Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 122
Filtrar
1.
Front Immunol ; 14: 1130735, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37033925

RESUMO

Diabetic retinopathy, a microvascular disease characterized by irreparable vascular damage, neurodegeneration and neuroinflammation, is a leading complication of diabetes mellitus. There is no cure for DR, and medical interventions marginally slow the progression of disease. Microglia-mediated inflammation in the diabetic retina is regulated via CX3CR1-FKN signaling, where FKN serves as a calming signal for microglial activation in several neuroinflammatory models. Polymorphic variants of CX3CR1, hCX3CR1I249/M280 , found in 25% of the human population, result in a receptor with lower binding affinity for FKN. Furthermore, disrupted CX3CR1-FKN signaling in CX3CR1-KO and FKN-KO mice leads to exacerbated microglial activation, robust neuronal cell loss and substantial vascular damage in the diabetic retina. Thus, studies to characterize the effects of hCX3CR1I249/M280 -expression in microglia-mediated inflammation in the diseased retina are relevant to identify mechanisms by which microglia contribute to disease progression. Our results show that hCX3CR1I249/M280 mice are significantly more susceptible to microgliosis and production of Cxcl10 and TNFα under acute inflammatory conditions. Inflammation is exacerbated under diabetic conditions and coincides with robust neuronal loss in comparison to CX3CR1-WT mice. Therefore, to further investigate the role of hCX3CR1I249/M280 -expression in microglial responses, we pharmacologically depleted microglia using PLX-5622, a CSF-1R antagonist. PLX-5622 treatment led to a robust (~70%) reduction in Iba1+ microglia in all non-diabetic and diabetic mice. CSF-1R antagonism in diabetic CX3CR1-WT prevented TUJ1+ axonal loss, angiogenesis and fibrinogen deposition. In contrast, PLX-5622 microglia depletion in CX3CR1-KO and hCX3CR1I249/M280 mice did not alleviate TUJ1+ axonal loss or angiogenesis. Interestingly, PLX-5622 treatment reduced fibrinogen deposition in CX3CR1-KO mice but not in hCX3CR1I249/M280 mice, suggesting that hCX3CR1I249/M280 expressing microglia influences vascular pathology differently compared to CX3CR1-KO microglia. Currently CX3CR1-KO mice are the most commonly used strain to investigate CX3CR1-FKN signaling effects on microglia-mediated inflammation and the results in this study indicate that hCX3CR1I249/M280 receptor variants may serve as a complementary model to study dysregulated CX3CR1-FKN signaling. In summary, the protective effects of microglia depletion is CX3CR1-dependent as microglia depletion in CX3CR1-KO and hCX3CR1I249/M280 mice did not alleviate retinal degeneration nor microglial morphological activation as observed in CX3CR1-WT mice.


Assuntos
Diabetes Mellitus Experimental , Microglia , Humanos , Camundongos , Animais , Microglia/metabolismo , Receptor 1 de Quimiocina CX3C/genética , Receptor 1 de Quimiocina CX3C/metabolismo , Diabetes Mellitus Experimental/patologia , Inflamação/metabolismo , Retina/patologia , Proteínas de Transporte/metabolismo , Fibrinogênio/metabolismo
2.
Artigo em Inglês | MEDLINE | ID: mdl-34667128

RESUMO

BACKGROUND AND OBJECTIVES: To evaluate the pathophysiology of neuromyelitis optica spectrum disorder (NMOSD) and the therapeutic mechanism and levels of interleukin-6 (IL-6) blockade (satralizumab), especially with respect to blood-brain barrier (BBB) disruption with the new in vitro and ex vivo human BBB models and in vivo model. METHODS: We constructed new static in vitro and flow-based ex vivo models for evaluating continued barrier function, leukocyte transmigration, and intracerebral transferability of neuromyelitis optica-immunoglobulin G (NMO-IgG) and satralizumab across the BBB using the newly established triple coculture system that are specialized to closely mimic endothelial cell contact of pericytes and endfeet of astrocytes. In the in vivo study, we assessed the effects of an anti-IL-6 receptor antibody for mice (MR16-1) on in vivo BBB disruption in mice with experimental autoimmune encephalomyelitis in which IL-6 concentration in the spinal cord dramatically increases. RESULTS: In vitro and ex vivo experiments demonstrated that NMO-IgG increased intracerebral transferability of satralizumab and NMO-IgG and that satralizumab suppressed the NMO-IgG-induced transmigration of T cells and barrier dysfunction. In the in vivo study, the blockade of IL-6 signaling suppressed the migration of T cells into the spinal cord and prevented the increased BBB permeability. DISCUSSION: These results suggest that (1) our triple-cultured in vitro and in ex vivo BBB models are ideal for evaluating barrier function, leukocyte transmigration, and intracerebral transferability; (2) NMO-IgG increased the intracerebral transferability of NMO-IgG via decreasing barrier function and induced secretion of IL-6 from astrocytes causing more dysfunction of the barrier and disrupting controlled cellular infiltration; and (3) satralizumab, which can pass through the BBB in the presence of NMO-IgG, suppresses the BBB dysfunction and the infiltration of inflammatory cells, leading to prevention of onset of NMOSD.


