920725

RESUMO

BACKGROUND: Lysophosphatidic acid (LPA) is a pleiotropic lipid messenger that addresses at least six specific G-protein coupled receptors. Accumulating evidence indicates a significant involvement of LPA in immune cell regulation as well as Schwann cell physiology, with potential relevance for the pathophysiology of peripheral neuroinflammation. However, the role of LPA signaling in inflammatory neuropathies has remained completely undefined. Given the broad expression of LPA receptors on both Schwann cells and cells of the innate and adaptive immune system, we hypothesized that inhibition of LPA signaling may ameliorate the course of disease in experimental autoimmune neuritis (EAN). METHODS: We induced active EAN by inoculation of myelin protein 2 peptide (P255-78) in female Lewis rats. Animals received the orally available LPA receptor antagonist AM095, specifically targeting the LPA1 receptor subtype. AM095 was administered daily via oral gavage in a therapeutic regimen from 10 until 28 days post-immunization (dpi). Analyses were based on clinical testing, hemogram profiles, immunohistochemistry and morphometric assessment of myelination. RESULTS: Lewis rats treated with AM095 displayed a significant improvement in clinical scores, most notably during the remission phase. Cellular infiltration of sciatic nerve was only discretely affected by AM095. Hemogram profiles indicated no impact on circulating leukocytes. However, sciatic nerve immunohistochemistry revealed a reduction in the number of Schwann cells expressing the dedifferentiation marker Sox2 paralleled by a corresponding increase in differentiating Sox10-positive Schwann cells. In line with this, morphometric analysis of sciatic nerve semi-thin sections identified a significant increase in large-caliber myelinated axons at 28 dpi. Myelin thickness was unaffected by AM095. CONCLUSION: Thus, LPA1 signaling may present a novel therapeutic target for the treatment of inflammatory neuropathies, potentially affecting regenerative responses in the peripheral nerve by modulating Schwann cell differentiation.


Assuntos
Desdiferenciação Celular/fisiologia , Neurite Autoimune Experimental/imunologia , Receptores de Ácidos Lisofosfatídicos/imunologia , Células de Schwann/imunologia , Transdução de Sinais/fisiologia , Animais , Compostos de Bifenilo/farmacologia , Compostos de Bifenilo/uso terapêutico , Desdiferenciação Celular/efeitos dos fármacos , Feminino , Isoxazóis/farmacologia , Isoxazóis/uso terapêutico , Neurite Autoimune Experimental/tratamento farmacológico , Neurite Autoimune Experimental/metabolismo , Ratos , Ratos Endogâmicos Lew , Receptores de Ácidos Lisofosfatídicos/antagonistas & inibidores , Receptores de Ácidos Lisofosfatídicos/metabolismo , Células de Schwann/efeitos dos fármacos , Células de Schwann/metabolismo , Transdução de Sinais/efeitos dos fármacos
7.
J Peripher Nerv Syst ; 26 Suppl 2: S61-S68, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34498774

RESUMO

To understand the pathology of axonal degeneration and demyelination in peripheral neuropathy, histological investigations in different animal models that mimic some aspects of human peripheral neuropathy are needed. Thus, in the following section of this special issue, the main pathological features of experimental autoimmune neuritis, animal models of chemotherapy-induced peripheral neuropath and of human inherited peripheral neuropathies (IPNs) will be illustrated. When possible, micrographs from animal models and selected human biopsy will be shown side by side.


