Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
STAR Protoc ; 5(1): 102916, 2024 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-38451820

RESUMO

Apoptosis-associated speck-like protein containing a c-terminal caspase activation and recruitment domain (ASC) specks are elevated in the cerebrospinal fluid (CSF) of Alzheimer's disease and related dementias (AD/ADRDs) patients. Here, we present a flow cytometry protocol to quantify ASC specks. We describe steps for fluorescently labeling ASC specks using antibody technology, visualizing with imaging flow cytometry, and gating based on physical characteristics. CSF ASC specks levels positively correlate with phosphorylated tau (Thr181) and negatively correlate with amyloid ß ratio (42/40), thus serving as a neuroinflammatory biomarker for diagnosing AD/ADRDs. For complete details on the use and execution of this protocol, please refer to Jiang et al.1.


Assuntos
Peptídeos beta-Amiloides , Proteínas Adaptadoras de Sinalização CARD , Humanos , Citometria de Fluxo/métodos , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Peptídeos beta-Amiloides/metabolismo , Inflamassomos/metabolismo , Apoptose
2.
Biosensors (Basel) ; 13(2)2023 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-36831917

RESUMO

The identification of protein aggregates as biomarkers for neurodegeneration is an area of interest for disease diagnosis and treatment development. In this work, we present novel super luminescent conjugated polyelectrolyte molecules as ex vivo sensors for tau-paired helical filaments (PHFs) and amyloid-ß (Aß) plaques. We evaluated the use of two oligo-p-phenylene ethynylenes (OPEs), anionic OPE12- and cationic OPE24+, as stains for fibrillar protein pathology in brain sections of transgenic mouse (rTg4510) and rat (TgF344-AD) models of Alzheimer's disease (AD) tauopathy, and post-mortem brain sections from human frontotemporal dementia (FTD). OPE12- displayed selectivity for PHFs in fluorimetry assays and strong staining of neurofibrillary tangles (NFTs) in mouse and human brain tissue sections, while OPE24+ stained both NFTs and Aß plaques. Both OPEs stained the brain sections with limited background or non-specific staining. This novel family of sensors outperformed the gold-standard dye Thioflavin T in sensing capacities and co-stained with conventional phosphorylated tau (AT180) and Aß (4G8) antibodies. As the OPEs readily bind protein amyloids in vitro and ex vivo, they are selective and rapid tools for identifying proteopathic inclusions relevant to AD. Such OPEs can be useful in understanding pathogenesis and in creating in vivo diagnostically relevant detection tools for neurodegenerative diseases.


Assuntos
Doença de Alzheimer , Emaranhados Neurofibrilares , Camundongos , Humanos , Ratos , Animais , Emaranhados Neurofibrilares/metabolismo , Emaranhados Neurofibrilares/patologia , Placa Amiloide , Proteínas tau , Doença de Alzheimer/diagnóstico , Encéfalo/metabolismo , Peptídeos beta-Amiloides , Coloração e Rotulagem , Etilenos/metabolismo
3.
Commun Biol ; 5(1): 125, 2022 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-35149761

RESUMO

With increased research funding for Alzheimer's disease (AD) and related disorders across the globe, large amounts of data are being generated. Several studies employed machine learning methods to understand the ever-growing omics data to enhance early diagnosis, map complex disease networks, or uncover potential drug targets. We describe results based on a Target Central Resource Database protein knowledge graph and evidence paths transformed into vectors by metapath matching. We extracted features between specific genes and diseases, then trained and optimized our model using XGBoost, termed MPxgb(AD). To determine our MPxgb(AD) prediction performance, we examined the top twenty predicted genes through an experimental screening pipeline. Our analysis identified potential AD risk genes: FRRS1, CTRAM, SCGB3A1, FAM92B/CIBAR2, and TMEFF2. FRRS1 and FAM92B are considered dark genes, while CTRAM, SCGB3A1, and TMEFF2 are connected to TREM2-TYROBP, IL-1ß-TNFα, and MTOR-APP AD-risk nodes, suggesting relevance to the pathogenesis of AD.