Assuntos
Anticorpos Bloqueadores/farmacologia , Autoanticorpos/farmacologia , Barreira Hematoencefálica , Encefalomielite Autoimune Experimental/imunologia , Interleucina-6/imunologia , Neuromielite Óptica , Animais , Anticorpos Monoclonais Humanizados/farmacologia , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/fisiopatologia , Células Cultivadas , Técnicas de Cocultura , Modelos Animais de Doenças , Feminino , Humanos , Imunoglobulina G , Camundongos , Camundongos Endogâmicos C57BL , Neuromielite Óptica/imunologia , Neuromielite Óptica/prevenção & controle
3.
Mol Neurodegener ; 15(1): 44, 2020 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-32727516

RESUMO

BACKGROUND: Identified as an Alzheimer's disease (AD) susceptibility gene by genome wide-association studies, BIN1 has 10 isoforms that are expressed in the Central Nervous System (CNS). The distribution of these isoforms in different cell types, as well as their role in AD pathology still remains unclear. METHODS: Utilizing antibodies targeting specific BIN1 epitopes in human post-mortem tissue and analyzing mRNA expression data from purified microglia, we identified three isoforms expressed in neurons and astrocytes (isoforms 1, 2 and 3) and four isoforms expressed in microglia (isoforms 6, 9, 10 and 12). The abundance of selected peptides, which correspond to groups of BIN1 protein isoforms, was measured in dorsolateral prefrontal cortex, and their relation to neuropathological features of AD was assessed. RESULTS: Peptides contained in exon 7 of BIN1's N-BAR domain were found to be significantly associated with AD-related traits and, particularly, tau tangles. Decreased expression of BIN1 isoforms containing exon 7 is associated with greater accumulation of tangles and subsequent cognitive decline, with astrocytic rather than neuronal BIN1 being the more likely culprit. These effects are independent of the BIN1 AD risk variant. CONCLUSIONS: Exploring the molecular mechanisms of specific BIN1 isoforms expressed by astrocytes may open new avenues for modulating the accumulation of Tau pathology in AD.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Astrócitos/metabolismo , Microglia/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Proteínas tau/metabolismo , Doença de Alzheimer/metabolismo , Encéfalo/metabolismo , Humanos , Neurônios/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
4.
PLoS One ; 14(8): e0220125, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31408457

RESUMO

BIN1 is the most important risk locus for Late Onset Alzheimer's Disease (LOAD), after ApoE. BIN1 AD-associated SNPs correlate with Tau deposition as well as with brain atrophy. Furthermore, the level of neuronal-specific BIN1 isoform 1 protein is decreased in sporadic AD cases in parallel with neuronal loss, despite an overall increase in BIN1 total mRNA. To address the relationship between reduction of BIN1 and neuronal cell loss in the context of Tau pathology, we knocked-down endogenous murine Bin1 via stereotaxic injection of AAV-Bin1 shRNA in the hippocampus of mice expressing Tau P301S (PS19). We observed a statistically significant reduction in the number of neurons in the hippocampus of mice injected with AAV-Bin1 shRNA in comparison with mice injected with AAV control. To investigate whether neuronal loss is due to deletion of Bin1 selectively in neurons in presence Tau P301S, we bred Bin1flox/flox with Thy1-Cre and subsequently with PS19 mice. Mice lacking neuronal Bin1 and expressing Tau P301S showed increased mortality, without increased neuropathology, when compared to neuronal Bin1 and Tau P301S-expressing mice. The loss of Bin1 isoform 1 resulted in reduced excitability in primary neurons in vitro, reduced neuronal c-fos expression as well as in altered microglia transcriptome in vivo. Taken together, our data suggest that the contribution of genetic variation in BIN1 locus to AD risk could result from a cell-autonomous reduction of neuronal excitability due to Bin1 decrease, exacerbated by the presence of aggregated Tau, coupled with a non-cell autonomous microglia activation.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Doença de Alzheimer/patologia , Biomarcadores/metabolismo , Encéfalo/fisiopatologia , Modelos Animais de Doenças , Hipocampo/fisiopatologia , Proteínas do Tecido Nervoso/fisiologia , Neurônios/patologia , Proteínas Supressoras de Tumor/fisiologia , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Animais , Comportamento Animal , Encéfalo/metabolismo , Feminino , Perfilação da Expressão Gênica , Hipocampo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/metabolismo , Ratos , Proteínas tau/metabolismo
5.
Sci Rep ; 9(1): 9477, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31263146