Assuntos
Doença de Charcot-Marie-Tooth , Animais , Doença de Charcot-Marie-Tooth/patologia , Humanos , Modelos Animais
8.
Int J Neurosci ; 130(11): 1109-1117, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32009498

RESUMO

Background: Guillain-Barré syndrome (GBS), an autoimmune disease and an acute inflammation disorder, is currently the most frequent cause of acute flaccid paralysis worldwide. EAN, an animal model of GBS, is a CD4+ T cell-mediated autoimmune disease of the PNS. Wnt/ß-catenin signals are critically important to several fundamental aspects of peripheral nerve development and play a crucial role in Schwann cell proliferation. Here, we investigate the role of Wnt/ß-catenin signalling cascades in EAN rats.Methods: 28 male Lewis rats weighing 170 ± 10 g were randomly divided into control group (n = 7) and EAN groups (Early group; Peak group and Recovery group. n = 7 per group). EAN rats were immunized with P257-81 peptide; weighed daily, and the neurologic signs of EAN were evaluated every day. The sciatic nerve was taken on the days 10, 17, and 30 p.i. for H&E staining, transmission electron microscopy and immunohistochemical staining; blood samples were collected weekly from caudal vein to detect IFN-γ, IL-4, TGF-ß1; and the sciatic nerve was taken to examinate the dynamics expression of Wnt/ß-catenin pathway molecules.Results: In our study, we chose tail-root injection to better model GBS. Moreover, we observed that IFN-γ levels paralleled clinical EAN, and the levels of TGF-ß1 and IL-4 gradually increased and peaked in the recovery phase. In addition, we have shown that canonical Wnt signalling is upregulated and reached a peak in the late recovery phase.Conclusion: Our findings suggest that Wnt/ß-catenin signalling is associated with the promotion of remyelination in EAN rats.


Assuntos
Síndrome de Guillain-Barré , Interferon gama/sangue , Interleucina-4/sangue , Neurite Autoimune Experimental , Remielinização , Nervo Isquiático , Fator de Crescimento Transformador beta1/sangue , Via de Sinalização Wnt , Animais , Síndrome de Guillain-Barré/imunologia , Síndrome de Guillain-Barré/metabolismo , Síndrome de Guillain-Barré/patologia , Masculino , Neurite Autoimune Experimental/imunologia , Neurite Autoimune Experimental/metabolismo , Neurite Autoimune Experimental/patologia , Ratos , Ratos Endogâmicos Lew , Remielinização/fisiologia , Nervo Isquiático/imunologia , Nervo Isquiático/metabolismo , Nervo Isquiático/patologia , Regulação para Cima , Via de Sinalização Wnt/fisiologia
9.
J Neuroinflammation ; 16(1): 58, 2019 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-30851725

RESUMO

BACKGROUND: Corticosteroids dominate in the treatment of chronic autoimmune neuropathies although long-term use is characterized by devastating side effects. METHODS: We introduce the intrathecal application of the synthetic steroid triamcinolone (TRIAM) as a novel therapeutic option in experimental autoimmune neuritis in Lewis rats RESULTS: After immunization with neuritogenic P2 peptide, we show a dose-dependent therapeutic effect of one intrathecal injection of 0.3 or 0.6 mg/kg TRIAM on clinical and electrophysiological parameters of neuritis with a lower degree of inflammatory infiltrates (T cells and macrophages) and demyelination in the sciatic nerve. In vitro studies in Schwann cell cultures showed an increased expression of IL-1 receptor antagonist and reduced expression of Toll-like receptor 4 after incubation with TRIAM as well as a protective effect of TRIAM against oxidative stress after H2O2 exposure. CONCLUSION: Intrathecal TRIAM application could be a novel immunomodulatory and potentially neuroprotective option for autoimmune neuropathies with a direct effect on Schwann cells.


Assuntos
Anti-Inflamatórios/administração & dosagem , Neurite Autoimune Experimental/tratamento farmacológico , Neurite Autoimune Experimental/patologia , Estresse Oxidativo/efeitos dos fármacos , Células de Schwann/efeitos dos fármacos , Triancinolona Acetonida/administração & dosagem , Animais , Antígenos CD/metabolismo , Técnicas de Cultura de Células , Modelos Animais de Doenças , Adjuvante de Freund/toxicidade , Regulação da Expressão Gênica/efeitos dos fármacos , Injeções Espinhais/métodos , Linfonodos/citologia , Masculino , Condução Nervosa/efeitos dos fármacos , Neurite Autoimune Experimental/induzido quimicamente , Ratos , Ratos Endogâmicos Lew , Fatores de Transcrição SOXE/metabolismo , Antígenos Thy-1/metabolismo
10.
Clin Exp Immunol ; 198(2): 184-197, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31344254