Assuntos
Doença de Alzheimer , Doença de Alzheimer/diagnóstico , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Diagnóstico Precoce , Humanos , Aprendizado de Máquina , Proteínas de Membrana/metabolismo , Proteínas de Neoplasias
4.
Cell Rep ; 36(12): 109720, 2021 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-34551296

RESUMO

Pathological hyperphosphorylation and aggregation of tau (pTau) and neuroinflammation, driven by interleukin-1ß (IL-1ß), are the major hallmarks of tauopathies. Here, we show that pTau primes and activates IL-1ß. First, RNA-sequence analysis suggests paired-helical filaments (PHFs) from human tauopathy brain primes nuclear factor κB (NF-κB), chemokine, and IL-1ß signaling clusters in human primary microglia. Treating microglia with pTau-containing neuronal media, exosomes, or PHFs causes IL-1ß activation, which is NLRP3, ASC, and caspase-1 dependent. Suppression of pTau or ASC reduces tau pathology and inflammasome activation in rTg4510 and hTau mice, respectively. Although the deletion of MyD88 prevents both IL-1ß expression and activation in the hTau mouse model of tauopathy, ASC deficiency in myeloid cells reduces pTau-induced IL-1ß activation and improves cognitive function in hTau mice. Finally, pTau burden co-exists with elevated IL-1ß and ASC in autopsy brains of human tauopathies. Together, our results suggest pTau activates IL-1ß via MyD88- and NLRP3-ASC-dependent pathways in myeloid cells, including microglia.


Assuntos
Inflamassomos/metabolismo , Interleucina-1beta/metabolismo , Transdução de Sinais , Tauopatias/patologia , Proteínas tau/metabolismo , Animais , Proteínas Adaptadoras de Sinalização CARD/genética , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Caspase 1/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Doxorrubicina/farmacologia , Humanos , Interleucina-1beta/genética , Camundongos , Camundongos Endogâmicos C57BL , Microglia/citologia , Microglia/metabolismo , Células Mieloides/citologia , Células Mieloides/metabolismo , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , NF-kappa B/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Tauopatias/metabolismo , Proteínas tau/genética
5.
J Neuroinflammation ; 18(1): 161, 2021 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-34275478

RESUMO

BACKGROUND: The presence of hyperphosphorylated microtubule-associated protein tau is strongly correlated with cognitive decline and neuroinflammation in Alzheimer's disease and related tauopathies. However, the role of inflammation and anti-inflammatory interventions in tauopathies is unclear. Our goal was to determine if removing anti-inflammatory interleukin-10 (IL-10) during an acute inflammatory challenge has any effect on neuronal tau pathology. METHODS: We induce systemic inflammation in Il10-deficient (Il10-/-) versus Il10+/+ (Non-Tg) control mice using a single intraperitoneal (i.p.) injection of lipopolysaccharide (LPS) to examine microglial activation and abnormal hyperphosphorylation of endogenous mouse tau protein. Tau phosphorylation was quantified by Western blotting and immunohistochemistry. Microglial morphology was quantified by skeleton analysis. Cytokine expression was determined by multiplex electro chemiluminescent immunoassay (MECI) from Meso Scale Discovery (MSD). RESULTS: Our findings show that genetic deletion of Il10 promotes enhanced neuroinflammation and tau phosphorylation. First, LPS-induced tau hyperphosphorylation was significantly increased in Il10-/- mice compared to controls. Second, LPS-treated Il10-/- mice showed signs of neurodegeneration. Third, LPS-treated Il10-/- mice showed robust IL-6 upregulation and direct treatment of primary neurons with IL-6 resulted in tau hyperphosphorylation on Ser396/Ser404 site. CONCLUSIONS: These data support that loss of IL-10 activates microglia, enhances IL-6, and leads to hyperphosphorylation of tau on AD-relevant epitopes in response to acute systemic inflammation.


Assuntos
Inflamação/metabolismo , Interleucina-10/deficiência , Interleucina-10/metabolismo , Tauopatias/metabolismo , Tauopatias/patologia , Proteínas tau/metabolismo , Animais , Técnicas de Cultura de Células , Citocinas/metabolismo , Modelos Animais de Doenças , Imuno-Histoquímica , Interleucina-6/metabolismo , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microglia/efeitos dos fármacos , Microglia/metabolismo , Microglia/patologia , Neurônios/metabolismo , Fosforilação/efeitos dos fármacos
6.
Front Mol Neurosci ; 13: 586731, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33177989

RESUMO

Tauopathies are a class of neurodegenerative diseases, including Alzheimer's disease (AD), Frontotemporal Dementia (FTD), Progressive Supranuclear Palsy (PSP), Corticobasal Degeneration (CBD), and many others where microtubule-associated protein tau (MAPT or tau) is hyperphosphorylated and aggregated to form insoluble paired helical filaments (PHFs) and ultimately neurofibrillary tangles (NFTs). Autophagic-endolysosomal networks (AELN) play important roles in tau clearance. Excessive soluble neurotoxic forms of tau and tau hyperphosphorylated at specific sites are cleared through the ubiquitin-proteasome system (UPS), Chaperon-mediated Autophagy (CMA), and endosomal microautophagy (e-MI). On the other hand, intra-neuronal insoluble tau aggregates are often degraded within lysosomes by macroautophagy. AELN defects have been observed in AD, FTD, CBD, and PSP, and lysosomal dysfunction was shown to promote the cleavage and neurotoxicity of tau. Moreover, several AD risk genes (e.g., PICALM, GRN, and BIN1) have been associated with dysregulation of AELN in the late-onset sporadic AD. Conversely, tau dissociation from microtubules interferes with retrograde transport of autophagosomes to lysosomes, and that tau fragments can also lead to lysosomal dysfunction. Recent studies suggest that tau is not merely an intra-neuronal protein, but it can be released to brain parenchyma via extracellular vesicles, like exosomes and ectosomes, and thus spread between neurons. Extracellular tau can also be taken up by microglial cells and astrocytes, either being degraded through AELN or propagated via exosomes. This article reviews the complex roles of AELN in the degradation and transmission of tau, potential diagnostic/therapeutic targets and strategies based on AELN-mediated tau clearance and propagation, and the current state of drug development targeting AELN and tau against tauopathies.

7.
NPJ Vaccines ; 4: 26, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31231552

RESUMO

Tauopathies, including frontotemporal dementia (FTD) and Alzheimer's disease (AD) are progressive neurodegenerative diseases clinically characterized by cognitive decline and could be caused by the aggregation of hyperphosphorylated pathological tau (pTau) as neurofibrillary tangles (NFTs) inside neurons. There is currently no FDA-approved treatment that cures, slows or prevents tauopathies. Current immunotherapy strategies targeting pTau have generated encouraging data but may pose concerns about scalability, affordability, and efficacy. Here, we engineered a virus-like particle (VLP)-based vaccine in which tau peptide, phosphorylated at threonine 181, was linked at high valency to Qß bacteriophage VLPs (pT181-Qß). We demonstrate that vaccination with pT181-Qß is sufficient to induce a robust and long-lived anti-pT181 antibody response in the sera and the brains of both Non-Tg and rTg4510 mice. Only sera from pT181-Qß vaccinated mice are reactive to classical somatodendritic pTau in human FTD and AD post-mortem brain sections. Finally, we demonstrate that pT181-Qß vaccination reduces both soluble and insoluble species of hyperphosphorylated pTau in the hippocampus and cortex, avoids a Th1-mediated pro-inflammatory cell response, prevents hippocampal and corpus callosum atrophy and rescues cognitive dysfunction in a 4-month-old rTg4510 mouse model of FTD. These studies provide a valid scientific premise for the development of VLP-based immunotherapy to target pTau and potentially prevent Alzheimer's diseases and related tauopathies.

8.
Front Mol Neurosci ; 10: 69, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28367114

RESUMO

Increasing evidence suggests that hyperphosphorylation and aggregation of microtubule-associated protein tau (MAPT or tau) correlates with the development of cognitive impairment in Alzheimer's disease (AD) and related tauopathies. While numerous attempts have been made to model AD-relevant tau pathology in various animal models, there has been very limited success for these models to fully recapitulate the progression of disease as seen in human tauopathies. Here, we performed whole genome gene expression in a genomic mouse model of tauopathy that expressed human MAPT gene under the control of endogenous human MAPT promoter and also were complete knockout for endogenous mouse tau [referred to as 'hTau MaptKO(Duke)' mice]. First, whole genome expression analysis revealed 64 genes, which were differentially expressed (32 up-regulated and 32 down-regulated) in the hippocampus of 6-month-old hTau MaptKO(Duke) mice compared to age-matched non-transgenic controls. Genes relevant to neuronal function or neurological disease include up-regulated genes: PKC-alpha (Prkca), MECP2 (Mecp2), STRN4 (Strn4), SLC40a1 (Slc40a1), POLD2 (Pold2), PCSK2 (Pcsk2), and down-regulated genes: KRT12 (Krt12), LASS1 (Cers1), PLAT (Plat), and NRXN1 (Nrxn1). Second, network analysis suggested anatomical structure development, cellular metabolic process, cell death, signal transduction, and stress response were significantly altered biological processes in the hTau MaptKO(Duke) mice as compared to age-matched non-transgenic controls. Further characterization of a sub-group of significantly altered genes revealed elevated phosphorylation of MECP2 (methyl-CpG-binding protein-2), which binds to methylated CpGs and associates with chromatin, in hTau MaptKO(Duke) mice compared to age-matched controls. Third, phoshpho-MECP2 was elevated in autopsy brain samples from human AD compared to healthy controls. Finally, siRNA-mediated knockdown of MECP2 in human tau expressing N2a cells resulted in a significant decrease in total and phosphorylated tau. Together, these results suggest that MECP2 is a potential novel regulator of tau pathology relevant to AD and tauopathies.

9.
Autophagy ; 13(6): 1084-1085, 2017 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-28368721

RESUMO

Macroautophagy/autophagy plays a role in unconventional secretion of leaderless cytosolic proteins. Whether and how secretory autophagy diverges from conventional degradative autophagy is unclear. We have shown that the prototypical secretory autophagy cargo IL1B/IL-1ß (interleukin 1 ß) is recognized by TRIM16, and that this first to be identified secretory autophagy receptor interacts with the R-SNARE SEC22B to jointly deliver cargo to the MAP1LC3B-II-positive sequestration membranes. Cargo secretion is unaffected by knockdowns of STX17, a SNARE catalyzing autophagosome-lysosome fusion as a prelude to cargo degradation. Instead, SEC22B in combination with plasma membrane syntaxins completes cargo secretion. Thus, secretory autophagy diverges from degradative autophagy by using specialized receptors and a dedicated SNARE machinery to bypass fusion with lysosomes.


Assuntos
Autofagia , Via Secretória , Humanos , Lisossomos/metabolismo , Fusão de Membrana , Modelos Biológicos , Fagossomos/metabolismo , Proteínas SNARE/metabolismo
10.
J Alzheimers Dis ; 57(4): 1123-1135, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28372329

RESUMO

Alzheimer's disease (AD) is the most common form of dementia affecting nearly 45 million people worldwide. However, the etiology of AD is still unclear. Accumulations of amyloid-ß plaques and tau tangles, neuroinflammation, and synaptic and neuronal loss are the major neuropathological hallmarks of AD, with synaptic loss being the strongest correlating factor with memory and cognitive impairment in AD. Many of these pathological hallmarks influence each other during the onset and progression of the disease. Recent genetic evidence suggests the possibility of a causal link between altered immune pathways and synaptic dysfunction in AD. Emerging studies also suggest that immune system-mediated synaptic pruning could initiate early-stage pathogenesis of AD. This comprehensive review is toward understanding the crosstalk of neuron-microglia-astrocyte and dynamics of complement, cytokine, and chemokine systems in the regulation of synaptic function and dysfunction relevant to AD. We start with summarizing several immune pathways, involving complements, MHC-I and CX3CL1, which mediate synaptic elimination during development and in AD. We then will discuss the potential of targeting these molecules as therapeutic interventions or as biomarkers for AD.


Assuntos
Doença de Alzheimer/imunologia , Proteínas do Sistema Complemento/metabolismo , Citocinas/metabolismo , Sinapses/metabolismo , Animais , Humanos
11.
EMBO J ; 36(1): 42-60, 2017 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-27932448

RESUMO

Autophagy is a process delivering cytoplasmic components to lysosomes for degradation. Autophagy may, however, play a role in unconventional secretion of leaderless cytosolic proteins. How secretory autophagy diverges from degradative autophagy remains unclear. Here we show that in response to lysosomal damage, the prototypical cytosolic secretory autophagy cargo IL-1ß is recognized by specialized secretory autophagy cargo receptor TRIM16 and that this receptor interacts with the R-SNARE Sec22b to recruit cargo to the LC3-II+ sequestration membranes. Cargo secretion is unaffected by downregulation of syntaxin 17, a SNARE promoting autophagosome-lysosome fusion and cargo degradation. Instead, Sec22b in combination with plasma membrane syntaxin 3 and syntaxin 4 as well as SNAP-23 and SNAP-29 completes cargo secretion. Thus, secretory autophagy utilizes a specialized cytosolic cargo receptor and a dedicated SNARE system. Other unconventionally secreted cargo, such as ferritin, is secreted via the same pathway.


Assuntos
Autofagia , Proteínas de Ligação a DNA/metabolismo , Interleucina-1beta/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas R-SNARE/metabolismo , Fatores de Transcrição/metabolismo , Linhagem Celular , Ferritinas/metabolismo , Humanos , Monócitos/metabolismo , Proteínas Qa-SNARE/metabolismo , Proteínas Qb-SNARE/metabolismo , Proteínas Qc-SNARE/metabolismo , Proteínas com Motivo Tripartido , Ubiquitina-Proteína Ligases
12.
Alzheimers Res Ther ; 8(1): 54, 2016 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-27974048

RESUMO

BACKGROUND: Hyperphosphorylation and aggregation of tau protein are the pathological hallmarks of Alzheimer's disease and related tauopathies. We previously demonstrated that the microglial activation induces tau hyperphosphorylation and cognitive impairment via activation of p38 mitogen-activated protein kinase (p38 MAPK) in the hTau mouse model of tauopathy that was deficient for microglial fractalkine receptor CX3CR1. METHOD: We report an isoform-selective, brain-permeable, and orally bioavailable small molecule inhibitor of p38α MAPK (MW181) and its effects on tau phosphorylation in vitro and in hTau mice. RESULTS: First, pretreatment of mouse primary cortical neurons with MW181 completely blocked inflammation-induced p38α MAPK activation and AT8 (pS199/pS202) site tau phosphorylation, with the maximum effect peaking at 60-90 min after stimulation. Second, treatment of old (~20 months of age) hTau mice with MW181 (1 mg/kg body weight; 14 days via oral gavage) significantly reduced p38α MAPK activation compared with vehicle-administered hTau mice. This also resulted in a significant reduction in AT180 (pT231) site tau phosphorylation and Sarkosyl-insoluble tau aggregates. Third, MW181 treatment significantly increased synaptophysin protein expression and resulted in improved working memory. Fourth, MW181 administration reduced phosphorylated MAPK-activated protein kinase 2 (pMK2) and phosphorylated activating transcription factor 2 (pATF2), which are known substrates of p38α MAPK. Finally, MW181 reduced the expression of interferon-γ and interleukin-1ß. CONCLUSIONS: Taken together, these studies support p38α MAPK as a valid therapeutic target for the treatment of tauopathies.


Assuntos
Fator 2 Ativador da Transcrição/efeitos dos fármacos , Interferon gama/efeitos dos fármacos , Interleucina-1beta/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular/efeitos dos fármacos , Memória de Curto Prazo/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/efeitos dos fármacos , Piridazinas/farmacologia , Piridinas/farmacologia , Tauopatias/tratamento farmacológico , Proteínas Quinases p38 Ativadas por Mitógeno/efeitos dos fármacos , Proteínas tau/metabolismo , Animais , Comportamento Animal , Córtex Cerebral/efeitos dos fármacos , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Inibidores de Proteínas Quinases/administração & dosagem , Piridazinas/administração & dosagem , Piridinas/administração & dosagem , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas tau/efeitos dos fármacos
13.
Nat Commun ; 6: 8620, 2015 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-26503418

RESUMO

Autophagy is a conserved homeostatic process active in all human cells and affecting a spectrum of diseases. Here we use a pharmaceutical screen to discover new mechanisms for activation of autophagy. We identify a subset of pharmaceuticals inducing autophagic flux with effects in diverse cellular systems modelling specific stages of several human diseases such as HIV transmission and hyperphosphorylated tau accumulation in Alzheimer's disease. One drug, flubendazole, is a potent inducer of autophagy initiation and flux by affecting acetylated and dynamic microtubules in a reciprocal way. Disruption of dynamic microtubules by flubendazole results in mTOR deactivation and dissociation from lysosomes leading to TFEB (transcription factor EB) nuclear translocation and activation of autophagy. By inducing microtubule acetylation, flubendazole activates JNK1 leading to Bcl-2 phosphorylation, causing release of Beclin1 from Bcl-2-Beclin1 complexes for autophagy induction, thus uncovering a new approach to inducing autophagic flux that may be applicable in disease treatment.


Assuntos
Doença de Alzheimer/fisiopatologia , Autofagia/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Bibliotecas de Moléculas Pequenas/farmacologia , Acetilação/efeitos dos fármacos , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Humanos , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Mebendazol/análogos & derivados , Mebendazol/farmacologia , Microtúbulos/efeitos dos fármacos , Microtúbulos/metabolismo
14.
Brain ; 138(Pt 6): 1738-55, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25833819

RESUMO

Pathological aggregation of tau is a hallmark of Alzheimer's disease and related tauopathies. We have previously shown that the deficiency of the microglial fractalkine receptor (CX3CR1) led to the acceleration of tau pathology and memory impairment in an hTau mouse model of tauopathy. Here, we show that microglia drive tau pathology in a cell-autonomous manner. First, tau hyperphosphorylation and aggregation occur as early as 2 months of age in hTauCx3cr1(-/-) mice. Second, CD45(+) microglial activation correlates with the spatial memory deficit and spread of tau pathology in the anatomically connected regions of the hippocampus. Third, adoptive transfer of purified microglia derived from hTauCx3cr1(-/-) mice induces tau hyperphosphorylation within the brains of non-transgenic recipient mice. Finally, inclusion of interleukin 1 receptor antagonist (Kineret®) in the adoptive transfer inoculum significantly reduces microglia-induced tau pathology. Together, our results suggest that reactive microglia are sufficient to drive tau pathology and correlate with the spread of pathological tau in the brain.


Assuntos
Encéfalo/metabolismo , Transtornos da Memória/metabolismo , Microglia/metabolismo , Tauopatias/metabolismo , Tauopatias/patologia , Proteínas tau/metabolismo , Animais , Encéfalo/patologia , Receptor 1 de Quimiocina CX3C , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Proteína Antagonista do Receptor de Interleucina 1/uso terapêutico , Transtornos da Memória/complicações , Transtornos da Memória/patologia , Camundongos , Camundongos Knockout , Fosforilação/genética , Cultura Primária de Células , Agregados Proteicos/genética , Receptores de Quimiocinas/deficiência , Receptores de Quimiocinas/genética , Tauopatias/complicações , Tauopatias/tratamento farmacológico , Tauopatias/genética , Proteínas tau/genética
15.
Curr Opin Microbiol ; 16(3): 355-65, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23790398

RESUMO

The now well-accepted innate immunity paradigm that autophagy acts as a cell-autonomous defense against intracellular bacteria has its key origins in studies with Mycobacterium tuberculosis, an important human pathogen and a model microorganism infecting macrophages. A number of different factors have been identified that play into the anti-mycobacterial functions of autophagy, and recent in vivo studies in the mouse model of tuberculosis have uncovered additional anti-inflammatory and tissue-sparing functions of autophagy. Complementing these observations, genome wide association studies indicate a considerable overlap between autophagy, human susceptibility to mycobacterial infections and predisposition loci for inflammatory bowel disease. Finally, recent studies show that autophagy is an important regulator and effector of IL-1 responses, and that autophagy intersects with type I interferon pathology-modulating responses.


Assuntos
Autofagia , Interações Hospedeiro-Patógeno , Imunidade Inata , Mycobacterium tuberculosis/imunologia , Tuberculose/imunologia , Tuberculose/microbiologia , Animais , Modelos Animais de Doenças , Humanos , Camundongos
16.
J Innate Immun ; 5(5): 471-9, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23445716

RESUMO

Autophagy (macroautophagy) is often defined as a degradative process and a tributary of the lysosomal pathway. In this context, autophagy carries out cytoplasmic quality control and nutritional functions by removing defunct or disused organelles, particulate targets and invading microbes, and by bulk digestion of the cytoplasm. However, recent studies indicate that autophagy surprisingly affects multiple secretory pathways. Autophagy participates in extracellular delivery of a number of cytosolic proteins that do not enter the conventional secretory pathway via the Golgi apparatus but are instead unconventionally secreted directly from the cytosol. In mammalian cells, a prototypical example of this manifestation of autophagy is the unconventional secretion of a major proinflammatory cytokine, IL-1ß. This review examines the concept of secretory autophagy and compares and contrasts the role of autophagy in the secretion of IL-1α and IL-1ß. Although IL-1α and IL-1ß have closely related extracellular inflammatory functions, they differ in intracellular activation, secretory mechanisms and how they are affected by autophagy. This example indicates that the role of autophagy in secretion is more complex, at least in mammalian cells, than the simplistic view that autophagosomes provide carriers for unconventional secretion of cytosolic proteins.


Assuntos
Autofagia/fisiologia , Proteólise , Via Secretória , Animais , Comunicação Celular , Humanos , Mediadores da Inflamação/metabolismo , Interleucina-1/metabolismo , Mamíferos
17.
Proc Natl Acad Sci U S A ; 109(46): E3168-76, 2012 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-23093667

RESUMO

Autophagy is a cell biological pathway affecting immune responses. In vitro, autophagy acts as a cell-autonomous defense against Mycobacterium tuberculosis, but its role in vivo is unknown. Here we show that autophagy plays a dual role against tuberculosis: antibacterial and anti-inflammatory. M. tuberculosis infection of Atg5(fl/fl) LysM-Cre(+) mice relative to autophagy-proficient littermates resulted in increased bacillary burden and excessive pulmonary inflammation characterized by neutrophil infiltration and IL-17 response with increased IL-1α levels. Macrophages from uninfected Atg5(fl/fl) LysM-Cre(+) mice displayed a cell-autonomous IL-1α hypersecretion phenotype, whereas T cells showed propensity toward IL-17 polarization during nonspecific activation or upon restimulation with mycobacterial antigens. Thus, autophagy acts in vivo by suppressing both M. tuberculosis growth and damaging inflammation.


Assuntos
Autofagia/imunologia , Proteínas Associadas aos Microtúbulos/imunologia , Mycobacterium tuberculosis/imunologia , Tuberculose/imunologia , Animais , Autofagia/genética , Proteína 5 Relacionada à Autofagia , Interleucina-17/imunologia , Interleucina-1alfa/genética , Interleucina-1alfa/imunologia , Macrófagos/imunologia , Camundongos , Camundongos Transgênicos , Proteínas Associadas aos Microtúbulos/genética , Infiltração de Neutrófilos/genética , Infiltração de Neutrófilos/imunologia , Neutrófilos/imunologia , Neutrófilos/microbiologia , Linfócitos T/imunologia , Linfócitos T/microbiologia , Tuberculose/genética , Tuberculose/microbiologia
18.
Immunity ; 37(2): 223-34, 2012 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-22921120

RESUMO

Autophagy is a fundamental biological process of the eukaryotic cell contributing to diverse cellular and physiological functions including cell-autonomous defense against intracellular pathogens. Here, we screened the Rab family of membrane trafficking regulators for effects on autophagic elimination of Mycobacterium tuberculosis var. bovis BCG and found that Rab8b and its downstream interacting partner, innate immunity regulator TBK-1, are required for autophagic elimination of mycobacteria in macrophages. TBK-1 was necessary for autophagic maturation. TBK-1 coordinated assembly and function of the autophagic machinery and phosphorylated the autophagic adaptor p62 (sequestosome 1) on Ser-403, a residue essential for its role in autophagic clearance. A key proinflammatory cytokine, IL-1ß, induced autophagy leading to autophagic killing of mycobacteria in macrophages, and this IL-1ß activity was dependent on TBK-1. Thus, TBK-1 is a key regulator of immunological autophagy and is responsible for the maturation of autophagosomes into lytic bactericidal organelles.


Assuntos
Autofagia/imunologia , Macrófagos/imunologia , Proteínas Serina-Treonina Quinases/imunologia , Proteínas rab de Ligação ao GTP/imunologia , Proteínas Adaptadoras de Transdução de Sinal/imunologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Proteína 7 Relacionada à Autofagia , Proteínas de Fluorescência Verde , Células HeLa , Proteínas de Choque Térmico/imunologia , Proteínas de Choque Térmico/metabolismo , Humanos , Interleucina-1beta/imunologia , Interleucina-1beta/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Microscopia Confocal , Proteínas Associadas aos Microtúbulos/antagonistas & inibidores , Proteínas Associadas aos Microtúbulos/genética , Mycobacterium bovis/imunologia , Fagossomos/efeitos dos fármacos , Fagossomos/imunologia , Fagossomos/metabolismo , Fosforilação , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , RNA Interferente Pequeno , Proteína Sequestossoma-1 , Serina/imunologia , Serina/metabolismo , Tuberculose/imunologia , Proteínas rab de Ligação ao GTP/genética
19.
Trends Cell Biol ; 22(8): 397-406, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22677446

RESUMO

Autophagy is a cell biological process ubiquitous to all eukaryotic cells, often referred to as a catabolic, lysosomal degradative pathway. However, current studies in mammalian systems suggest that autophagy plays an unexpectedly broad biogenesis role in protein trafficking and secretion. Autophagy supports alternative trafficking pathways for delivery of integral membrane proteins to the plasma membrane and affects secretion, including the constitutive, regulated and unconventional secretion pathways. Autophagy-based unconventional secretion, termed here 'autosecretion', is one of the pathways enabling leaderless cytosolic proteins to exit the cell without entering the endoplasmic reticulum (ER)-to-Golgi secretory pathway. In this review, we discuss the emerging underlying mechanisms of how autophagy affects different facets of secretion. We also describe the physiological roles of autosecretory cargos that are often associated with inflammatory processes and also play a role in the formation of specialized tissues and in tissue remodeling, expanding the immediate sphere of influence of autophagy from the intracellular to the extracellular space.


Assuntos
Autofagia , Animais , Membrana Celular/metabolismo , Retículo Endoplasmático/metabolismo , Complexo de Golgi/metabolismo , Humanos , Inflamação/metabolismo , Transporte Proteico
20.
EMBO J ; 30(23): 4701-11, 2011 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-22068051

RESUMO

Autophagy controls the quality and quantity of the eukaryotic cytoplasm while performing two evolutionarily highly conserved functions: cell-autonomous provision of energy and nutrients by cytosol autodigestion during starvation, and removal of defunct organelles and large aggregates exceeding the capacity of other cellular degradative systems. In contrast to these autodigestive processes, autophagy in yeast has additional, biogenesis functions. However, no equivalent biosynthetic roles have been described for autophagy in mammals. Here, we show that in mammalian cells, autophagy has a hitherto unappreciated positive contribution to the biogenesis and secretion of the proinflammatory cytokine IL-1ß via an export pathway that depends on Atg5, inflammasome, at least one of the two mammalian Golgi reassembly stacking protein (GRASP) paralogues, GRASP55 (GORASP2) and Rab8a. This process, which is a type of unconventional secretion, expands the functional manifestations of autophagy beyond autodigestive and quality control roles in mammals. It enables a subset of cytosolic proteins devoid of signal peptide sequences, and thus unable to access the conventional pathway through the ER, to enter an autophagy-based secretory pathway facilitating their exit from the cytoplasm.


Assuntos
Autofagia/fisiologia , Proteínas de Transporte/metabolismo , Interleucina-1beta , Proteínas de Membrana/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , Animais , Autofagia/efeitos dos fármacos , Proteína 5 Relacionada à Autofagia , Células Cultivadas , Proteína HMGB1/metabolismo , Inflamassomos/metabolismo , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas Luminescentes/metabolismo , Macrolídeos/farmacologia , Macrófagos/metabolismo , Camundongos , Nigericina/farmacologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Via Secretória/efeitos dos fármacos , Via Secretória/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...