RESUMO

Despite Bridging INtegrator 1 (BIN1) being the second most statistically-significant locus associated to Late Onset Alzheimer's Disease, its role in disease pathogenesis remains to be clarified. As reports suggest a link between BIN1, Tau and extracellular vesicles, we investigated whether BIN1 could affect Tau spreading via exosomes secretion. We observed that BIN1-associated Tau-containing extracellular vesicles purified from cerebrospinal fluid of AD-affected individuals are seeding-competent. We showed that BIN1 over-expression promotes the release of Tau via extracellular vesicles in vitro as well as exacerbation of Tau pathology in vivo in PS19 mice. Genetic deletion of Bin1 from microglia resulted in reduction of Tau secretion via extracellular vesicles in vitro, and in decrease of Tau spreading in vivo in male, but not female, mice, in the context of PS19 background. Interestingly, ablation of Bin1 in microglia of male mice resulted in significant reduction in the expression of heat-shock proteins, previously implicated in Tau proteostasis. These observations suggest that BIN1 could contribute to the progression of AD-related Tau pathology by altering Tau clearance and promoting release of Tau-enriched extracellular vesicles by microglia.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/biossíntese , Doença de Alzheimer/metabolismo , Vesículas Extracelulares/metabolismo , Proteínas do Tecido Nervoso/biossíntese , Proteínas Nucleares/biossíntese , Proteínas Supressoras de Tumor/biossíntese , Proteínas tau/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Animais , Vesículas Extracelulares/genética , Vesículas Extracelulares/patologia , Feminino , Regulação da Expressão Gênica , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Microglia/metabolismo , Microglia/patologia , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Proteostase , Caracteres Sexuais , Proteínas Supressoras de Tumor/genética , Proteínas tau/genética
6.
Methods Mol Biol ; 1936: 23-36, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30820891

RESUMO

Remyelination is the regenerative process whereby myelin sheaths are restored around axons following nervous system injury, allowing reinstatement of electrical impulse conduction, trophic/metabolic support, and axon health. Failure of remyelination in progressive multiple sclerosis is considered to contribute to axon loss, a correlate of clinical decline. Lack of approved pro-regenerative therapies for MS highlights the need to understand the cellular and molecular mechanisms underpinning successful remyelination. One approach is to conduct nonbiased gene expression analyses of cell types which regulate remyelination, such as microglia and monocyte-derived macrophages. Recent technological advances address the challenges of RNA sequencing of small tissue samples, thus allowing relatively small numbers of cells to be isolated from discrete lesions for analysis. Here, we present methods for FACS-based isolation of cells from focal remyelinating lesions of the adult mouse brain and subsequent RNA extraction for sequencing, using isolation of microglia/macrophages as an example.


Assuntos
Encéfalo/citologia , Remielinização , Análise de Sequência de RNA/métodos , Animais , Separação Celular , Sistema Nervoso Central/química , Citometria de Fluxo , Regulação da Expressão Gênica , Macrófagos/química , Macrófagos/citologia , Camundongos , Microglia/química , Microglia/citologia
7.
Nat Commun ; 9(1): 2450, 2018 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-29934589

RESUMO

Inflammation is a characteristic feature of Parkinson's disease (PD). We examined the role of TLR9 and its regulation by glucocorticoid receptors (GRs) in degeneration of substantia nigra dopamine neurons (DNs). TLR9 agonist, CpG-ODN, induced DN degeneration in mice lacking GR in microglia but not in controls. TLR9 deletion reduced DN loss in neurotoxin, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of PD. GR regulates TLR9 activation during MPTP neurotoxicity as TLR9 antagonist suppressed increased DN loss in microglia/macrophage GR mutant mice. GR absence in microglia enhanced TLR9 translocation to endolysosomes and facilitated its cleavage leading to pro-inflammatory gene expression. GR-dependent TLR9 activation also triggered DN loss following intranigral injection of mitochondrial DNA. Finally, microglial GR sensitivity to A53T-alpha-synuclein induced DN degeneration as well as decreased microglial GR expression observed in SN of PD brain samples, all suggest that reduced microglial GR activity in SN can stimulate TLR9 activation and DN loss in PD pathology.