RESUMO

Guillain-Barré syndrome (GBS) is characterized by acute immune-mediated peripheral neuropathy, which may result in rapidly progressive paralysis and fatal respiratory failure. As the underlying pathological mechanisms of GBS are unclear, we surveyed the transcriptome of rats with experimental autoimmune neuritis (EAN), a model of GBS. Briefly, sciatic nerves on both sides were collected from 8-10-week-old Lewis rats during early (10 days post-induction), peak (19 days) and late neuritis (30 days). Total RNA was sequenced to identify differentially expressed genes. Compared to control rats without induced neuritis, 33 genes were differentially expressed in the early phase (14 up-regulated and 19 down-regulated), with an adjusted P-value < 0·05 and |log2 fold-change| >1, as were 137 genes in the peak phase (126 up-regulated and 11 down-regulated) and 60 genes in the late phase (58 up-regulated and two down-regulated). Eleven of these genes were common to all stages, suggesting their crucial roles throughout the disease course. Analysis of protein-protein interactions revealed Fos, Ccl2, Itgax and C3 as node genes at different stages. Functional analysis of differentially expressed genes identified biological processes and pathways that are activated as neuritis progresses. This is the first genomewide gene expression study of peripheral nerves in experimental autoimmune neuritis model. Dynamic gene expression and significantly altered biological functions were detected in different phases of the disease, increasing our understanding of the molecular mechanisms underlying EAN and highlighting potential targets for its diagnosis and treatment.


Assuntos
Perfilação da Expressão Gênica , Regulação da Expressão Gênica/imunologia , Neurite Autoimune Experimental , Nervo Isquiático/imunologia , Análise de Sequência de RNA , Transcriptoma/imunologia , Animais , Feminino , Neurite Autoimune Experimental/genética , Neurite Autoimune Experimental/imunologia , Neurite Autoimune Experimental/patologia , Ratos , Nervo Isquiático/patologia
11.
Toxicol Pathol ; 47(4): 542-552, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30987532

RESUMO

Experimental autoimmune neuritis (EAN) is an animal model for Guillain-Barré syndrome (GBS), which results in neurological symptoms and histopathological changes in peripheral nerves. In this model, the correlation between the progression of the disease and the histopathological changes is not clear. To further examine histopathological changes in peripheral nerves in EAN rats, sciatic nerves were sampled at onset (day 10), peak (day 16), and recovery (days 22 and 25) of neurological symptoms in P2(57-81)-peptide-administered rats. Axon and myelin degeneration was observed by light microscopy at onset, degeneration became severe at peak, and persisted at recovery. Densities of myelinated nerve fibers and myelin areas decreased from day 10 to a minimum on day 22. Slight axon and myelin degeneration, such as accumulation of vesicles in axons and focal myelin splitting and folding, was observed by transmission electron microscopy at onset; severe degeneration, such as axonal loss, myelin ovoid, and demyelination, increased at peak; and regenerative changes, such as remyelination and enlargement of Schwann cell cytoplasm, occurred at recovery. These results suggest that EAN rats have histopathological similarities to some types of GBS patients and that EAN rats are a useful model to understand the pathogenesis of GBS.


Assuntos
Axônios/ultraestrutura , Síndrome de Guillain-Barré/patologia , Bainha de Mielina/ultraestrutura , Neurite Autoimune Experimental/patologia , Nervo Isquiático/patologia , Animais , Síndrome de Guillain-Barré/imunologia , Masculino , Microscopia Eletrônica de Transmissão , Proteína P2 de Mielina/imunologia , Fibras Nervosas Mielinizadas/ultraestrutura , Neurite Autoimune Experimental/imunologia , Fragmentos de Peptídeos/imunologia , Ratos Endogâmicos Lew
12.
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
13.
Cell Physiol Biochem ; 47(1): 390-402, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29772575

RESUMO

BACKGROUND/AIMS: This study aimed to explore whether the adoptive transfusion of autologous CD4+CD25+ regulatory T cells (CD4+CD25+ Tregs) has a therapeutic effect on Experimental autoimmune neuritis (EAN) model rats, and it provides new experimental and theoretical bases for the immunotherapy of Guillain-Barre syndrome (GBS). METHODS: CD4+CD25+ Tregs were sorted from the spleens of rats using immunomagnetic bead separation techniques combined with flow cytometry. Their in vitro inhibitory function was determined using a lymphocyte proliferation inhibition test, and their purity was confirmed by flow cytometry. Cells were stimulated using CD3/CD28 monoclonal antibodies and were cultured in culture medium containing interleukin 2 (IL-2), transforming growth factor-ß (TGF-ß) and rapamycin. After 15 days of amplification, CD4+CD25+ Tregs were collected and transfused into EAN model rats. Changes in the pathology and electron microscopical morphology of rat sciatic nerves in the normal group, untreated group, low-dose group (2 × 107) and high-dose group (4 × 107) were observed, and the expression of CD4+CD25+FOXP3 in peripheral blood in the four groups of rats was detected by flow cytometry. RESULTS: Compared with rats in the untreated group, rats in the treatment groups had significantly reduced infiltration of inflammatory cells in the sciatic nerve, as well as myelin and axonal damage. Additionally, the CD4+CD25+ Tregs levels in peripheral blood were significantly higher than those in the untreated group (P< 0. 05). Moreover, the therapeutic effect became more significant with an increase in the dose of adoptive transfusion. CONCLUSION: Adoptive transfusion of CD4+CD25+ Tregs into EAN model rats has significant therapeutic effects.


Assuntos
Transferência Adotiva , Antígenos CD4/imunologia , Subunidade alfa de Receptor de Interleucina-2/imunologia , Neurite Autoimune Experimental/terapia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/transplante , Transferência Adotiva/métodos , Animais , Células Cultivadas , Modelos Animais de Doenças , Feminino , Neurite Autoimune Experimental/imunologia , Neurite Autoimune Experimental/patologia , Ratos , Ratos Endogâmicos Lew
14.
J Neuroinflammation ; 15(1): 51, 2018 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-29467007

RESUMO

BACKGROUND: With the recognition of the key roles of cellular metabolism in immunity, targeting metabolic pathway becomes a new strategy for autoimmune disease treatment. Guillain-Barré syndrome (GBS) is an acute immune-mediated inflammatory demyelinating disease of the peripheral nervous system, characterized by inflammatory cell infiltration. These inflammatory cells, including activated macrophages, Th1 cells, and Th17 cells, generally undergo metabolic reprogramming and rely mainly on glycolysis to exert functions. This study aimed to explore whether enhanced glycolysis contributed to the pathogenesis of experimental autoimmune neuritis (EAN), a classic model of GBS. METHODS: Preventive and therapeutic treatments with glycolysis inhibitor, 2-deoxy-D-glucose (2-DG), were applied to EAN rats. The effects of treatments were determined by clinical scoring, weighting, and tissue examination. Flow cytometry and ELISA were used to evaluate T cell differentiation, autoantibody level, and macrophage functions in vivo and in vitro. RESULTS: Glycolysis inhibition with 2-DG not only inhibited the initiation, but also prevented the progression of EAN, evidenced by the improved clinical scores, weight loss, inflammatory cell infiltration, and demyelination of sciatic nerves. 2-DG inhibited the differentiation of Th1, Th17, and Tfh cells but enhanced Treg cell development, accompanied with reduced autoantibody secretion. Further experiments in vitro proved glycolysis inhibition decreased the nitric oxide production and phagocytosis of macrophages and suppressed the maturation of dendritic cells (DC). CONCLUSION: The effects of glycolysis inhibition on both innate and adaptive immune responses and the alleviation of animal clinical symptoms indicated that enhanced glycolysis contributed to the pathogenesis of EAN. Glycolysis inhibition may be a new therapy for GBS.


Assuntos
Glicólise/fisiologia , Neurite Autoimune Experimental/induzido quimicamente , Neurite Autoimune Experimental/metabolismo , Animais , Desoxiglucose/farmacologia , Desoxiglucose/uso terapêutico , Glicólise/efeitos dos fármacos , Imunidade Celular/efeitos dos fármacos , Imunidade Celular/fisiologia , Lipopolissacarídeos/toxicidade , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/metabolismo , Camundongos , Neurite Autoimune Experimental/tratamento farmacológico , Células RAW 264.7 , Ratos , Ratos Endogâmicos Lew
15.
J Neuroinflammation ; 15(1): 61, 2018 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-29486771

RESUMO

BACKGROUND: Small-diameter, myelinated axons are selectively susceptible to dysfunction in several inflammatory PNS and CNS diseases, resulting in pain and degeneration, but the mechanism is not known. METHODS: We used in vivo confocal microscopy to compare the effects of inflammation in experimental autoimmune neuritis (EAN), a model of Guillain-Barré syndrome (GBS), on mitochondrial function and transport in large- and small-diameter axons. We have compared mitochondrial function and transport in vivo in (i) healthy axons, (ii) axons affected by experimental autoimmune neuritis, and (iii) axons in which mitochondria were focally damaged by laser induced photo-toxicity. RESULTS: Mitochondria affected by inflammation or laser damage became depolarized, fragmented, and immobile. Importantly, the loss of functional mitochondria was accompanied by an increase in the number of mitochondria transported towards, and into, the damaged area, perhaps compensating for loss of ATP and allowing buffering of the likely excessive Ca2+ concentration. In large-diameter axons, healthy mitochondria were found to move into the damaged area bypassing the dysfunctional mitochondria, re-populating the damaged segment of the axon. However, in small-diameter axons, the depolarized mitochondria appeared to "plug" the axon, obstructing, sometimes completely, the incoming (mainly anterograde) transport of mitochondria. Over time (~ 2 h), the transported, functional mitochondria accumulated at the obstruction, and the distal part of the small-diameter axons became depleted of functional mitochondria. CONCLUSIONS: The data show that neuroinflammation, in common with photo-toxic damage, induces depolarization and fragmentation of axonal mitochondria, which remain immobile at the site of damage. The damaged, immobile mitochondria can "plug" myelinated, small-diameter axons so that successful mitochondrial transport is prevented, depleting the distal axon of functioning mitochondria. Our observations may explain the selective vulnerability of small-diameter axons to dysfunction and degeneration in a number of neurodegenerative and neuroinflammatory disorders.


Assuntos
Axônios/metabolismo , Mitocôndrias/metabolismo , Fibras Nervosas Mielinizadas/metabolismo , Neurite Autoimune Experimental/metabolismo , Nervos Periféricos/metabolismo , Animais , Axônios/patologia , Transporte Biológico/fisiologia , Inflamação/metabolismo , Inflamação/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mitocôndrias/patologia , Fibras Nervosas Mielinizadas/patologia , Neurite Autoimune Experimental/patologia , Nervos Periféricos/patologia
16.
J Neuroinflammation ; 15(1): 122, 2018 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-29690884

RESUMO

BACKGROUND: Autoimmune neuropathies are common PNS disorders and effective treatment is challenging. Environmental influence and dietary components are known to affect the course of autoimmune diseases. Capsaicin as pungent component of chili-peppers is common in human nutrition. An influence of capsaicin on autoimmune diseases has been postulated. METHODS: We tested capsaicin in the animal model of experimental autoimmune neuritis (EAN) in Lewis rat. Rats were immunized with P2-peptide and were treated with capsaicin in different preventive settings. Electrophysiological, histological, and molecular biological analyses of the sciatic nerve were performed to analyze T-cell and macrophage cell count, TRPV1, and cytokine expression. Moreover, FACS analyses including the intestinal immune system were executed. RESULTS: We observed an immunomodulatory effect of an early preventive diet-concept, where a physiological dosage of oral capsaicin was given 10 days before immunization in EAN. A reduced inflammation of the sciatic nerve was significant detectable clinically, electrophysiologically (CMAPs reduced in control group p < 0.01; increase of nerve conduction blocks in control group p < 0.05), histologically (significant reduction of T-cells, macrophages and demyelination), and at cytokine level. In contrast, this therapeutic effect was missing with capsaicin given from the day of immunization onwards. As possible underlying mechanism, we were able to show changes in the expression of the capsaicin receptor in the sciatic nerve and the small intestine, as well as altered immune cell populations in the small intestine. CONCLUSION: This is the first report about the immunomodulatory effect of the common nutrient, capsaicin, in an experimental model for autoimmune neuropathies.


Assuntos
Capsaicina/uso terapêutico , Dieta/métodos , Neurite Autoimune Experimental/dietoterapia , Animais , Movimento Celular/efeitos dos fármacos , Citocinas/metabolismo , Modelos Animais de Doenças , Potencial Evocado Motor/efeitos dos fármacos , Feminino , Adjuvante de Freund/toxicidade , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/patologia , Macrófagos/efeitos dos fármacos , Macrófagos/patologia , Condução Nervosa/efeitos dos fármacos , Neurite Autoimune Experimental/patologia , Neurite Autoimune Experimental/fisiopatologia , Ratos , Ratos Endogâmicos Lew , Proteínas S100/metabolismo , Nervo Isquiático/efeitos dos fármacos , Nervo Isquiático/metabolismo , Nervo Isquiático/patologia , Linfócitos T/efeitos dos fármacos , Canais de Cátion TRPV/metabolismo
17.
J Neuroinflammation ; 15(1): 217, 2018 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-30068351

RESUMO

BACKGROUND: Autoimmune polyneuropathies are acquired inflammatory disorders of the peripheral nervous system (PNS) characterized by inflammation, demyelination, and axonal degeneration. Although the pathogenesis has not been fully elucidated, T cells recognizing self-antigens are believed to initiate inflammation in a subgroup of patients. However, the route and time of T cell entry into the PNS have not yet been described in detail. In this study, we analyzed both kinetics as well as localization of retrovirally transfected green fluorescent protein (GFP)-expressing neuritogenic T lymphocytes in experimental autoimmune neuritis (EAN). METHODS: T lymphocytes obtained from rats following EAN induction by immunization with peripheral nerve protein peptide P255-78 were retrovirally engineered to express GFP. Non-specific T cells were negatively selected by in vitro restimulation, whereas GFP-expressing neuritogenic T cells (reactive to P255-78) were adoptively transferred into healthy rats (AT-EAN). Antigen-specific T cell tracking and localization was performed by flow cytometry and immunohistochemistry during the course of disease. RESULTS: After induction of autoimmune neuritis, P2-reactive T cells were detectable in the liver, spleen, lymph nodes, lung, peripheral blood, and the sciatic nerves with distinct kinetics. A significant number of GFP+ T cells appeared early in the lung with a peak at day four. In the peripheral nerves within the first days, GFP-negative T cells rapidly accumulated and exceeded the number of GFP-expressing cells, but did not enter the endoneurium. Very early after adoptive transfer, T cells are found in proximity to peripheral nerves and in the epineurium. However, only GFP-expressing neuritogenic T cells are able to enter the endoneurium from day five after transfer. CONCLUSIONS: Our findings suggest that neuritogenic T cells invade the PNS early in the course of disease. However, neuritogenic T cells cross the blood-nerve barrier with a certain delay without preference to dorsal roots. Further understanding of the pathophysiological role of autoagressive T cells may help to improve therapeutic strategies in immune-mediated neuropathies.


Assuntos
Neurite Autoimune Experimental/imunologia , Neurite Autoimune Experimental/patologia , Nervos Periféricos/patologia , Linfócitos T/fisiologia , Transferência Adotiva , Animais , Antígenos CD4/metabolismo , Proliferação de Células/fisiologia , Modelos Animais de Doenças , Feminino , Citometria de Fluxo , Adjuvante de Freund/toxicidade , Regulação da Expressão Gênica/imunologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Proteína P2 de Mielina/metabolismo , Neurite Autoimune Experimental/induzido quimicamente , Neurite Autoimune Experimental/cirurgia , Fragmentos de Peptídeos/metabolismo , Ratos , Ratos Endogâmicos Lew , Linfócitos T/metabolismo , Fatores de Tempo , Transdução Genética
18.
Cell Immunol ; 329: 41-49, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29724464

RESUMO

The Class I phosphatidylinositol 3-kinase inhibitor, 2-(2-difluoromethy lbenzimidazol-1-yl)-4,6-dimorpholino-1,3,5-triazine (ZSTK474), has anti-inflammatory and immunoregulatory properties. However, whether it can be used to treat Guillain-Barré syndrome (GBS)-a neuroinflammatory disorder-is unknown. We induced experimental autoimmune neuritis (EAN) in Lewis rats, an established model of GBS. Orally administered ZSTK474 decreased neurological deficits in the GBS model, as demonstrated by diminished inflammatory cell infiltration, and ameliorated demyelination of sciatic nerves. Additionally, ZSTK474 decreased the number of Th1/Th17 cells and levels of the proinflammatory cytokines interleukin (IL)-1α, IL-1ß, IL-17, IL-23, interferon-γ, and tumor necrosis factor-α. We propose that the phosphoinositide 3-kinase/AKT/mammalian target of rapamycin complex 1 (PI3K/AKT/mTORC1) pathway likely contributed to the neuroprotective effect of ZSTK474. ZSTK474 effectively decreases the frequency of Th1/Th17 cells, thereby reducing the production of proinflammatory cytokines and successfully alleviating the symptoms of EAN. Thus, the neuroprotective effect of ZSTK474 indicates its potential utility as anti-inflammatory therapy for GBS.


Assuntos
Citocinas/efeitos dos fármacos , Neurite Autoimune Experimental/tratamento farmacológico , Triazinas/farmacologia , Animais , Doenças Desmielinizantes/tratamento farmacológico , Modelos Animais de Doenças , Síndrome de Guillain-Barré/tratamento farmacológico , Masculino , Fármacos Neuroprotetores/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Ratos , Ratos Endogâmicos Lew , Nervo Isquiático , Células Th1/efeitos dos fármacos , Células Th17/efeitos dos fármacos , Triazinas/metabolismo
19.
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
20.
J Neuroinflammation ; 14(1): 232, 2017 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-29179723

RESUMO

Inflammatory neuropathies are rare autoimmune-mediated disorders affecting the peripheral nervous system. Considerable progress has recently been made in understanding pathomechanisms of these disorders which will be essential for developing novel diagnostic and therapeutic strategies in the future. Here, we summarize our current understanding of antigenic targets and the relevance of new immunological concepts for inflammatory neuropathies. In addition, we provide an overview of available animal models of acute and chronic variants and how new diagnostic tools such as magnetic resonance imaging and novel therapeutic candidates will benefit patients with inflammatory neuropathies in the future. This review thus illustrates the gap between pre-clinical and clinical findings and aims to outline future directions of development.


Assuntos
Síndrome de Guillain-Barré/fisiopatologia , Polirradiculoneuropatia Desmielinizante Inflamatória Crônica/fisiopatologia , Animais , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA