RESUMO
The role of Pannexin (PANX) channels during collective and single cell migration is increasingly recognized. Amongst many functions that are relevant to cell migration, here we focus on the role of PANX-mediated adenine nucleotide release and associated autocrine and paracrine signaling. We also summarize the contribution of PANXs with the cytoskeleton, which is also key regulator of cell migration. PANXs, as mechanosensitive ATP releasing channels, provide a unique link between cell migration and purinergic communication. The functional association with several purinergic receptors, together with a plethora of signals that modulate their opening, allows PANX channels to integrate physical and chemical cues during inflammation. Ubiquitously expressed in almost all immune cells, PANX1 opening has been reported in different immunological contexts. Immune activation is the epitome coordination between cell communication and migration, as leukocytes (i.e., T cells, dendritic cells) exchange information while migrating towards the injury site. In the current review, we summarized the contribution of PANX channels during immune cell migration and recruitment; although we also compile the available evidence for non-immune cells (including fibroblasts, keratinocytes, astrocytes, and cancer cells). Finally, we discuss the current evidence of PANX1 and PANX3 channels as a both positive and/or negative regulator in different inflammatory conditions, proposing a general mechanism of these channels contribution during cell migration.
Assuntos
Movimento Celular/fisiologia , Conexinas/fisiologia , Células Dendríticas/fisiologia , Leucócitos/fisiologia , Fagócitos/fisiologia , Nucleotídeos de Adenina/fisiologia , Envelhecimento/imunologia , Envelhecimento/fisiologia , Animais , Astrócitos/fisiologia , Polaridade Celular , Quimiotaxia de Leucócito/fisiologia , Citoesqueleto/fisiologia , Fibroblastos/fisiologia , Humanos , Inflamação/imunologia , Inflamação/fisiopatologia , Queratinócitos/fisiologia , Mecanotransdução Celular/fisiologia , Neoplasias/imunologia , Degeneração Neural/imunologia , Degeneração Neural/fisiopatologia , Proteínas do Tecido Nervoso/fisiologia , Receptores Purinérgicos/fisiologiaAssuntos
Doenças Cerebelares/induzido quimicamente , Neoplasias Cerebelares/terapia , Inibidores de Checkpoint Imunológico/efeitos adversos , Síndrome Miastênica de Lambert-Eaton/induzido quimicamente , Degeneração Neural/induzido quimicamente , Tumores Neuroendócrinos/terapia , Nivolumabe/efeitos adversos , Amifampridina/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Canais de Cálcio Tipo P , Canais de Cálcio Tipo Q , Doenças Cerebelares/tratamento farmacológico , Doenças Cerebelares/imunologia , Doenças Cerebelares/fisiopatologia , Neoplasias Cerebelares/secundário , Feminino , Glucocorticoides/uso terapêutico , Humanos , Imunoglobulinas Intravenosas/uso terapêutico , Fatores Imunológicos/uso terapêutico , Síndrome Miastênica de Lambert-Eaton/tratamento farmacológico , Síndrome Miastênica de Lambert-Eaton/imunologia , Síndrome Miastênica de Lambert-Eaton/fisiopatologia , Neoplasias Pulmonares/diagnóstico por imagem , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/terapia , Linfonodos/diagnóstico por imagem , Imageamento por Ressonância Magnética , Pessoa de Meia-Idade , Degeneração Neural/tratamento farmacológico , Degeneração Neural/imunologia , Degeneração Neural/fisiopatologia , Tumores Neuroendócrinos/secundário , Fármacos Neuromusculares/uso terapêutico , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Prednisona/uso terapêutico , Radiocirurgia , Radioterapia , Rituximab/uso terapêutico , Carcinoma de Pequenas Células do Pulmão/diagnóstico por imagem , Carcinoma de Pequenas Células do Pulmão/secundário , Carcinoma de Pequenas Células do Pulmão/terapia , Tomografia Computadorizada por Raios XRESUMO
BACKGROUND: The neuronal ceroid lipofuscinoses (CLN diseases) are fatal lysosomal storage diseases causing neurodegeneration in the CNS. We have previously shown that neuroinflammation comprising innate and adaptive immune reactions drives axonal damage and neuron loss in the CNS of palmitoyl protein thioesterase 1-deficient (Ppt1-/-) mice, a model of the infantile form of the diseases (CLN1). Therefore, we here explore whether pharmacological targeting of innate immune cells modifies disease outcome in CLN1 mice. METHODS: We applied treatment with PLX3397 (150 ppm in the chow), a potent inhibitor of the colony stimulating factor-1 receptor (CSF-1R) to target innate immune cells in CLN1 mice. Experimental long-term treatment was non-invasively monitored by longitudinal optical coherence tomography and rotarod analysis, as well as analysis of visual acuity, myoclonic jerks, and survival. Treatment effects regarding neuroinflammation, neural damage, and neurodegeneration were subsequently analyzed by histology and immunohistochemistry. RESULTS: We show that PLX3397 treatment attenuates neuroinflammation in CLN1 mice by depleting pro-inflammatory microglia/macrophages. This leads to a reduction of T lymphocyte recruitment, an amelioration of axon damage and neuron loss in the retinotectal system, as well as reduced thinning of the inner retina and total brain atrophy. Accordingly, long-term treatment with the inhibitor also ameliorates clinical outcomes in CLN1 mice, such as impaired motor coordination, visual acuity, and myoclonic jerks. However, we detected a sex- and region-biased efficacy of CSF-1R inhibition, with male microglia/macrophages showing higher responsiveness toward depletion, especially in the gray matter of the CNS. This results in a better treatment outcome in male Ppt1-/- mice regarding some histopathological and clinical readouts and reflects heterogeneity of innate immune reactions in the diseased CNS. CONCLUSIONS: Our results demonstrate a detrimental impact of innate immune reactions in the CNS of CLN1 mice. These findings provide insights into CLN pathogenesis and may guide in the design of immunomodulatory treatment strategies.
Assuntos
Aminopiridinas/uso terapêutico , Encéfalo/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Microglia/efeitos dos fármacos , Lipofuscinoses Ceroides Neuronais/tratamento farmacológico , Pirróis/uso terapêutico , Aminopiridinas/farmacologia , Animais , Encéfalo/patologia , Modelos Animais de Doenças , Feminino , Macrófagos/patologia , Masculino , Camundongos , Microglia/patologia , Degeneração Neural/tratamento farmacológico , Degeneração Neural/imunologia , Degeneração Neural/patologia , Lipofuscinoses Ceroides Neuronais/imunologia , Lipofuscinoses Ceroides Neuronais/patologia , Neurônios/efeitos dos fármacos , Neurônios/patologia , Pirróis/farmacologia , Retina/efeitos dos fármacos , Retina/patologia , Fatores Sexuais , Linfócitos T/efeitos dos fármacos , Linfócitos T/patologia , Tomografia de Coerência ÓpticaRESUMO
Analysis of isolated meninges and cerebrospinal fluid (CSF) of post-mortem MS cases has shown increased gene and protein expression for the pro-inflammatory cytokines: tumour necrosis factor (TNF) and interferon-γ (IFNγ). Here we tested the hypothesis that persistent production of these cytokines in the meningeal compartment and diffusion into underlying GM can drive chronic MS-like GM pathology. Lentiviral transfer vectors were injected into the sagittal sulcus of DA rats to deliver continuous expression of TNF + IFNγ transgenes in the meninges and the resulting neuropathology analysed after 1 and 2 months. Injection of TNF + IFNγ viral vectors, with or without prior MOG immunisation, induced extensive immune cell infiltration (CD4+ and CD8+ T-cells, CD79a + B-cells and macrophages) in the meninges by 28 dpi, which remained at 2 months. Control GFP viral vector did not induce infiltration. Subpial demyelination was seen underlying these infiltrates, which was partly dependant on prior myelin oligodendrocyte glycoprotein (MOG) immunisation. A significant decrease in neuronal numbers was seen at 28 and 56 days in cortical layers II-V that was independent of MOG immunisation. RNA analysis at 28 dpi showed an increase in expression of necroptotic pathway genes, including RIP3, MLKL, cIAP2 and Nox2. PhosphoRIP3+ and phosphoMLKL+ neurons were present in TNF + IFNγ vector injected animals, indicating activation of necroptosis. Our results suggest that persistent expression of TNF in the presence of IFNγ is a potent inducer of meningeal inflammation and can activate TNF signalling pathways in cortical cells leading to neuronal death and subpial demyelination and thus may contribute to clinical progression in MS.
Assuntos
Doenças Desmielinizantes/metabolismo , Encefalomielite Autoimune Experimental/patologia , Interferon gama/metabolismo , Degeneração Neural/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Córtex Cerebral/imunologia , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Citocinas , Doenças Desmielinizantes/imunologia , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/metabolismo , Humanos , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/patologia , Interferon gama/imunologia , Meninges/imunologia , Meninges/metabolismo , Meninges/patologia , Esclerose Múltipla/imunologia , Esclerose Múltipla/metabolismo , Esclerose Múltipla/patologia , Degeneração Neural/imunologia , Ratos , Fator de Necrose Tumoral alfa/imunologiaRESUMO
Complement pathway overactivation can lead to neuronal damage in various neurological diseases. Although Alzheimer's disease (AD) is characterized by ß-amyloid plaques and tau tangles, previous work examining complement has largely focused on amyloidosis models. We find that glial cells show increased expression of classical complement components and the central component C3 in mouse models of amyloidosis (PS2APP) and more extensively tauopathy (TauP301S). Blocking complement function by deleting C3 rescues plaque-associated synapse loss in PS2APP mice and ameliorates neuron loss and brain atrophy in TauP301S mice, improving neurophysiological and behavioral measurements. In addition, C3 protein is elevated in AD patient brains, including at synapses, and levels and processing of C3 are increased in AD patient CSF and correlate with tau. These results demonstrate that complement activation contributes to neurodegeneration caused by tau pathology and suggest that blocking C3 function might be protective in AD and other tauopathies.
Assuntos
Doença de Alzheimer/imunologia , Amiloidose/imunologia , Complemento C3/metabolismo , Degeneração Neural/imunologia , Tauopatias/imunologia , Doença de Alzheimer/genética , Animais , Atrofia , Comportamento Animal , Biomarcadores/metabolismo , Encéfalo/patologia , Complemento C1q/metabolismo , Complemento C3/líquido cefalorraquidiano , Complemento C3/genética , Modelos Animais de Doenças , Feminino , Deleção de Genes , Regulação da Expressão Gênica , Humanos , Masculino , Camundongos Transgênicos , Degeneração Neural/genética , Neurônios/metabolismo , Neurônios/patologia , Placa Amiloide/metabolismo , Sinapses/metabolismoRESUMO
Neuroinflammation has been involved in pathogenesis of Parkinson's disease (PD), a chronic neurodegenerative disease characterized neuropathologically by progressive dopaminergic neuronal loss in the substantia nigra (SN). We recently have shown that helper T (Th)17 cells facilitate dopaminergic neuronal loss in vitro. Herein, we demonstrated that interleukin (IL)-17A, a proinflammatory cytokine produced mainly by Th17 cells, contributed to PD pathogenesis depending on microglia. Mouse and rat models for PD were prepared by intraperitoneal injection of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) or striatal injection of 1-methyl-4-phenylpyridinium (MPP+), respectively. Both in MPTP-treated mice and MPP+-treated rats, blood-brain barrier (BBB) was disrupted and IL-17A level increased in the SN but not in cortex. Effector T (Teff) cells that were adoptively transferred via tail veins infiltrated into the brain of PD mice but not into that of normal mice. The Teff cell transfer aggravated nigrostriatal dopaminergic neurodegeneration, microglial activation and motor impairment. Contrarily, IL-17A deficiency alleviated BBB disruption, dopaminergic neurodegeneration, microglial activation and motor impairment. Anti-IL-17A-neutralizing antibody that was injected into lateral cerebral ventricle in PD rats ameliorated the manifestations mentioned above. IL-17A activated microglia but did not directly affect dopaminergic neuronal survival in vitro. IL-17A exacerbated dopaminergic neuronal loss only in the presence of microglia, and silencing IL-17A receptor gene in microglia abolished the IL-17A effect. IL-17A-treated microglial medium that contained higher concentration of tumor necrosis factor (TNF)-α facilitated dopaminergic neuronal death. Further, TNF-α-neutralizing antibody attenuated MPP+-induced neurotoxicity. The findings suggest that IL-17A accelerates neurodegeneration in PD depending on microglial activation and at least partly TNF-α release.
Assuntos
Interleucina-17/imunologia , Microglia/imunologia , Doença de Parkinson/imunologia , 1-Metil-4-fenilpiridínio/farmacologia , Animais , Morte Celular/imunologia , Corpo Estriado/imunologia , Modelos Animais de Doenças , Dopamina/imunologia , Neurônios Dopaminérgicos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Degeneração Neural/imunologia , Degeneração Neural/patologia , Doenças Neurodegenerativas/imunologia , Doenças Neurodegenerativas/patologia , Neuroimunomodulação/imunologia , Ratos , Ratos Sprague-Dawley , Substância Negra/imunologia , Células Th17/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismoRESUMO
Purpose: Herpes simplex virus type-1 (HSV-1) is a leading cause of neurotrophic keratitis, characterized by decreased or absent corneal sensation due to damage to the sensory corneal innervation. We previously reported the elicited immune response to infection contributes to the mechanism of corneal nerve regression/damage during acute HSV-1 infection. Our aim is to further establish the involvement of infiltrated macrophages in the mechanism of nerve loss upon infection. Methods: Macrophage Fas-Induced Apoptosis (MAFIA) transgenic C57BL/6 mice were systemically treated with AP20187 dimerizer or vehicle (VEH), and their corneas, lymph nodes, and blood were assessed for CD45+CD11b+GFP+ cell depletion by flow cytometry (FC). Mice were ocularly infected with HSV-1 or left uninfected. At 2, 4, and/or 6 days post infection (PI), corneas were assessed for sensitivity and harvested for FC, nerve structure by immunohistochemistry, viral content by plaque assay, soluble factor content by suspension array, and activation of signaling pathways by Western blot analysis. C57BL6 mice were used to compare to the MAFIA mouse model. Results: MAFIA mice treated with AP20187 had efficient depletion of CD45+CD11b+GFP+ cells in the tissues analyzed. The reduction of CD45+CD11b+GFP+ cells recruited to the infected corneas of AP20187-treated mice correlated with preservation of corneal nerve structure and function, decreased protein concentration of inflammatory cytokines, and decreased STAT3 activation despite no changes in viral content in the cornea compared to VEH-treated animals. Conclusions: Our results suggest infiltrated macrophages are early effectors in the nerve regression following HSV-1 infection. We propose the neurodegeneration mechanism involves macrophages, local up-regulation of IL-6, and activation of STAT3.
Assuntos
Córnea/inervação , Herpesvirus Humano 1/crescimento & desenvolvimento , Ceratite Herpética/imunologia , Macrófagos/fisiologia , Degeneração Neural/imunologia , Receptor de Fator Estimulador de Colônias de Macrófagos/metabolismo , Doenças do Nervo Trigêmeo/imunologia , Animais , Western Blotting , Modelos Animais de Doenças , Citometria de Fluxo , Imuno-Histoquímica , Interleucina-6/metabolismo , Ceratite Herpética/patologia , Ceratite Herpética/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Degeneração Neural/patologia , Degeneração Neural/virologia , Fator de Transcrição STAT3/metabolismo , Tacrolimo/análogos & derivados , Tacrolimo/farmacologia , Nervo Trigêmeo/metabolismo , Doenças do Nervo Trigêmeo/patologia , Doenças do Nervo Trigêmeo/virologia , Ensaio de Placa ViralRESUMO
Inflammation arising from central and/or peripheral sources contributes to the pathogenesis of multiple neurodegenerative diseases including Parkinson's disease (PD). Emerging data suggest that differential activation of glia could lead to the pathogenesis and progression of PD. Here, we sought to determine the relationship between lipopolysaccharide (LPS) treatment, loss of dopaminergic neurons and differential activation of glia. Using a model of repeated injections with LPS (1mg/kg, i.p. for 4days), we found that LPS induced a 34% loss of dopamine neurons in the substantia nigra 19days after initiation of treatment, but no further cell loss was observed at 36days. LPS induced a strong pro-inflammatory response with increased mRNA expression of pro-inflammatory markers, including tumor necrosis factor-α (4.8-fold), inducible nitric oxide synthase (2.0-fold), interleukin-1 beta (8.9-fold), interleukin-6 (10.7-fold), and robust glial activation were observed at 1day after final dose of LPS. These pro-inflammatory genes were then reduced at 19days after treatment, when there was a rise in the anti-inflammatory genes Ym1 (1.8-fold) and arginase-1 (2.6-fold). Additionally, 36days after the last LPS injection there was a significant increase in interleukin-10 (2.1-fold) expression. The qPCR data results were supported by protein data, including cytokine measurements, western blotting, and immunofluorescence in brain microglia. Taken together, these data demonstrate that progressive neurodegeneration in the substantia nigra following LPS is likely arrested by microglia shifting to an anti-inflammatory phenotype. Thus, strategies to promote resolution of neuroinflammation may be a promising avenue to slow the progressive loss of dopamine neurons in PD.
Assuntos
Neurônios Dopaminérgicos/imunologia , Lipopolissacarídeos/toxicidade , Microglia/imunologia , Degeneração Neural/imunologia , Animais , Astrócitos/imunologia , Astrócitos/patologia , Morte Celular/fisiologia , Corpo Estriado/imunologia , Corpo Estriado/patologia , Citocinas/metabolismo , Progressão da Doença , Neurônios Dopaminérgicos/patologia , Inflamação/patologia , Inflamação/fisiopatologia , Masculino , Camundongos Endogâmicos C57BL , Microglia/patologia , Degeneração Neural/patologia , Doenças Neurodegenerativas/imunologia , Doenças Neurodegenerativas/patologia , Neuroimunomodulação/fisiologia , RNA Mensageiro/metabolismo , Distribuição Aleatória , Fatores de TempoRESUMO
OBJECTIVE: The etiological initiators of neuroinflammation remain inconclusive, and effective interventions to block neurodegeneration are unavailable. Surprisingly, we found collagen II-combined complete Freund's adjuvant (CC) that usually induces rheumatoid arthritis (RA) also drives Alzheimer's disease (AD)-like neurodegeneration in mice. CC not only upregulates the cerebral pro-inflammatory cytokines including tumor necrosis factor α (TNF-α) and interleukin 8 (IL-8), but also downregulates the cerebral interleukin 10 (IL-10), an anti-inflammatory cytokine, and tyrosine hydroxylase (TH), a ratelimiting enzyme for biosynthesis of the anti-inflammatory neurotransmitter dopamine. In contrast, electroacupuncture (EA) elevates TNF-α/IL-8 and declines IL-10/TH at first, but declines TNF-α/IL-8 and elevates IL-10/TH later. Upon impact on mitochondrial biogenesis, ubiquitination, and autophagy, EA firstly potentates but secondly attenuates CC-triggered signaling cascades leading to oxidation, nitrosylation, hypoxia, and angiogenesis. Eventually, EA compromises neurodegeneration by decreasing amyloid- ß peptide (Aß) and phosphorylated tau protein (p-tau), and also rectifies neuronal dysfunctions by increasing the cholinergic neurotransmitter acetylcholine (Ach) and its rate-limiting biosynthetic enzyme choline acetyltransferase (ChAT). RESULTS: Conclusively, EA initially aggravates and subsequently ameliorates CC-evoked AD-like earlyphase brain pathogenesis via conversion from pro-inflammatory microglia to anti-inflammatory microglia.
Assuntos
Doença de Alzheimer/imunologia , Doença de Alzheimer/terapia , Encéfalo/imunologia , Encéfalo/patologia , Eletroacupuntura , Doença de Alzheimer/patologia , Animais , Animais não Endogâmicos , Modelos Animais de Doenças , Feminino , Colágenos Associados a Fibrilas , Adjuvante de Freund , Masculino , Camundongos , Microglia/imunologia , Microglia/patologia , Degeneração Neural/imunologia , Degeneração Neural/patologia , Neuroimunomodulação/fisiologia , Neurônios/imunologia , Neurônios/patologia , Óxido Nítrico Sintase/metabolismo , Distribuição Aleatória , Transmissão Sináptica/fisiologiaRESUMO
Despite the recognized role of tumor necrosis factor (TNF) in inflammation and neuronal degeneration, anti-TNF therapeutics failed to treat neurodegenerative diseases. Animal disease models had revealed the antithetic effects of the two TNF receptors (TNFR) in the central nervous system, whereby TNFR1 has been associated with inflammatory degeneration and TNFR2 with neuroprotection. We here show the therapeutic potential of selective inhibition of TNFR1 and activation of TNFR2 by ATROSAB, a TNFR1-selective antagonistic antibody, and EHD2-scTNFR2, an agonistic TNFR2-selective TNF, respectively, in a mouse model of NMDA-induced acute neurodegeneration. Coadministration of either ATROSAB or EHD2-scTNFR2 into the magnocellular nucleus basalis significantly protected cholinergic neurons and their cortical projections against cell death, and reverted the neurodegeneration-associated memory impairment in a passive avoidance paradigm. Simultaneous blocking of TNFR1 and TNFR2 signaling, however, abrogated the therapeutic effect. Our results uncover an essential role of TNFR2 in neuroprotection. Accordingly, the therapeutic activity of ATROSAB is mediated by shifting the balance of the antithetic activity of endogenous TNF toward TNFR2, which appears essential for neuroprotection. Our data also explain earlier results showing that complete blocking of TNF activity by anti-TNF drugs was detrimental rather than protective and argue for the use of next-generation TNFR-selective TNF therapeutics as an effective approach in treating neurodegenerative diseases.
Assuntos
Inflamação/tratamento farmacológico , Degeneração Neural/tratamento farmacológico , Receptores Tipo II do Fator de Necrose Tumoral/genética , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Fator de Necrose Tumoral alfa/genética , Animais , Anticorpos/farmacologia , Núcleo Basal de Meynert/metabolismo , Núcleo Basal de Meynert/patologia , Proteínas de Transporte/genética , Proteínas de Transporte/imunologia , Morte Celular/efeitos dos fármacos , Neurônios Colinérgicos/efeitos dos fármacos , Neurônios Colinérgicos/patologia , Células HEK293 , Humanos , Inflamação/genética , Inflamação/patologia , Camundongos , N-Metilaspartato/genética , Degeneração Neural/induzido quimicamente , Degeneração Neural/genética , Degeneração Neural/imunologia , Receptores Tipo I de Fatores de Necrose Tumoral/antagonistas & inibidores , Receptores Tipo II do Fator de Necrose Tumoral/antagonistas & inibidores , Receptores Tipo II do Fator de Necrose Tumoral/imunologia , Fator de Necrose Tumoral alfa/antagonistas & inibidoresRESUMO
Poly(ADP-ribose), identified in 1966 independently by three groups Strassbourg, Kyoto and Tokyo, is synthesized by poly(ADP-ribose) polymerases (PARP) from NAD(+) as a substrate in the presence of Mg(2+). The structure was unique in that it has ribose-ribose linkage. In the early-1970s, however, its function in vivo/in vitro was still controversial and the antibody against it was desired to help clear its significance. Thereupon, the author tried to produce antibody against poly(ADP-ribose) in rabbits and succeeded in it for the first time in the world. Eventually, this success has led to the following two groundbreaking papers in Nature: "Naturally-occurring antibody against poly(ADP-ribose) in patients with autoimmune disease SLE", and "Induction of anti-poly(ADP-ribose) antibody by immunization with synthetic double-stranded RNA, poly(A)·poly(U)".On the way to the publication of the first paper, a reviewer gave me a friendly comment that there is "heteroclitic" fashion as a mechanism of the production of natural antibody. This comment was really a God-send for me, and became a train of power for publication of another paper, as described above. Accordingly, I thought this, I would say, episode is worth describing herein. Because of its importance in biomedical phenomena, a certain number of articles related to "heteroclitic" have become to be introduced in this review, although they were not always directly related to immuno-biological works on poly(ADP-ribose). Also, I tried to speculate on the future prospects of poly(ADP-ribose), product of PARP, as an immuno-regulatory molecule, including either induced or naturally-occurring antibodies, in view of "heteroclitic".
Assuntos
Poli Adenosina Difosfato Ribose/imunologia , Poli Adenosina Difosfato Ribose/uso terapêutico , Animais , Anticorpos/imunologia , Histonas/imunologia , Humanos , Lúpus Eritematoso Sistêmico/sangue , Lúpus Eritematoso Sistêmico/tratamento farmacológico , Lúpus Eritematoso Sistêmico/imunologia , Degeneração Neural/tratamento farmacológico , Degeneração Neural/imunologia , Poli Adenosina Difosfato Ribose/química , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Inibidores de Poli(ADP-Ribose) Polimerases/uso terapêuticoRESUMO
Immunological mediators that originate outside the nervous system can affect neuronal health. However, their roles in neurodegeneration remain largely unknown. Here, we show that the p38MAPK-mediated immune pathway activated in intestinal cells of Caenorhabditis elegans upon mitochondrial dysfunction protects neurons in a cell-non-autonomous fashion. Specifically, mitochondrial complex I dysfunction induced by rotenone activates the p38MAPK/CREB/ATF-7-dependent innate immune response pathway in intestinal cells of C. elegans. Activation of p38MAPK in the gut is neuroprotective. Enhancing the p38MAPK-mediated immune pathway in intestinal cells alone suppresses rotenone-induced dopaminergic neuron loss, while downregulating it in the intestine exacerbates neurodegeneration. The p38MAPK/ATF-7 immune pathway modulates autophagy and requires autophagy and the PTEN-induced putative kinase PINK-1 for conferring neuroprotection. Thus, mitochondrial damage induces the clearance of mitochondria by the immune pathway, protecting the organism from the toxic effects of mitochondrial dysfunction. We propose that mitochondria are subject to constant surveillance by innate immune mechanisms.
Assuntos
Fatores Ativadores da Transcrição/genética , Proteínas de Caenorhabditis elegans/genética , Caenorhabditis elegans/imunologia , Neurônios Dopaminérgicos/imunologia , Complexo I de Transporte de Elétrons/genética , Mitocôndrias/imunologia , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Fatores Ativadores da Transcrição/imunologia , Animais , Caenorhabditis elegans/efeitos dos fármacos , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/imunologia , Dopamina/metabolismo , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/patologia , Complexo I de Transporte de Elétrons/deficiência , Complexo I de Transporte de Elétrons/imunologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/imunologia , Células Epiteliais/patologia , Trato Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/patologia , Regulação da Expressão Gênica , Imunidade Inata , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/patologia , Mitofagia/efeitos dos fármacos , Mitofagia/genética , Degeneração Neural/induzido quimicamente , Degeneração Neural/genética , Degeneração Neural/imunologia , Rotenona/toxicidade , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno/imunologiaRESUMO
CONTEXT: Neuroimmunological response is associated with neurodegeneration in the human substantia nigra (SN) in Parkinson's disease (PD). OBJECTIVE: To explore the possibility that the neurotoxin, 6-hydroxydopamine (6-OHDA), could be used as a tool in mice to understand the immune response in PD. MATERIALS AND METHODS: We employed unilateral administration of 6-OHDA into the mouse SN. At 1 week, 2 weeks and 4 weeks post-injection, we used immunohistochemistry for the markers Iba-1 and gp91PHOX to investigate activated microglia in the SN. To examine the adaptive immune response, we used immunohistochemistry for CD3-positive T-lymphocytes, CD45R-positive B-lymphocytes and anti-mouse immunoglobulin-G (IgG). Dopamine neuron loss was examined using immunohistochemistry for the dopamine neuron marker, tyrosine hydroxylase. RESULTS: Compared to vehicle, 6-OHDA administration induced an intense IgG deposition in the SN as well as increased infiltration of both T- and B- lymphocytes into the injected side of the midbrain. The adaptive immune response was associated with extensive destruction of dopamine neurons and extensive microglial activation at every time point in the 6-OHDA groups. CONCLUSION: Our results suggest that 6-OHDA administration in mice can a potential tool for understanding mechanisms underlying adaptive immune activation-induced neurodegeneration in PD.
Assuntos
Imunidade Adaptativa/efeitos dos fármacos , Degeneração Neural/tratamento farmacológico , Oxidopamina/farmacologia , Transtornos Parkinsonianos/imunologia , Substância Negra/efeitos dos fármacos , Animais , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Complexo CD3/imunologia , Proteínas de Ligação ao Cálcio/metabolismo , Modelos Animais de Doenças , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/imunologia , Neurônios Dopaminérgicos/patologia , Imunoglobulina G/imunologia , Imuno-Histoquímica , Antígenos Comuns de Leucócito/imunologia , Masculino , Glicoproteínas de Membrana/metabolismo , Camundongos Endogâmicos C57BL , Proteínas dos Microfilamentos/metabolismo , Microglia/efeitos dos fármacos , Microglia/imunologia , Microglia/metabolismo , NADPH Oxidase 2 , NADPH Oxidases/metabolismo , Degeneração Neural/imunologia , Degeneração Neural/patologia , Transtornos Parkinsonianos/patologia , Substância Negra/imunologia , Substância Negra/patologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologiaRESUMO
OBJECTIVES: Paraneoplastic neurological syndromes (PNSs) are rare disorders in association with cancer and sub-divided into "classical" and "non-classical" syndromes according to a 2004 consensus paper proposed by a panel of PNS experts. "Classical" PNSs are regarded to account for the vast majority of cases. However, systematic reports on clinical PNS manifestations are rare. Therefore, we analyzed the spectrum of PNS in our clinic. METHODS: We retrospectively investigated medical records from consecutive patients diagnosed with definite PNS and serological evidence of well-characterized onconeural antibodies (anti-Hu, Yo, Ri, CV2/CRMP5, Ma1, Ma2, and amphiphysin) analyzed between 1991 and 2014 in our clinic. RESULTS: Of the 50 patients identified with onconeural antibody-positive PNS, 28 patients (56.0%) had "classical" PNS, and 22 (44.0%) "non-classical" PNS. Subacute cerebellar degeneration was the most frequent "classical" syndrome, brainstem encephalitis and subacute sensorimotor neuronopathy the most frequent "non-classical" syndromes. Anti-Hu antibodies were most frequent in both groups. 86.1% of patients developed neurological symptoms before the cancer was known. No differences between "classical" and "non-classical" syndromes were detected with respect to age, tumor entities and median time to diagnosis. However, whereas most patients with "classical" syndromes were females, there was no gender predominance in patients with "non-classical" PNS and the latter had significantly more frequent peripheral neurological syndromes. CONCLUSIONS: The so-called "non-classical" PNSs in association with well-characterized onconeural antibodies were more common in our patient population than expected. Therefore, in neurological disorders of unclear etiology with a subacute onset and atypical presentation further diagnostic work-up including investigation of onconeural antibodies is necessary.
Assuntos
Anticorpos Antineoplásicos/sangue , Síndromes Paraneoplásicas do Sistema Nervoso/etiologia , Síndromes Paraneoplásicas do Sistema Nervoso/imunologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Tronco Encefálico/imunologia , Doenças Cerebelares/imunologia , Proteínas ELAV/imunologia , Encefalite/imunologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Degeneração Neural/imunologia , Síndromes Paraneoplásicas do Sistema Nervoso/classificação , Síndromes Paraneoplásicas do Sistema Nervoso/fisiopatologia , Doenças do Sistema Nervoso Periférico/imunologia , Estudos RetrospectivosRESUMO
The paracaspase Malt1 is a central regulator of antigen receptor signaling that is frequently mutated in human lymphoma. As a scaffold, it assembles protein complexes for NF-κB activation, and its proteolytic domain cleaves negative NF-κB regulators for signal enforcement. Still, the physiological functions of Malt1-protease are unknown. We demonstrate that targeted Malt1-paracaspase inactivation induces a lethal inflammatory syndrome with lymphocyte-dependent neurodegeneration in vivo. Paracaspase activity is essential for regulatory T cell (Treg) and innate-like B cell development, but it is largely dispensable for overcoming Malt1-dependent thresholds for lymphocyte activation. In addition to NF-κB inhibitors, Malt1 cleaves an entire set of mRNA stability regulators, including Roquin-1, Roquin-2, and Regnase-1, and paracaspase inactivation results in excessive interferon gamma (IFNγ) production by effector lymphocytes that drive pathology. Together, our results reveal distinct threshold and modulatory functions of Malt1 that differentially control lymphocyte differentiation and activation pathways and demonstrate that selective paracaspase blockage skews systemic immunity toward destructive autoinflammation.
Assuntos
Autoimunidade , Caspases/metabolismo , Inflamação/imunologia , Inflamação/patologia , Proteínas de Neoplasias/metabolismo , Animais , Linfócitos B/imunologia , Caspases/deficiência , Diferenciação Celular/imunologia , Regulação da Expressão Gênica , Homeostase/imunologia , Humanos , Imunidade nas Mucosas/imunologia , Interferon gama/biossíntese , Ativação Linfocitária/imunologia , Camundongos Mutantes , Proteína de Translocação 1 do Linfoma de Tecido Linfoide Associado à Mucosa , Proteínas de Neoplasias/deficiência , Degeneração Neural/imunologia , Degeneração Neural/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Transdução de Sinais/imunologia , Linfócitos T Reguladores/imunologiaRESUMO
Multiple sclerosis (MS) is a chronic autoimmune disease characterized by inflammation, demyelination, and neurodegeneration in the CNS. Although it is important to prevent neurodegeneration for alleviating neurological disability, the molecular mechanism of neurodegeneration remains largely unknown. Here, we report that repulsive guidance molecule-a (RGMa), known to regulate axonal growth, is associated with neurodegeneration in experimental autoimmune encephalomyelitis (EAE), a mouse model of MS. RGMa is highly expressed in interleukin-17-producing CD4(+) T cells (Th17 cells). We induced EAE by adoptive transfer of myelin oligodendrocyte glycoprotein (MOG)-specific Th17 cells and then inhibited RGMa with a neutralizing antibody. Inhibition of RGMa improves EAE scores and reduces neuronal degeneration without altering immune or glial responses. Th17 cells induce cultured cortical neuron death through RGMa-neogenin and Akt dephosphorylation. Our results demonstrate that RGMa is involved in Th17-cell-mediated neurodegeneration and that RGMa-specific antibody may have a therapeutic effect in MS.
Assuntos
Encefalomielite Autoimune Experimental/complicações , Encefalomielite Autoimune Experimental/imunologia , Degeneração Neural/complicações , Degeneração Neural/imunologia , Proteínas do Tecido Nervoso/metabolismo , Células Th17/imunologia , Animais , Especificidade de Anticorpos/imunologia , Axônios/patologia , Adesão Celular , Morte Celular , Encefalomielite Autoimune Experimental/patologia , Proteínas Ligadas por GPI/antagonistas & inibidores , Proteínas Ligadas por GPI/metabolismo , Imunidade , Camundongos Endogâmicos C57BL , Degeneração Neural/patologia , Proteínas do Tecido Nervoso/antagonistas & inibidores , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Células Th17/metabolismoRESUMO
Multiple sclerosis is an inflammatory demyelinating disease affecting the central nervous system and considered one of the leading causes of disability in young adults. The precise cause of multiple sclerosis is unknown, although the current evidence points towards a combination of genetic and environmental factors leading to an autoimmune response that promotes neuronal degeneration. In this review, we will describe the association between the immune response and neurodegeneration in multiple sclerosis.
Assuntos
Imunidade Celular/imunologia , Esclerose Múltipla/imunologia , Degeneração Neural/imunologia , Linfócitos B/imunologia , Humanos , Inflamação/imunologia , Macrófagos/imunologia , Bainha de Mielina/imunologia , Doenças Neurodegenerativas/imunologia , Neuroglia/imunologia , Linfócitos T/imunologiaRESUMO
La esclerosis múltiple es una enfermedad inflamatoria desmielinizante que afecta el sistema nervioso central y que es considerada una de las principales causas de discapacidad en jóvenes adultos. Las causas de la esclerosis múltiple son aún desconocidas, aunque se cree que una combinación de factores genéticos y ambientales resulta en una respuesta autoinmune que promueve la degeneración neuronal/axonal. En esta revisión se analiza la asociación entre la respuesta inmune y la neurodegeneración en la esclerosis múltiple.
Multiple sclerosis is an inflammatory demyelinating disease affecting the central nervous system and considered one of the leading causes of disability in young adults. The precise cause of multiple sclerosis is unknown, although the current evidence points towards a combination of genetic and environmental factors leading to an autoimmune response that promotes neuronal degeneration. In this review, we will describe the association between the immune response and neurodegeneration in multiple sclerosis.
Assuntos
Humanos , Imunidade Celular/imunologia , Esclerose Múltipla/imunologia , Degeneração Neural/imunologia , Linfócitos B/imunologia , Inflamação/imunologia , Macrófagos/imunologia , Bainha de Mielina/imunologia , Doenças Neurodegenerativas/imunologia , Neuroglia/imunologia , Linfócitos T/imunologiaRESUMO
Sedentary lifestyle leads to the accumulation of visceral fat. This is accompanied by the infiltration of immune cells with pro-inflammatory characteristics in adipose tissue, causing an increased release of cytokines and generating a low-grade inflammatory state. It has been associated with the development of insulin resistance, atherosclerosis, neurodegeneration, and development of tumors. Exercise can be used as a treatment to improve symptoms of many of these conditions because it promotes an anti-inflammatory effect. In this review we analyze the pro-inflammatory factors present in obesity and the induction of antiinflammatory factors that occur with exercise.
La vida sedentaria induce la acumulación de grasa visceral, que se acompaña de la infiltración de células inmunitarias con características proinflamatorias en el tejido adiposo, que ocasiona mayor liberación de citocinas y genera un estado inflamatorio de bajo grado, éste se ha asociado con resistencia a la insulina, aterosclerosis, neurodegeneración y tumores. El ejercicio físico puede indicarse como tratamiento para disminuir los síntomas de muchas afecciones porque promueve un estado antiinflamatorio. En esta revisión se analizarán los factores proinflamatorios que coexisten en la obesidad, y la inducción de factores antiinflamatorios que se originan con el ejercicio físico.
Assuntos
Exercício Físico , Inflamação/etiologia , Obesidade/complicações , Comportamento Sedentário , Adipocinas/metabolismo , Aterosclerose/etiologia , Aterosclerose/imunologia , Aterosclerose/metabolismo , Doenças Cardiovasculares/etiologia , Doenças Cardiovasculares/imunologia , Doenças Cardiovasculares/metabolismo , Citocinas/metabolismo , Diabetes Mellitus Tipo 2/etiologia , Diabetes Mellitus Tipo 2/imunologia , Diabetes Mellitus Tipo 2/metabolismo , Epinefrina/metabolismo , Exercício Físico/fisiologia , Terapia por Exercício , Humanos , Hidrocortisona/metabolismo , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/prevenção & controle , Resistência à Insulina , Interleucina-6/metabolismo , Gordura Intra-Abdominal/metabolismo , Macrófagos/metabolismo , Contração Muscular , Neoplasias/etiologia , Neoplasias/imunologia , Neoplasias/metabolismo , Degeneração Neural/etiologia , Degeneração Neural/imunologia , Degeneração Neural/metabolismo , Obesidade/imunologia , Obesidade/metabolismo , Estresse Oxidativo , Linfócitos T Reguladores/imunologia , Receptores Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/metabolismoRESUMO
OBJECT: In this report, the authors describe the pathological changes in the human trigeminal nerve after Gamma Knife radiosurgery. METHODS: Three trigeminal nerves of patients with multiple sclerosis (MS)-related trigeminal neuralgia (MSTN) after Gamma Knife radiosurgery and other ablative procedures were examined by a neuropathologist. These cases were compared with 3 patients with typical TN who underwent partial surgical rhizotomy following recurrent symptoms after gasserian injury procedures, as well as with autopsy specimens from patients with and without MSTN. RESULTS: The three irradiated MS-TN specimens exhibited axon loss, demyelination, myelin debris, and fibrosis. Mild lymphocytic infiltrate was present in all 3 samples from MS-TN patients. The nonirradiated trigeminal nerve samples were generally well myelinated with rare degenerating axons. The microscopic findings in trigeminal nerve autopsy specimens were normal in patients without TN, with MS but not TN, and MS-TN. CONCLUSIONS: The inflammation observed in MS-TN specimens collected following Gamma Knife radiosurgery has not previously been described in the literature. These data provide new insight into the changes that occur in trigeminal nerve following stereotactic radiosurgery.