Assuntos
Microglia/metabolismo , Doença de Parkinson/etiologia , Receptores de Glucocorticoides/metabolismo , Substância Negra/metabolismo , Receptor Toll-Like 9/metabolismo , Idoso , Idoso de 80 Anos ou mais , Animais , Sobrevivência Celular , Cisteína Endopeptidases/metabolismo , DNA Mitocondrial/metabolismo , Neurônios Dopaminérgicos/fisiologia , Feminino , Humanos , Lisossomos/metabolismo , Masculino , Camundongos , Camundongos Knockout , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Substância Negra/patologia
8.
J Neuroinflammation ; 15(1): 26, 2018 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-29382353

RESUMO

BACKGROUND: Traumatic brain injury (TBI) is a critical public health and socio-economic problem worldwide. A growing body of evidence supports the involvement of inflammatory events in TBI. It has been reported that resident microglia and infiltrating monocytes promote an inflammatory reaction that leads to neuronal death and eventually behavioral and cognitive impairment. Currently, there is no effective treatment for TBI and the development of new therapeutic strategies is a scientific goal of highest priority. Laquinimod, an orally administered neuroimmunomodulator initially developed for the treatment of multiple sclerosis, might be a promising neuroprotective therapy for TBI. Herein, we aim to investigate the hypothesis that laquinimod will reduce the central nervous system (CNS) damage caused by TBI. METHODS: To test our hypothesis, Ccr2rfp/+ Cx3cr1 gfp/+ mice were submitted to a moderate TBI induced by fluid percussion. Sham controls were submitted only to craniotomy. Mice were treated daily by oral gavage with laquinimod (25 mg/kg) 7 days before and 3 days after TBI. The brains of mice treated or not treated with laquinimod were collected at 3 and 120 days post injury, and brain morphological changes, axonal injury, and neurogenesis were evaluated by microscopy analysis. We also isolated microglia from infiltrating monocytes, and the expression of immune gene mRNAs were analyzed by employing a quantitative NanoString nCounter technique. RESULTS: Laquinimod prevented ventricle enlargement caused by TBI in the long term. Immunohistochemical analyses revealed decreased axonal damage and restored neurogenesis in the laquinimod-treated TBI group at early stage (3 days post injury). Notably, laquinimod inhibited the monocytes infiltration to the brain. Hierarchial clustering demonstrated that the microglial gene expression from the TBI group treated with laquinimod resembles the sham group more than the TBI-water control group. CONCLUSIONS: Administration of laquinimod reduced lesion volume and axonal damage and restored neurogenesis after TBI. Laquinimod might be a potential therapy strategy to improve TBI long-term prognosis.


Assuntos
Lesões Encefálicas Traumáticas/tratamento farmacológico , Lesões Encefálicas Traumáticas/patologia , Modelos Animais de Doenças , Macrófagos/efeitos dos fármacos , Macrófagos/fisiologia , Quinolonas/uso terapêutico , Animais , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Inflamação/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Quinolonas/farmacologia
9.
J Neuroimmunol ; 317: 55-66, 2018 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-29325905

RESUMO

Acute skeletal muscle injury repair requires an adequate inflammatory response predominated by macrophage infiltration. We studied the activation of infiltrating macrophages by analyzing the expression of M1/M2 signature genes. Most of the intramuscular macrophages were Ly6Chi at day 1 after BaCl2 injection, while many were Ly6Clo at day 3. Ly6Chi macrophages at day 1 expressed a high level of both M1 and M2 genes, and the Ly6Chi and Ly6Clo macrophages at day 3 expressed a similar level of many M1/M2 genes. Infiltrating macrophages are broadly activated rather than polarized at the early stage to support acute skeletal muscle injury repair.


Assuntos
Ativação de Macrófagos/fisiologia , Macrófagos/fisiologia , Músculo Esquelético/lesões , Músculo Esquelético/fisiologia , Regeneração/fisiologia , Animais , Camundongos , Camundongos Endogâmicos C57BL
10.
J Crohns Colitis ; 12(2): 188-196, 2018 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-28961770

RESUMO

BACKGROUND AND AIMS: Progressive multifocal leukoencephalopathy [PML], a brain infection associated with anti-integrin drugs that inhibit lymphocyte translocation from bloodstream to tissue, can be fatal. Decreased central nervous system [CNS] immune surveillance leading to this infection has been reported in patients with multiple sclerosis or Crohn's disease treated with anti-integrin antibody natalizumab. PF-00547659 is an investigational human monoclonal antibody for inflammatory bowel disease, targeted against α4ß7-mucosal addressin cell-adhesion molecule-1 [the integrin ligand selectively expressed in the gut]. We hypothesised that this selective agent would not affect central nervous system immune surveillance. METHODS: Cerebrospinal fluid from five healthy volunteers, and from 10 patients with Crohn's disease previously treated with immunosuppressants, was evaluated to assess the feasibility of the study. Subsequently, 39 patients with active Crohn's disease and previous immunosuppression were evaluated over 12 weeks of PF-00547659-induction therapy. We measured total lymphocytes, T cell subsets in cerebrospinal fluid, and circulating ß7+ memory cells. Disease activity was assessed using the Harvey-Bradshaw Index. RESULTS: Patients treated with PF-00547659 had no reduction of cerebrospinal fluid lymphocytes, T-lymphocyte subsets, or CD4:CD8 ratio, whereas circulating ß7+ memory cells increased significantly. A total of 28/35 [80%] patients had a clinical response and 27/34 [79%] had disease remission. Treatment-related adverse events, none serious, were reported in 23/49 [47%] patients. CONCLUSIONS: In patients with active Crohn's disease, natalizumab therapy increases the risk for PML, and the increased risk is thought to be associated with iatrogenic leukopenia within the CNS. PML under PF-00547659 may be a lesser concern, as this agent did not reduce lymphocytes or T cell subsets in the cerebrospinal fluid.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Sistema Nervoso Central/imunologia , Doença de Crohn/imunologia , Cadeias beta de Integrinas/metabolismo , Linfócitos T/metabolismo , Adolescente , Adulto , Anticorpos Monoclonais Humanizados/farmacologia , Doença de Crohn/sangue , Doença de Crohn/líquido cefalorraquidiano , Feminino , Humanos , Vigilância Imunológica/efeitos dos fármacos , Imunossupressores/uso terapêutico , Leucoencefalopatia Multifocal Progressiva/induzido quimicamente , Contagem de Linfócitos , Masculino , Pessoa de Meia-Idade , Subpopulações de Linfócitos T , Adulto Jovem
11.
J Neurotrauma ; 35(1): 73-84, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-28859549

RESUMO

Traumatic brain injury (TBI) induces widespread neuroinflammation and accumulation of microtubule associated protein tau (MAPT): two key pathological features of tauopathies. This study sought to characterize the microglial/macrophage response to TBI in genomic-based MAPT transgenic mice in a Mapt knockout background (called hTau). Two-month-old hTau and age-matched control male and female mice received a single lateral fluid percussion TBI or sham injury. Separate groups of mice were aged to an acute (3 days post-injury [DPI]) or chronic (135 DPI) post-injury time point. As judged by tissue immunostaining for macrophage markers, microglial/macrophage response to TBI was enhanced at 3 DPI in hTau mice compared with control TBI and sham mice. However, MAPT phosphorylation increased in hTau mice regardless of injury group. Flow cytometric analysis revealed distinct populations of microglia and macrophages within all groups at 135 DPI. Unexpectedly, microglial reactivity was significantly reduced in hTau TBI mice compared with all other groups. Instead, hTau TBI mice showed a persistent macrophage response. In addition, TBI enhanced MAPT pathology in the temporal cortex and hippocampus of hTau TBI mice compared with controls 135 DPI. A battery of behavioral tests revealed that TBI in hTau mice resulted in compromised use of spatial search strategies to complete a water maze task, despite lack of motor or visual deficits. Collectively, these data indicate that the presence of wild-type human tau alters the microglial/macrophage response to a single TBI, induces delayed, region-specific MAPT pathology, and alters cognitive recovery; however, the causal relationship between these events remains unclear. These results highlight the potential significance of communication between MAPT and microglia/macrophages following TBI, and emphasize the role of neuroinflammation in post-injury recovery.


Assuntos
Lesões Encefálicas Traumáticas/complicações , Lesões Encefálicas Traumáticas/patologia , Macrófagos/patologia , Tauopatias/complicações , Tauopatias/patologia , Animais , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microglia/patologia , Proteínas tau/genética
12.
Cell Rep ; 21(2): 366-380, 2017 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-29020624

RESUMO

Microglia, the tissue-resident macrophages in the brain, are damage sensors that react to nearly any perturbation, including neurodegenerative diseases such as Alzheimer's disease (AD). Here, using single-cell RNA sequencing, we determined the transcriptome of more than 1,600 individual microglia cells isolated from the hippocampus of a mouse model of severe neurodegeneration with AD-like phenotypes and of control mice at multiple time points during progression of neurodegeneration. In this neurodegeneration model, we discovered two molecularly distinct reactive microglia phenotypes that are typified by modules of co-regulated type I and type II interferon response genes, respectively. Furthermore, our work identified previously unobserved heterogeneity in the response of microglia to neurodegeneration, discovered disease stage-specific microglia cell states, revealed the trajectory of cellular reprogramming of microglia in response to neurodegeneration, and uncovered the underlying transcriptional programs.


Assuntos
Doença de Alzheimer/metabolismo , Ativação de Macrófagos , Microglia/metabolismo , Transcriptoma , Animais , Encéfalo/citologia , Encéfalo/metabolismo , Células Cultivadas , Perfilação da Expressão Gênica , Interferon Tipo I/genética , Interferon gama/genética , Macrófagos/metabolismo , Camundongos , Microglia/citologia , Fenótipo , Análise de Célula Única
13.
Sci Rep ; 7(1): 4942, 2017 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-28694451

RESUMO

Microglia are the resident macrophages of the central nervous system and play complex roles in the milieu of diseases including the primary diseases of myelin. Although mitochondria are critical for cellular functions and survival in the nervous system, alterations in and the roles of mitochondrial dynamics and associated signaling in microglia are still poorly understood. In the present study, by combining immunohistochemistry and 3D ultrastructural analyses, we show that mitochondrial fission/fusion in reactive microglia is differentially regulated from that in monocyte-derived macrophages and the ramified microglia of normal white matter in myelin disease models. Mouse cerebral microglia in vitro demonstrated that stimulation of TLR4 with lipopolysaccharide, widely used to examine microglial reactions, caused the activation of the mitochondrial fission protein, dynamin-related protein 1 (Drp1) and enhanced production of reactive oxygen species (ROS). The increase in the ROS level activated 5' adenosine monophosphate-activated protein kinase (AMPK), and facilitated elongation of mitochondria along the microtubule tracks. These results suggest that the polymorphic regulation of mitochondrial fission and fusion in reactive microglia is mediated by distinct signaling under inflammatory conditions, and modulates microglial phenotypes through the production of ROS.


Assuntos
Microglia/metabolismo , Dinâmica Mitocondrial , Fenótipo , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Biomarcadores , Sistema Nervoso Central/imunologia , Sistema Nervoso Central/metabolismo , Doenças Desmielinizantes/etiologia , Doenças Desmielinizantes/metabolismo , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental , Inflamação/imunologia , Inflamação/metabolismo , Lipopolissacarídeos/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Mitocôndrias/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais
14.
Nat Commun ; 8: 15508, 2017 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-28561022

RESUMO

NOTCH1 signalling contributes to defective remyelination by impairing differentiation of oligodendrocyte progenitor cells (OPCs). Here we report that IL-17 stimulation induces NOTCH1 activation in OPCs, contributing to Th17-mediated demyelinating disease. Mechanistically, IL-17R interacts with NOTCH1 via the extracellular domain, which facilitates the cleavage of NOTHC1 intracellular domain (NICD1). IL-17-induced NOTCH1 activation results in the interaction of IL-17R adaptor Act1 with NICD1, followed by the translocation of the Act1-NICD1 complex into the nucleus. Act1-NICD1 are recruited to the promoters of several NOTCH1 target genes (including STEAP4, a metalloreductase important for inflammation and cell proliferation) that are specifically induced in the spinal cord by Th17 cells. A decoy peptide disrupting the IL-17RA-NOTCH1 interaction inhibits IL-17-induced NOTCH1 activation and attenuates Th17-mediated experimental autoimmune encephalitis (EAE). Taken together, these findings demonstrate critical crosstalk between the IL-17 and NOTCH1 pathway, regulating Th17-induced inflammatory and proliferative genes to promote demyelinating disease.


Assuntos
Encefalomielite Autoimune Experimental/imunologia , Interleucina-17/metabolismo , Esclerose Múltipla/imunologia , Células Precursoras de Oligodendrócitos/fisiologia , Receptor Notch1/imunologia , Células Th17/imunologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Astrócitos , Diferenciação Celular/imunologia , Proliferação de Células/fisiologia , Técnicas de Cocultura , Feminino , Células HEK293 , Células HeLa , Humanos , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/genética , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/imunologia , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/metabolismo , Interleucina-17/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Cultura Primária de Células , Ligação Proteica/imunologia , Domínios Proteicos/fisiologia , Receptor Notch1/genética , Receptor Notch1/metabolismo , Receptores de Interleucina-17/metabolismo , Remielinização/fisiologia , Transdução de Sinais/imunologia , Células Th1/imunologia , Células Th17/metabolismo , Peptídeos e Proteínas Associados a Receptores de Fatores de Necrose Tumoral/metabolismo
15.
Science ; 356(6344)2017 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-28546318

RESUMO

Microglia play essential roles in central nervous system (CNS) homeostasis and influence diverse aspects of neuronal function. However, the transcriptional mechanisms that specify human microglia phenotypes are largely unknown. We examined the transcriptomes and epigenetic landscapes of human microglia isolated from surgically resected brain tissue ex vivo and after transition to an in vitro environment. Transfer to a tissue culture environment resulted in rapid and extensive down-regulation of microglia-specific genes that were induced in primitive mouse macrophages after migration into the fetal brain. Substantial subsets of these genes exhibited altered expression in neurodegenerative and behavioral diseases and were associated with noncoding risk variants. These findings reveal an environment-dependent transcriptional network specifying microglia-specific programs of gene expression and facilitate efforts to understand the roles of microglia in human brain diseases.


Assuntos
Meio Ambiente , Redes Reguladoras de Genes/fisiologia , Microglia/citologia , Microglia/fisiologia , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/fisiopatologia , Células Cultivadas , Epilepsia/genética , Epilepsia/fisiopatologia , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL
16.
J Immunol ; 197(12): 4750-4761, 2016 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-27913649

RESUMO

We identified and characterized the function of CD45+/collagen I+ fibrocytes in acutely injured skeletal muscle of wild-type (WT) and Ccr2-/- mice, and in quadriceps and diaphragm muscles of mdx5cv mice, a mouse model for Duchenne muscular dystrophy. Fibrocytes were not detected in peripheral blood of WT mice after acute muscle injury or mdx5cv mice. Fibrocytes were detected in acutely injured muscles and in mdx5cv quadriceps and diaphragm muscles. These cells expressed F4/80 and CCR2, and they were mostly Ly6Clo They expressed a low level of collagens but a high level of profibrotic growth factors as compared with i.m. fibroblasts. Fibrocyte expression of collagens and profibrotic growth factors was not increased in Ccr2-/- mice as compared with WT controls. Fibrocyte expression of both proinflammatory and profibrotic cytokines was significantly higher in mdx5cv diaphragm than in mdx5cv quadriceps. In cocultures, fibrocytes from the mdx5cv diaphragm stimulated a higher level of fibroblast expression of extracellular matrix genes than did those from the mdx5cv quadriceps. Our findings suggest that i.m. fibrocytes most likely originate from infiltrating monocytes/macrophages and differentiate within injured muscles. They likely contribute to the normal muscle injury repair by producing growth factors. They do not appear to contribute to the persistent muscle fibrosis associated with poor injury repair in Ccr2-/- mice. However, they likely contribute to the persistent inflammation and progressive fibrosis in the mdx5cv diaphragm.


Assuntos
Diafragma/patologia , Células-Tronco Mesenquimais/fisiologia , Músculo Esquelético/patologia , Distrofia Muscular de Duchenne/patologia , Cicatrização , Doença Aguda , Animais , Antígenos de Diferenciação/metabolismo , Células Cultivadas , Técnicas de Cocultura , Colágeno Tipo I/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Fibrose , Humanos , Mediadores da Inflamação/metabolismo , Antígenos Comuns de Leucócito/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Camundongos Knockout , Músculo Esquelético/imunologia , Distrofia Muscular de Duchenne/imunologia , Receptores CCR2/genética , Receptores CCR2/metabolismo , Cicatrização/genética
17.
Nat Neurosci ; 19(8): 987-91, 2016 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-27459405

RESUMO

Microglial research has entered a fertile, dynamic phase characterized by novel technologies including two-photon imaging, whole-genome transcriptomic and epigenomic analysis with complementary bioinformatics, unbiased proteomics, cytometry by time of flight (CyTOF; Fluidigm) cytometry, and complex high-content experimental models including slice culture and zebrafish. Against this vivid background of newly emerging data, investigators will encounter in the microglial research literature a body of published work using the terminology of macrophage polarization, most commonly into the M1 and M2 phenotypes. It is the assertion of this opinion piece that microglial polarization has not been established by research findings. Rather, the adoption of this schema was undertaken in an attempt to simplify data interpretation at a time when the ontogeny and functional significance of microglia had not yet been characterized. Now, terminology suggesting established meaningful pathways of microglial polarization hinders rather than aids research progress and should be discarded.


Assuntos
Polaridade Celular/fisiologia , Citocinas/metabolismo , Macrófagos/metabolismo , Microglia/metabolismo , Animais , Polaridade Celular/efeitos dos fármacos , Humanos , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Microglia/efeitos dos fármacos , Fenótipo
18.
Stem Cells ; 34(8): 2026-39, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27145382

RESUMO

Shifting the balance away from tumor-mediated immune suppression toward tumor immune rejection is the conceptual foundation for a variety of immunotherapy efforts currently being tested. These efforts largely focus on activating antitumor immune responses but are confounded by multiple immune cell populations, including myeloid-derived suppressor cells (MDSCs), which serve to suppress immune system function. We have identified immune-suppressive MDSCs in the brains of GBM patients and found that they were in close proximity to self-renewing cancer stem cells (CSCs). MDSCs were selectively depleted using 5-flurouracil (5-FU) in a low-dose administration paradigm, which resulted in prolonged survival in a syngeneic mouse model of glioma. In coculture studies, patient-derived CSCs but not nonstem tumor cells selectively drove MDSC-mediated immune suppression. A cytokine screen revealed that CSCs secreted multiple factors that promoted this activity, including macrophage migration inhibitory factor (MIF), which was produced at high levels by CSCs. Addition of MIF increased production of the immune-suppressive enzyme arginase-1 in MDSCs in a CXCR2-dependent manner, whereas blocking MIF reduced arginase-1 production. Similarly to 5-FU, targeting tumor-derived MIF conferred a survival advantage to tumor-bearing animals and increased the cytotoxic T cell response within the tumor. Importantly, tumor cell proliferation, survival, and self-renewal were not impacted by MIF reduction, demonstrating that MIF is primarily an indirect promoter of GBM progression, working to suppress immune rejection by activating and protecting immune suppressive MDSCs within the GBM tumor microenvironment. Stem Cells 2016;34:2026-2039.


Assuntos
Neoplasias Encefálicas/imunologia , Glioblastoma/imunologia , Evasão da Resposta Imune , Fatores Inibidores da Migração de Macrófagos/metabolismo , Células Supressoras Mieloides/metabolismo , Células-Tronco Neoplásicas/metabolismo , Animais , Arginase/metabolismo , Neoplasias Encefálicas/patologia , Carcinogênese/metabolismo , Carcinogênese/patologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Meios de Cultivo Condicionados/farmacologia , Feminino , Glioblastoma/patologia , Humanos , Evasão da Resposta Imune/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Camundongos Nus , Células Supressoras Mieloides/efeitos dos fármacos , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/patologia , Microambiente Tumoral/efeitos dos fármacos
20.
Nat Immunol ; 17(5): 583-92, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26998763

RESUMO

Interleukin 1ß (IL-1ß) is critical for the in vivo survival, expansion and effector function of IL-17-producing helper T (T(H)17) cells during autoimmune responses, including experimental autoimmune encephalomyelitis (EAE). However, the spatiotemporal role and cellular source of IL-1ß during EAE pathogenesis are poorly defined. In the present study, we uncovered a T cell-intrinsic inflammasome that drives IL-1ß production during T(H)17-mediated EAE pathogenesis. Activation of T cell antigen receptors induced expression of pro-IL-1ß, whereas ATP stimulation triggered T cell production of IL-1ß via ASC-NLRP3-dependent caspase-8 activation. IL-1R was detected on T(H)17 cells but not on type 1 helper T (T(H)1) cells, and ATP-treated T(H)17 cells showed enhanced survival compared with ATP-treated T(H)1 cells, suggesting autocrine action of T(H)17-derived IL-1ß. Together these data reveal a critical role for IL-1ß produced by a T(H)17 cell-intrinsic ASC-NLRP3-caspase-8 inflammasome during inflammation of the central nervous system.


Assuntos
Proteínas Reguladoras de Apoptose/imunologia , Encefalomielite Autoimune Experimental/imunologia , Linfócitos T/imunologia , Células Th17/imunologia , Trifosfato de Adenosina/farmacologia , Animais , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Proteínas Adaptadoras de Sinalização CARD , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/imunologia , Proteínas de Transporte/metabolismo , Caspase 8/genética , Caspase 8/imunologia , Caspase 8/metabolismo , Sobrevivência Celular/genética , Sobrevivência Celular/imunologia , Células Cultivadas , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/metabolismo , Citometria de Fluxo , Expressão Gênica/imunologia , Immunoblotting , Inflamassomos/genética , Inflamassomos/imunologia , Inflamassomos/metabolismo , Interleucina-17/genética , Interleucina-17/imunologia , Interleucina-17/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/imunologia , Interleucina-1beta/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo , Células Th17/efeitos dos fármacos , Células Th17/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA