Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Stem Cell Res Ther ; 15(1): 258, 2024 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-39135132

RESUMO

BACKGROUND: Alzheimer disease (AD) is a heterogenous and multifactorial disease, and its pathology is partly driven by microglia and their activated phenotype. Brain organoids (BOs) are gaining prominence as a relevant model of the human brain for the study of AD; however, BOs are commonly devoid of microglia. To overcome this limitation, current protocols incorporate microglia through either (1) co-culture (BO co-culture), or (2) molecular manipulation at critical windows of BO development to have microglia arise innately (BO innate cultures). It is currently unclear whether the microglia incorporated into BOs by either of these two protocols differ in function. METHODS: At in vitro day 90, BO innate cultures and BO-co-cultures were challenged with the AD-related ß-amyloid peptide (Aß) for up to 72 h. After Aß challenge, BOs were collected for immunoblotting. Immunoblots compared immunodensity and protein banding of Aß and ionized calcium-binding adapter molecule 1 (IBA1, a marker of microglial activation) in BOs. The translational potential of these observations was supported using 56 human cortical samples from neurocognitively normal donors and patients with early-onset AD and late-onset AD. Statistical analyses were conducted using the Kruskal-Wallis test, a two-way ANOVA, or a simple linear regression, and where applicable, followed by Dunn's or Sidak's test. RESULTS: We show that BO co-cultures promote Aß oligomerization as early as 24 h and this coincides with a significant increase in IBA1 levels. In contrast, the Aßs do not oligomerize in BO innate cultures and the IBA1 response was modest and only emerged after 48 h. In human cortical samples, we found IBA1 levels correlated with age at onset, age at death, and the putative diagnostic Aß(1-42)/Aß(1-40) ratio (particularly in their oligomeric forms) in a sex-dependent manner. CONCLUSIONS: Our unique observations suggest that BOs with innate microglia model the response of a healthy brain to Aß, and by extension the initial stages of Aß challenge. It would be impossible to model these early stages of pathogenesis in BOs where microglia are already compromised, such as those with microglia incorporated by co-culture.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides , Encéfalo , Técnicas de Cocultura , Microglia , Organoides , Humanos , Microglia/metabolismo , Técnicas de Cocultura/métodos , Peptídeos beta-Amiloides/metabolismo , Organoides/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Feminino , Masculino , Idoso , Pessoa de Meia-Idade
2.
Front Cell Neurosci ; 18: 1383688, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38784709

RESUMO

Human brain organoids are emerging as translationally relevant models for the study of human brain health and disease. However, it remains to be shown whether human-specific protein processing is conserved in human brain organoids. Herein, we demonstrate that cell fate and composition of unguided brain organoids are dictated by culture conditions during embryoid body formation, and that culture conditions at this stage can be optimized to result in the presence of glia-associated proteins and neural network activity as early as three-months in vitro. Under these optimized conditions, unguided brain organoids generated from induced pluripotent stem cells (iPSCs) derived from male-female siblings are similar in growth rate, size, and total protein content, and exhibit minimal batch-to-batch variability in cell composition and metabolism. A comparison of neuronal, microglial, and macroglial (astrocyte and oligodendrocyte) markers reveals that profiles in these brain organoids are more similar to autopsied human cortical and cerebellar profiles than to those in mouse cortical samples, providing the first demonstration that human-specific protein processing is largely conserved in unguided brain organoids. Thus, our organoid protocol provides four major cell types that appear to process proteins in a manner very similar to the human brain, and they do so in half the time required by other protocols. This unique copy of the human brain and basic characteristics lay the foundation for future studies aiming to investigate human brain-specific protein patterning (e.g., isoforms, splice variants) as well as modulate glial and neuronal processes in an in situ-like environment.

3.
Mol Cell Neurosci ; 124: 103804, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36592800

RESUMO

Cardiolipin is a mitochondrial phospholipid that is also detected in serum inferring its extracellular release; however, this process has not been directly demonstrated for any of the brain cell types. Nevertheless, extracellular cardiolipin has been shown to modulate several neuroimmune functions of microglia and astrocytes, including upregulation of their endocytic activity. Low cardiolipin levels are associated with brain aging, and may thus hinder uptake of amyloid-ß (Αß) in Alzheimer's disease. We hypothesized that glial cells are one of the sources of extracellular cardiolipin in the brain parenchyma where this phospholipid interacts with neighboring cells to upregulate the endocytosis of Αß. Liquid chromatography-mass spectrophotometry identified 31 different species of cardiolipin released from murine BV-2 microglial cells and revealed this process was accelerated by exposure to Aß42. Extracellular cardiolipin upregulated internalization of fluorescently-labeled Aß42 by primary murine astrocytes, human U118 MG astrocytic cells, and murine BV-2 microglia. Increased endocytic activity in the presence of extracellular cardiolipin was also demonstrated by studying uptake of Aß42 and pHrodo™ Bioparticles™ by human induced pluripotent stem cells (iPSCs)-derived microglia, as well as iPSC-derived human brain organoids containing microglia, astrocytes, oligodendrocytes and neurons. Our observations indicate that Aß42 augments the release of cardiolipin from microglia into the extracellular space, where it can act on microglia and astrocytes to enhance their endocytosis of Aß42. Our observations suggest that the reduced glial uptake of Aß due to the decreased levels of cardiolipin could be at least partially responsible for the extracellular accumulation of Aß in aging and Alzheimer's disease.


Assuntos
Doença de Alzheimer , Células-Tronco Pluripotentes Induzidas , Humanos , Animais , Camundongos , Microglia/metabolismo , Cardiolipinas/metabolismo , Doença de Alzheimer/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Neuroglia/metabolismo , Peptídeos beta-Amiloides/metabolismo , Astrócitos/metabolismo
4.
Methods Mol Biol ; 2558: 143-161, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36169861

RESUMO

The influence of a protein is not determined exclusively by its level of expression, but also by its localization within the cell. The literature often refers to the enzyme monoamine oxidase (MAO) as a mitochondrial enzyme, yet there is evidence that mitochondria-independent pools of MAO exist. These pools of MAO could exert distinct influences across physiological as well as pathological phenotypes. Fluorescence microscopy is a powerful tool for spatially resolving target proteins in cell and tissue preparations. This can rely on an antibody-based probe that targets the endogenous protein, e.g., immunofluorescence. In the event that antibodies might not be readily available or if one is interested in characterizing a variant of the wild-type protein, then a recombinant protein with a fluorescent fusion "tag" is preferred. We now describe a protocol for the detection of endogenous MAO using indirect immunofluorescence and a version of the protocol with minor modification for detecting (green) fluorescent protein-tagged MAOs. One observation we can highlight using these easily adaptable approaches is that MAO A and MAO B do not follow similar patterns of distribution throughout the cell, suggesting potential expression of MAO A and MAO B on distinct pools of mitochondria. Furthermore, distinct subcellular compartmentalization is suggested by the fact that a pool of MAO A, but not MAO B, is associated with certain lysosomal compartments. However, directed and quantitative studies will be required before any definitive statement can be made on these intriguing possibilities.


Assuntos
Mitocôndrias , Monoaminoxidase , Imunofluorescência , Mitocôndrias/metabolismo , Monoaminoxidase/metabolismo , Proteínas Recombinantes/metabolismo , Coloração e Rotulagem
5.
J Neurochem ; 164(5): 560-582, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36517959

RESUMO

Brain organoids have the potential to improve clinical translation, with the added benefit of reducing any extraneous use of experimental animals. As brain organoids are three-dimensional in vitro constructs that emulate the human brain, they bridge in vitro and in vivo studies more appropriately than monocultures. Although many factors contribute to the failure of extrapolating monoculture-based information to animal-based experiments and clinical trials, for the purpose of this review, we will focus on glia (non-neuronal brain cells), whose functions and transcriptome are particularly abnormal in monocultures. As discussed herein, glia require signals from-and contact with-other cell types to exist in their homeostatic state, which likely contributes to some of the differences between data derived from monocultures and data derived from brain organoids and even two-dimensional co-cultures. Furthermore, we highlight transcriptomic differences between humans and mice in regard to aging and Alzheimer's disease, emphasizing need for a model using the human genome-again, a benefit of brain organoids-to complement data derived from animals. We also identify an urgency for guidelines to improve the reporting and transparency of research using organoids. The lack of reporting standards creates challenges for the comparison and discussion of data from different articles. Importantly, brain organoids mark the first human model enabling the study of brain cytoarchitecture and development.


Assuntos
Doença de Alzheimer , Neuroquímica , Humanos , Animais , Camundongos , Microglia , Encéfalo/fisiologia , Organoides/metabolismo , Doença de Alzheimer/metabolismo
6.
Mediators Inflamm ; 2022: 9946439, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35369030

RESUMO

Alzheimer's disease (AD) is characterized by chronic neuroinflammation, which is partially mediated by dysregulated functions of glial cells. Cardiolipin (CL) is a phospholipid normally confined to the inner mitochondrial membrane; however, it has been detected in human sera, indicating that it can exist in the extracellular space where it may interact with nearby cells. Although CL has been shown to modulate several functions of microglia in a toll-like receptor (TLR) 4-dependent manner, the effects of extracellular CL on astrocytes are unknown. In addition to their homeostatic functions, astrocytes participate in neuroimmune responses of the brain and express TLR 4. Therefore, we hypothesized that extracellular CL (1) modulates the secretion of cytokines and cytotoxins by astrocytes, as well as their phagocytic activity, and (2) acts by interacting with astrocyte TLR 4. We demonstrate that CL inhibits the lipopolysaccharide- (LPS-) induced secretion of cytotoxins and expression of glial fibrillary acidic protein (GFAP) by human U118 MG astrocytic cells. CL alone upregulates the phagocytic activity of human astrocytic cells and primary murine astrocytes. CL in combination with LPS upregulates secretion of interleukin (IL)-1ß by astrocytic cells. Furthermore, CL alone increases the secretion of monocyte chemoattractant protein (MCP)-1 by astrocytic cells, which is blocked by the TLR 4-specific antagonist TAK-242. We demonstrate that CL upregulates MCP-1 secretion in the absence of its natural carrier protein, ß2-glycoprotein 1, indicating that CL may be bioactive in the brain where this protein is not present. Lastly, we show that CL downregulates the expression of astrocytic TLR 4, implying that CL engages this receptor, as its activation has been shown to lead to its degradation. Overall, our study extends the list of cell type functions of which CL modulates and provides evidence that CL, or liposomes containing this phospholipid can be used to modulate specific neuroimmune functions of astrocytes.


Assuntos
Astrócitos , Receptor 4 Toll-Like , Animais , Astrócitos/metabolismo , Cardiolipinas/metabolismo , Cardiolipinas/farmacologia , Humanos , Imunidade , Camundongos , Microglia/metabolismo , Receptor 4 Toll-Like/metabolismo
7.
Biochim Biophys Acta Mol Basis Dis ; 1868(3): 166336, 2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-34973372

RESUMO

Specific diets regulate neuroimmune responses and modify risk of inflammatory bowel diseases, including ulcerative colitis. A link between gut and brain inflammation is also emerging. We hypothesized that adjusting dietary fatty acid composition modulates the neuroimmune responses in the mucin 2 knock out mice model of spontaneous colitis. Mice were randomly divided into three groups and fed isocaloric diets that only differed in their fatty acid composition. Diets enriched with anhydrous milk fat, corn oil, or Mediterranean diet fats were used. After nine weeks, brain and serum concentrations of ten inflammatory cytokines were measured. Three of these cytokines, including interleukin (IL)-2, IL-12 p70 and interferon-γ, were differentially expressed in the brains of animals from the three diet groups while there were no differences in the serum concentrations of these cytokines. Since only limited information is available about the functions of IL-2 in the central nervous system, in vitro experiments were performed to assess its effects on microglia. IL-2 had no effect on the secretion of neurotoxins and nitric oxide by microglia-like cells, but it selectively regulated phagocytic activity and reactive oxygen species production by stimulated microglia-like cells. Modulation of microglial reactive oxygen species through altered brain IL-2 concentrations could be one of the mechanisms linking diets with modified risk of neuroimmune disorders including Parkinson's disease.


Assuntos
Colite/complicações , Citocinas/metabolismo , Gorduras na Dieta/efeitos adversos , Microglia/patologia , Doenças Neuroinflamatórias/patologia , Animais , Ácidos Graxos/metabolismo , Feminino , Interferon gama/metabolismo , Interleucina-2/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/metabolismo , Mucina-2 , Doenças Neuroinflamatórias/etiologia , Doenças Neuroinflamatórias/metabolismo
8.
J Neuroimmunol ; 353: 577496, 2021 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-33517251

RESUMO

Microglia-driven neuroinflammation contributes to neurodegenerative diseases. Mitochondrial phospholipid cardiolipin acts as a signaling molecule when released from damaged cells. We demonstrate that extracellular cardiolipin induces the secretion of monocyte chemoattractant protein-1 and interferon gamma-induced protein 10 by resting microglia while inhibiting secretion of cytokines by microglia stimulated with lipopolysaccharide, amyloid Aß42 peptides, or α-synuclein. Extracellular cardiolipin also induces nitric oxide secretion by microglia-like cells and upregulates microglial phagocytosis. By using blocking antibodies, we determine that toll-like receptor 4 mediates the latter effect. Under physiological and pathological conditions characterized by cell death, extracellularly released cardiolipin may regulate immune responses of microglia.


Assuntos
Cardiolipinas/metabolismo , Citocinas/metabolismo , Microglia/metabolismo , Fagocitose/fisiologia , Receptor 4 Toll-Like/metabolismo , Animais , Cardiolipinas/imunologia , Citocinas/imunologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Microglia/imunologia , Células THP-1 , Receptor 4 Toll-Like/imunologia
9.
Mol Cell Neurosci ; 109: 103569, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33161065

RESUMO

Alzheimer's disease (AD) is a neurodegenerative disorder without a known cure or effective treatment. Research has identified several modifiable risk factors and suggested preventative measures to reduce the risk of developing AD, including alterations in diet. Polyunsaturated fatty acids (PUFAs) have been shown to regulate inflammatory responses in the central nervous system (CNS), the main site of inflammation in AD. In the CNS, microglia are immune cells responsible for the maintenance of homeostasis. However, in AD, microglia can become adversely activated, causing them to release increased levels of cytotoxins and inflammatory mediators, including nitric oxide (NO) and monocyte-chemoattractant protein (MCP)-1. We assessed the effects of two PUFAs, α-linolenic acid (ALA) and linoleic acid (LA), on select microglial immune functions, since the effects of these dietary fatty acids on neuroimmune responses are not well characterized. In BV-2 mouse microglia activated with lipopolysaccharide (LPS), exposure to LA reduced NO secretion and inducible nitric oxide synthase (iNOS) levels, whereas exposure to ALA reduced NO without a corresponding reduction of iNOS. Neither ALA nor LA altered MCP-1 levels or cytotoxins released by THP-1 human microglia-like cells stimulated with a combination of LPS and interferon (IFN)-γ. Specific receptor antagonists were used to demonstrate that the inhibitory effect of LA on NO secretion did not depend on the free fatty acid receptor (FFAR) 1 or FFAR4. Furthermore, gas chromatography with a flame ionization detector (GC-FID) revealed that exposure to LA or ALA did not alter the fatty acid composition of BV-2 microglia. Our data indicate that regulation of select microglial immune functions by ALA and LA could be one of the mechanisms underlying the observed link between certain dietary patterns and AD, such as reduced risk of cognitive decline and dementia associated with the Mediterranean diet.


Assuntos
Gorduras na Dieta/farmacologia , Ácido Linoleico/farmacologia , Microglia/efeitos dos fármacos , Óxido Nítrico/biossíntese , Ácido alfa-Linolênico/farmacologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/prevenção & controle , Animais , Linhagem Celular , Citocinas/metabolismo , Humanos , Mediadores da Inflamação/metabolismo , Lipídeos de Membrana/metabolismo , Camundongos , Microglia/imunologia , Microglia/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Receptores Acoplados a Proteínas G/fisiologia , Células THP-1
10.
CNS Neurol Disord Drug Targets ; 19(7): 483-494, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32614758

RESUMO

Glial cells, including microglia and astrocytes, facilitate the survival and health of all cells within the Central Nervous System (CNS) by secreting a range of growth factors and contributing to tissue and synaptic remodeling. Microglia and astrocytes can also secrete cytotoxins in response to specific stimuli, such as exogenous Pathogen-Associated Molecular Patterns (PAMPs), or endogenous Damage-Associated Molecular Patterns (DAMPs). Excessive cytotoxic secretions can induce the death of neurons and contribute to the progression of neurodegenerative disorders, such as Alzheimer's disease (AD). The transition between various activation states of glia, which include beneficial and detrimental modes, is regulated by endogenous molecules that include DAMPs, cytokines, neurotransmitters, and bioactive lipids, as well as a diverse group of mediators sometimes collectively referred to as Resolution-Associated Molecular Patterns (RAMPs). RAMPs are released by damaged or dying CNS cells into the extracellular space where they can induce signals in autocrine and paracrine fashions by interacting with glial cell receptors. While the complete range of their effects on glia has not been described yet, it is believed that their overall function is to inhibit adverse CNS inflammatory responses, facilitate tissue remodeling and cellular debris removal. This article summarizes the available evidence implicating the following RAMPs in CNS physiological processes and neurodegenerative diseases: cardiolipin (CL), prothymosin α (ProTα), binding immunoglobulin protein (BiP), heat shock protein (HSP) 10, HSP 27, and αB-crystallin. Studies on the molecular mechanisms engaged by RAMPs could identify novel glial targets for development of therapeutic agents that effectively slow down neuroinflammatory disorders including AD.


Assuntos
Mediadores da Inflamação/metabolismo , Doenças Neurodegenerativas/metabolismo , Neuroglia/metabolismo , Animais , Astrócitos/metabolismo , Sistema Nervoso Central/metabolismo , Citocinas/metabolismo , Proteínas de Choque Térmico , Humanos , Inflamação/metabolismo , Microglia/metabolismo , Chaperonas Moleculares , Neurônios/metabolismo , Transdução de Sinais
11.
Mol Cell Neurosci ; 105: 103493, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32333962

RESUMO

Neuroinflammation contributes to neurodegenerative disorders, including Alzheimer's disease (AD). Gut microbes are involved in regulating systemic inflammation. Short-chain fatty acids (SCFAs), which are among the many metabolites released by gut microbes, can cross the blood-brain barrier (BBB) and interact with microglia. High concentrations of individual SCFAs decrease the inflammatory responses of peripheral monocytes; therefore, we hypothesized that SCFAs act on their own or in combinations to reduce the inflammatory response of microglia. Cultured human THP-1 monocytic cells and differentiated human HL-60 myelomonocytic cells were used to model select immune functions of human microglia. Acetate, propionate, butyrate, formate, and valerate were added to cells alone or as a mixture containing SCFAs at an approximate physiological concentration ratio. The SCFA mixture, as well as several individual SCFAs at the highest concentrations used in the mixture (15-236 µM), decreased the secretion of interleukin (IL)-1ß, monocyte chemoattractant protein (MCP)-1, tumor necrosis factor (TNF)-α, and cytotoxins by immune-stimulated THP-1 cells. GLPG 0974, a free fatty acid receptor (FFAR) 2/3 antagonist, did not block the inhibitory effect of the SCFA mixture on IL-1ß secretion by THP-1 cells while blocking the inhibitory effect of formate alone. We demonstrated that formate and valerate alone reduced the phagocytic activity of immune-stimulated THP-1 cells. Formate, but not valerate, alone also inhibited the N-formylmethionine-leucyl-phenylalanine (fMLP)-induced respiratory burst of HL-60 cells, reducing the production of reactive oxygen species (ROS). Our data indicate that SCFAs could regulate select microglial functions that are disrupted in AD.


Assuntos
Butiratos/farmacologia , Ácidos Graxos Voláteis/metabolismo , Microglia/metabolismo , Monócitos/metabolismo , Tiofenos/farmacologia , Quimiocina CCL2/metabolismo , Ácidos Graxos Voláteis/imunologia , Humanos , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Microglia/efeitos dos fármacos , Microglia/imunologia , Monócitos/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo
12.
Expert Opin Ther Targets ; 23(10): 865-882, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31580163

RESUMO

Introduction: Adverse immune activation contributes to many central nervous system (CNS) disorders. All main CNS cell types express toll-like receptor 4 (TLR 4). This receptor is critical for a myriad of immune functions such as cytokine secretion and phagocytic activity of microglia; however, imbalances in TLR 4 activation can contribute to the progression of neurodegenerative diseases. Areas covered: We considered available evidence implicating TLR 4 activation in the following CNS pathologies: Alzheimer's disease, Parkinson's disease, ischemic stroke, traumatic brain injury, multiple sclerosis, multiple systems atrophy, and Huntington's disease. We reviewed studies reporting effects of TLR 4-specific antagonists and agonists in models of peripheral and CNS diseases from the perspective of possible future use of TLR 4 ligands in CNS disorders. Expert opinion: TLR 4-specific antagonists could suppress neuroinflammation by reducing overproduction of inflammatory mediators; however, they may interfere with protein clearance mechanisms and myelination. Agonists that specifically activate myeloid differentiation primary-response protein 88 (MyD88)-independent pathway of TLR 4 signaling could facilitate beneficial glial phagocytic activity with limited activity as inducers of proinflammatory mediators. Deciphering the disease stage-specific involvement of TLR 4 in CNS pathologies is crucial for the future clinical development of TLR 4 agonists and antagonists.


Assuntos
Doenças do Sistema Nervoso Central/tratamento farmacológico , Inflamação/tratamento farmacológico , Receptor 4 Toll-Like/metabolismo , Animais , Doenças do Sistema Nervoso Central/fisiopatologia , Desenvolvimento de Medicamentos , Humanos , Inflamação/fisiopatologia , Terapia de Alvo Molecular , Doenças Neurodegenerativas/tratamento farmacológico , Doenças Neurodegenerativas/fisiopatologia , Receptor 4 Toll-Like/agonistas , Receptor 4 Toll-Like/antagonistas & inibidores
13.
Biochim Biophys Acta Gen Subj ; 1863(11): 129400, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31344401

RESUMO

BACKGROUND: Chronic activation of glial cells contributes to neurodegenerative diseases. Cytochrome c (CytC) is a soluble mitochondrial protein that can act as a damage-associated molecular pattern (DAMP) when released into the extracellular space from damaged cells. CytC causes immune activation of microglia in a toll-like receptor (TLR) 4-dependent manner. The effects of extracellular CytC on astrocytes are unknown. Astrocytes, which are the most abundant glial cell type in the brain, express TLR 4 and secrete inflammatory mediators; therefore, we hypothesized that extracellular CytC can interact with the TLR 4 of astrocytes inducing their release of inflammatory molecules and cytotoxins. METHOD: Experiments were conducted using primary human astrocytes, U118 MG human astrocytic cells, BV-2 murine microglia, and SH-SY5Y human neuronal cells. RESULTS: Extracellularly applied CytC increased the secretion of interleukin (IL)-1ß, granulocyte-macrophage colony stimulating factor (GM-CSF) and IL-12 p70 by cultured primary human astrocytes. Anti-TLR 4 antibodies blocked the CytC-induced secretion of IL-1ß and GM-CSF by astrocytes. Supernatants from CytC-activated astrocytes were toxic to human SH-SY5Y neuronal cells. We also demonstrated CytC release from damaged glial cells by measuring CytC in the supernatants of BV-2 microglia after their exposure to cytotoxic concentrations of staurosporine, amyloid-ß peptides (Aß42) and tumor necrosis factor-α. CONCLUSION: CytC can be released into the extracellular space from damaged glial cells causing immune activation of astrocytes in a TLR 4-dependent manner. GENERAL SIGNIFICANCE: Astrocyte activation by CytC may contribute to neuroinflammation and neuronal death in neurodegenerative diseases. Astrocyte TLR 4 could be a potential therapeutic target in these diseases.


Assuntos
Astrócitos/metabolismo , Citocromos c/metabolismo , Interleucina-12/metabolismo , Interleucina-1beta/metabolismo , Receptor 4 Toll-Like/metabolismo , Astrócitos/patologia , Linhagem Celular Tumoral , Humanos , Inflamação/metabolismo , Inflamação/patologia
14.
Brain Res Bull ; 146: 153-163, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30625370

RESUMO

Cardiolipin is a mitochondrial membrane phospholipid with several well-defined metabolic roles. Cardiolipin can be released extracellularly by damaged cells and has been shown to affect peripheral immune functions. We hypothesized that extracellular cardiolipin can also regulate functions of microglia, the resident immune cells of the central nervous system (CNS). We demonstrate that extracellular cardiolipin increases microglial phagocytosis and neurotrophic factor expression, as well as decreases the release of inflammatory mediators and cytotoxins by activated microglia-like cells. These results identify extracellular cardiolipin as a potential CNS intercellular signaling molecule that can regulate key microglial immune functions associated with neurodegenerative diseases.


Assuntos
Cardiolipinas/fisiologia , Microglia/imunologia , Microglia/metabolismo , Animais , Cardiolipinas/metabolismo , Técnicas de Cultura de Células , Linhagem Celular , Sistema Nervoso Central/metabolismo , Citocinas/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Lipopolissacarídeos/metabolismo , Macrófagos , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Crescimento Neural/metabolismo , Doenças Neurodegenerativas/metabolismo , Fagocitose , Transdução de Sinais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA