Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 124
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Biochem Biophys Res Commun ; 709: 149828, 2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38537596

RESUMO

Long intergenic non-coding RNA 346 (LINC00346) has been reported to be involved in the development of atherosclerosis and specific cancers by affecting signaling pathways. However, its function in inflammation has not been thoroughly studied. Therefore, its expression pattern and function were determined in the human macrophage-like cell line THP-1. Lipopolysaccharide (LPS) treatment induced the expression of LINC00346. LPS-induced NF-κB activation and proinflammatory cytokine expression were suppressed or enhanced by the overexpression or knockdown of LINC00346, respectively. Analyses using dual luciferase assay and decoy RNAs that could block RNA-RNA interactions indicated that LINC00346 improves phosphatase and tensin homolog (PTEN) expression by sponging miR-25-3p. Subsequently, PTEN suppresses phosphoinositide-3 kinase (PI3K)-mediated conversion of phosphatidylinositol-4,5-bisphosphate (PIP2) into phosphatidylinositol-3,4,5-trisphosphate (PIP3) as well as consequent activation of protein kinase B (AKT) and NF-κB. Interestingly, database analysis revealed that the expression levels of LINC00346 and PTEN were simultaneously decreased in breast cancer tissues. Further analyses conducted using a breast cancer cell line, MDA-MB-231, confirmed the functional relationship among LINC00346, miR-25-3p, and PTEN in LPS-induced activation of NF-κB. These results indicate that miR-25-3p-sponging activity of LINC00346 affects the balance between PTEN and PI3K as well as the downstream activation of AKT/NF-κB pathway in inflammatory conditions.


Assuntos
Neoplasias da Mama , MicroRNAs , Feminino , Humanos , Lipopolissacarídeos/farmacologia , MicroRNAs/genética , MicroRNAs/metabolismo , NF-kappa B/metabolismo , Fosfatidilinositol 3-Quinase , Fosfatidilinositol 3-Quinases/metabolismo , Fosfatidilinositóis , Proteínas Proto-Oncogênicas c-akt/metabolismo , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo
2.
Int J Mol Sci ; 25(5)2024 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-38473915

RESUMO

Over the past century, molecular biology's focus has transitioned from proteins to DNA, and now to RNA. Once considered merely a genetic information carrier, RNA is now recognized as both a vital element in early cellular life and a regulator in complex organisms. Long noncoding RNAs (lncRNAs), which are over 200 bases long but do not code for proteins, play roles in gene expression regulation and signal transduction by inducing epigenetic changes or interacting with various proteins and RNAs. These interactions exhibit a range of functions in various cell types, including macrophages. Notably, some macrophage lncRNAs influence the activation of NF-κB, a crucial transcription factor governing immune and inflammatory responses. Macrophage NF-κB is instrumental in the progression of various pathological conditions including sepsis, atherosclerosis, cancer, autoimmune disorders, and hypersensitivity. It orchestrates gene expression related to immune responses, inflammation, cell survival, and proliferation. Consequently, its malfunction is a key contributor to the onset and development of these diseases. This review aims to summarize the function of lncRNAs in regulating NF-κB activity in macrophage activation and inflammation, with a particular emphasis on their relevance to human diseases and their potential as therapeutic targets. The insights gained from studies on macrophage lncRNAs, as discussed in this review, could provide valuable knowledge for the development of treatments for various pathological conditions involving macrophages.


Assuntos
NF-kappa B , RNA Longo não Codificante , Humanos , NF-kappa B/metabolismo , RNA Longo não Codificante/genética , Macrófagos/metabolismo , Transdução de Sinais/genética , Inflamação/metabolismo
3.
Expert Rev Proteomics ; 20(10): 197-209, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37724426

RESUMO

INTRODUCTION: Alzheimer's disease (AD) is a neurodegenerative disorder characterized by progressive cognitive decline, memory loss, and changes in behavior. Accumulating evidence indicates that dysfunction of glial cells, including astrocytes, microglia, and oligodendrocytes, may contribute to the development and progression of AD. Large-scale analysis of glial proteins sheds light on their roles in cellular processes and diseases. In AD, glial proteomics has been utilized to understand glia-based pathophysiology and identify potential biomarkers and therapeutic targets. AREA COVERED: In this review, we provide an updated overview of proteomic analysis of glia in the context of AD. Additionally, we discuss current challenges in the field, involving glial complexity and heterogeneity, and describe some cutting-edge proteomic technologies to address them. EXPERT OPINION: Unbiased comprehensive analysis of glial proteomes aids our understanding of the molecular and cellular mechanisms of AD pathogenesis. These investigations highlight the crucial role of glial cells and provide novel insights into the mechanisms of AD pathology. A deeper understanding of the AD-related glial proteome could offer a repertoire of potential biomarkers and therapeutics. Further technical advancement of glial proteomics will enable us to identify proteins within individual cells and specific cell types, thus significantly enhancing our comprehension of AD pathogenesis.


Assuntos
Doença de Alzheimer , Humanos , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Proteoma/genética , Proteoma/metabolismo , Proteômica , Neuroglia/metabolismo , Biomarcadores
4.
Expert Rev Proteomics ; 20(12): 371-379, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37978891

RESUMO

INTRODUCTION: Astrocytes are the most abundant cell type in the central nervous system (CNS). They play a pivotal role in supporting neuronal function and maintaining homeostasis by releasing a variety of bioactive proteins, collectively known as the astrocyte secretome. Investigating secretome provides insights into the molecular mechanisms underlying astrocyte function and dysfunction, as well as novel strategies to prevent and treat diseases affecting the CNS. AREAS COVERED: Proteomics databases are a valuable resource for studying the role of astrocytes in healthy and diseased brain function, as they provide information about gene expression, protein expression, and cellular function. In this review, we discuss existing databases that are useful for astrocyte secretome research. EXPERT OPINION: Astrocyte secretomics is a field that is rapidly progressing, yet the availability of dedicated databases is currently limited. To meet the increasing demand for comprehensive omics data in glia research, developing databases specifically focused on astrocyte secretome is crucial. Such databases would allow researchers to investigate the intricate molecular landscape of astrocytes and comprehend their involvement in diverse physiological and pathological processes. Expanding resources through the development of databases dedicated to the astrocyte secretome may facilitate further advancements in this field.


Assuntos
Astrócitos , Secretoma , Humanos , Astrócitos/metabolismo , Neuroglia/metabolismo , Neurônios/metabolismo , Proteínas/metabolismo
5.
Glia ; 70(10): 1902-1926, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35670184

RESUMO

Cathelicidin-related antimicrobial peptide (CRAMP) is an effector molecule of the innate immune system with direct antimicrobial and immunomodulatory activities; however, its role in neuroinflammatory responses and related diseases is not clearly understood. In particular, the expression of CRAMP and its functional role has not been previously studied in experimental autoimmune encephalomyelitis (EAE) or multiple sclerosis (MS). Here, we investigated the role of CRAMP in neuroinflammation, using an EAE mouse model of MS and postmortem patient tissues. We found that the CRAMP expression was increased in the spinal cords of EAE-induced mice. Immunofluorescence analysis revealed that CRAMP is mainly induced in reactive astrocytes in the inflamed spinal cord of EAE mice. A similar pattern of the LL-37 (human CRAMP) expression was observed in the brain and spinal cord tissues of patients with MS. An intrathecal injection of the CRAMP peptide in EAE mice accelerated the onset of symptoms and increased disease severity with augmented expression of inflammatory mediators, glial activation, infiltration of inflammatory cells, and demyelination. In addition, shRNA-mediated knockdown of Cramp in the spinal cord resulted in a milder disease course with less inflammation in EAE mice. We identified FPR2 on microglia as a CRAMP receptor and demonstrated that CRAMP potentiates IFN-γ-induced microglial activation via the STAT3 pathway. Taken together, our findings suggest that CRAMP is a novel mediator of astrocyte-microglia interactions in neuroinflammatory conditions such as EAE. Thus, CRAMP could be exploited as a biomarker or therapeutic target for the diagnosis or treatment of MS.


Assuntos
Encefalomielite Autoimune Experimental , Esclerose Múltipla , Animais , Peptídeos Catiônicos Antimicrobianos , Peptídeos Antimicrobianos , Astrócitos/metabolismo , Comunicação , Encefalomielite Autoimune Experimental/tratamento farmacológico , Encefalomielite Autoimune Experimental/metabolismo , Humanos , Inflamação/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Esclerose Múltipla/metabolismo , Doenças Neuroinflamatórias , Medula Espinal/metabolismo , Catelicidinas
6.
J Immunol ; 204(5): 1299-1309, 2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-31980577

RESUMO

LETM1 domain-containing protein 1 (LETMD1), also known as HCCR-1, is a mitochondrial protein and is known to regulate p53 and STAT3 activities in cancer cells. In this study, we present, for the first time (to our knowledge), data indicating that LETMD1 suppresses multiple immune responses in monocyte/macrophage lineage cells and mouse primary macrophages. Attenuation of LETMD1 expression with specific small interfering RNA and short hairpin RNA constructs enhanced LPS-induced expressions of inflammatory mediators in macrophages. In addition, LETMD1 attenuation caused potentiation of phagocytosis as well as migration in a macrophage-like cell line, U937. These enhancing effects were associated with altered activation of signaling adaptors (such as NF-κB, MAPKs, p53, and JAK-STAT) involved in TLR4 signaling. Especially, LETMD1 selectively regulated TLR4-induced NF-κB activation via MyD88 but not via TIR-domain-containing adapter-inducing IFN-ß (TRIF). Attenuation of LETMD1 expression caused mitochondrial hyperpolarization and subsequent decrease in ATP production and increase in mitochondrial/cellular reactive oxygen species (ROS) and intracellular calcium levels. LETMD1 attenuation also enhanced LPS-induced expression of NADPH oxidase (NOX) 2, the main producer of cellular ROS in phagocytes, through augmenting IFN regulatory factor 1. Accordingly, treatment with ROS scavenger, NOX2 suppressing agents, or calcium chelators resulted in suppression of LPS-induced cytokine production as well as NF-κB activation in cells with LETMD1 attenuation. These findings reveal a previously unknown function of LETMD1 and provide evidences showing LETMD1 negatively regulates macrophage functions by modulating mitochondrial function, subsequent ROS generation, and NF-κB activation.


Assuntos
Lipopolissacarídeos/toxicidade , Macrófagos/imunologia , NF-kappa B/imunologia , Fagocitose/efeitos dos fármacos , Proteínas Proto-Oncogênicas/imunologia , Espécies Reativas de Oxigênio/imunologia , Animais , Citocinas/imunologia , Células HEK293 , Humanos , Inflamação/induzido quimicamente , Inflamação/imunologia , Inflamação/patologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/imunologia , Macrófagos/patologia , Camundongos , Mitocôndrias/imunologia , Mitocôndrias/patologia , Células RAW 264.7 , Células THP-1 , Células U937
7.
J Biol Chem ; 295(50): 16906-16919, 2020 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-33060198

RESUMO

Kinases are critical components of intracellular signaling pathways and have been extensively investigated with regard to their roles in cancer. p21-activated kinase-1 (PAK1) is a serine/threonine kinase that has been previously implicated in numerous biological processes, such as cell migration, cell cycle progression, cell motility, invasion, and angiogenesis, in glioma and other cancers. However, the signaling network linked to PAK1 is not fully defined. We previously reported a large-scale yeast genetic interaction screen using toxicity as a readout to identify candidate PAK1 genetic interactions. En masse transformation of the PAK1 gene into 4,653 homozygous diploid Saccharomyces cerevisiae yeast deletion mutants identified ∼400 candidates that suppressed yeast toxicity. Here we selected 19 candidate PAK1 genetic interactions that had human orthologs and were expressed in glioma for further examination in mammalian cells, brain slice cultures, and orthotopic glioma models. RNAi and pharmacological inhibition of potential PAK1 interactors confirmed that DPP4, KIF11, mTOR, PKM2, SGPP1, TTK, and YWHAE regulate PAK1-induced cell migration and revealed the importance of genes related to the mitotic spindle, proteolysis, autophagy, and metabolism in PAK1-mediated glioma cell migration, drug resistance, and proliferation. AKT1 was further identified as a downstream mediator of the PAK1-TTK genetic interaction. Taken together, these data provide a global view of PAK1-mediated signal transduction pathways and point to potential new drug targets for glioma therapy.


Assuntos
Movimento Celular , Glioma/patologia , Saccharomyces cerevisiae/crescimento & desenvolvimento , Transdução de Sinais , Fuso Acromático/genética , Quinases Ativadas por p21/genética , Animais , Linhagem Celular , Proliferação de Células , Sobrevivência Celular , Modelos Animais de Doenças , Epistasia Genética , Feminino , Glioma/genética , Glioma/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Mitose , Inibidores de Proteínas Quinases/farmacologia , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Quinases Ativadas por p21/metabolismo
8.
Glia ; 69(4): 971-996, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33251681

RESUMO

Diabetic peripheral neuropathy (DPN) is a common complication of uncontrolled diabetes. The pathogenesis of DPN is associated with chronic inflammation in dorsal root ganglion (DRG), eventually causing structural and functional changes. Studies on DPN have primarily focused on neuronal component, and there is limited knowledge about the role of satellite glial cells (SGCs), although they completely enclose neuronal soma in DRG. Lipocalin-2 (LCN2) is a pro-inflammatory acute-phase protein found in high levels in diverse neuroinflammatory and metabolic disorders. In diabetic DRG, the expression of LCN2 was increased exclusively in the SGCs. This upregulation of LCN2 in SGCs correlated with increased inflammatory responses in DRG and sciatic nerve. Furthermore, diabetes-induced inflammation and morphological changes in DRG, as well as sciatic nerve, were attenuated in Lcn2 knockout (KO) mice. Lcn2 gene ablation also ameliorated neuropathy phenotype as determined by nerve conduction velocity and intraepidermal nerve fiber density. Mechanistically, studies using specific gene KO mice, adenovirus-mediated gene overexpression strategy, and primary cultures of DRG SGCs and neurons have demonstrated that LCN2 enhances the expression of mitochondrial gate-keeping regulator pyruvate dehydrogenase kinase-2 (PDK2) through PPARß/δ, thereby inhibiting pyruvate dehydrogenase activity and increasing production of glycolytic end product lactic acid in DRG SGCs and neurons of diabetic mice. Collectively, our findings reveal a crucial role of glial LCN2-PPARß/δ-PDK2-lactic acid axis in progression of DPN. Our results establish a link between pro-inflammatory LCN2 and glycolytic PDK2 in DRG SGCs and neurons and propose a novel glia-based mechanism and drug target for therapy of DPN. MAIN POINTS: Diabetes upregulates LCN2 in satellite glia, which in turn increases pyruvate dehydrogenase kinase-2 (PDK2) expression and lactic acid production in dorsal root ganglia (DRG). Glial LCN2-PDK2-lactic acid axis in DRG plays a crucial role in the pathogenesis of diabetic neuropathy.


Assuntos
Diabetes Mellitus Experimental , Neuropatias Diabéticas , Lipocalina-2 , PPAR beta , Animais , Camundongos , Gânglios Espinais , Inflamação , Ácido Láctico , Lipocalina-2/genética , Camundongos Knockout , Neuroglia , Piruvato Desidrogenase Quinase de Transferência de Acetil
9.
Biochem Cell Biol ; 98(2): 137-144, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31071273

RESUMO

Mitochondria affect cellular functions alone or in cooperation with other cellular organelles. Recent research has demonstrated the close relationship of mitochondria with the endoplasmic reticulum (ER), both at the physical and the functional level. In an effort to define the combined effect of mitochondrial dysfunction (MD) and ER stress in the proinflammatory activities of macrophages, the human macrophage-like monocytic leukemia cell line THP-1 was treated with mitochondrial electron transport chain (ETC) blockers, and changes in the cellular responses upon stimulation by interferon (IFN)-γ were analyzed. Inducing mitochondrial dysfunction (MD) with ETC blockers resulted in suppression of IFN-induced activation of JAK1 and STAT1/3, as well as the expression of STAT1-regulated genes. In addition, experiments utilizing pharmacological modulators of adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) and liver kinase B1 (LKB1)-deficient HeLa cells demonstrated that these suppressive effects are mediated by the LKB1-AMPK pathway. Treatment with pharmacological inhibitors of ER stress sensors failed to affect these processes, thus indicating that involvement of ER stress is not required. These results indicate that MD, induced by blocking the ETC, affects IFN-induced activation of JAK-STAT and associated inflammatory changes in THP-1 cells through the LKB1-AMPK pathway independently of ER stress.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Janus Quinase 1/metabolismo , Mitocôndrias/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Fator de Transcrição STAT1/metabolismo , Quinases Proteína-Quinases Ativadas por AMP , Transporte de Elétrons , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Glicosilação , Células HEK293 , Células HeLa , Humanos , Inflamação , Interferon gama/metabolismo , Fosforilação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Células THP-1
10.
Expert Rev Proteomics ; 17(3): 207-220, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32187501

RESUMO

Introduction: Glial cells are closely associated with neurons located throughout the nervous system and regulate neuronal activity and function through various mechanisms including the secretion of proteins and other signaling molecules. Glia-secreted proteins play crucial roles in modulating neuronal function in physiological and pathological conditions. Aberrant activation of glial cells leading to neuroinflammation is a common phenomenon observed in various neurological disorders. Aberrantly activated glial cells secrete proteins in disease-specific manner and can be exploited as a repository for novel biomarker discovery.Areas covered: In this review, we describe the recent advances in proteomic techniques, highlighting the need for their application to the secretomic field. Studies regarding the secretome profile of glial cells published within the last 5 years are discussed in detail. The use of glia-based biomarkers in various neuroinflammatory and neurodegenerative diseases is also discussed.Expert opinion: Precise diagnosis and timely treatment of neurological disorders remains a challenge and glia-focused research to identify specific biomarkers appears to be a promising approach to combat these disorders. Recent technological advancement in proteomic research would open new frontiers for more rigorous analysis of glial secretome variations over time and the discovery/development of novel biomarkers for neurological disorders.


Assuntos
Doenças Neurodegenerativas/genética , Neuroglia/metabolismo , Neurônios/metabolismo , Proteômica , Biomarcadores/metabolismo , Humanos , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Neuroglia/patologia , Neurônios/patologia
11.
Cell Biol Int ; 43(3): 313-322, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30632648

RESUMO

The functional and physical interaction between mitochondria and the endoplasmic reticulum (ER) has been the subject of intense study. To test the effect of this interaction on macrophage inflammatory activation, the human macrophage-like monocytic leukemia cell line THP-1 was treated with oligomycin, rotenone, or sodium azide, which induce mitochondrial dysfunction (MD) by blocking the electron transport chain (ETC). MD induced by these agents triggered activation of various sensors and markers of ER stress. This linkage affected macrophage function since LPS-induced expression of IL-23 was enhanced by the MD inducers, and this enhancing effect was abolished by inhibition of pancreatic endoplasmic reticulum kinase (PERK) activity. This MD-mediated ER stress may be universal since it was observed in human embryonic kidney HEK293 cells and colon cancer SW480 cells. On the other hand, MD regulated LPS-induced activation of the AKT/GSK3ß/ß-catenin pathway in a manner not affected by inhibition of PERK or inositol-requiring enzyme 1α (IRE1α) activities. These results indicate that the occurrence of MD can lead to ER stress and these two events, separately or in combination, can affect various cellular processes.


Assuntos
Estresse do Retículo Endoplasmático , Mediadores da Inflamação/metabolismo , Mitocôndrias/patologia , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Glicogênio Sintase Quinase 3 beta/metabolismo , Células HEK293 , Humanos , Interleucina-23/metabolismo , Lipopolissacarídeos/farmacologia , Mitocôndrias/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Transdução de Sinais/efeitos dos fármacos , Células THP-1 , Fator de Transcrição CHOP/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , beta Catenina/metabolismo
12.
Glia ; 65(9): 1471-1490, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28581123

RESUMO

Lipocalin-2 (LCN2) has diverse functions in multiple pathophysiological conditions; however, its pathogenic role in vascular dementia (VaD) is unknown. Here, we investigated the role of LCN2 in VaD using rodent models of global cerebral ischemia and hypoperfusion with cognitive impairment and neuroinflammation. Mice subjected to transient bilateral common carotid artery occlusion (tBCCAo) for 50 min showed neuronal death and gliosis in the hippocampus at 7 days post-tBCCAo. LCN2 expression was observed predominantly in the hippocampal astrocytes, whereas its receptor was mainly detected in neurons, microglia, and astrocytes. Furthermore, Lcn2-deficient mice, compared with wild-type animals, showed significantly weaker CA1 neuronal loss, cognitive decline, white matter damage, blood-brain barrier permeability, glial activation, and proinflammatory cytokine production in the hippocampus after tBCCAo. Lcn2 deficiency also attenuated hippocampal neuronal death and cognitive decline at 30 days after unilateral common carotid artery occlusion (UCCAo). Furthermore, intracerebroventricular (i.c.v) injection of recombinant LCN2 protein elicited CA1-neuronal death and a cognitive deficit. Our studies using cultured glia and hippocampal neurons supported the decisive role of LCN2 in hippocampal neurotoxicity and microglial activation, and the role of the HIF-1α-LCN2-VEGFA axis of astrocytes in vascular injury. Additionally, plasma levels of LCN2 were significantly higher in patients with VaD than in the healthy control subjects. These results indicate that hippocampal damage and cognitive impairment are mediated by LCN2 secreted from reactive astrocytes in VaD.


Assuntos
Astrócitos/metabolismo , Disfunção Cognitiva/metabolismo , Demência Vascular/metabolismo , Hipocampo/metabolismo , Lipocalina-2/metabolismo , Animais , Astrócitos/patologia , Biomarcadores/sangue , Células Cultivadas , Cognição/fisiologia , Disfunção Cognitiva/patologia , Demência Vascular/patologia , Modelos Animais de Doenças , Hipocampo/irrigação sanguínea , Hipocampo/patologia , Humanos , Lipocalina-2/administração & dosagem , Lipocalina-2/genética , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/metabolismo , Microglia/patologia , Microvasos/metabolismo , Microvasos/patologia , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
13.
Cell Immunol ; 315: 64-70, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28391993

RESUMO

Sodium azide (NaN3) is a chemical compound with multiple toxic effects on vascular and neuronal systems, causing hypotension and neurotoxicity, respectively. In order to test its effects on the immune system, human and mouse macrophage-like cell lines were treated with nontoxic doses of NaN3 and the changes in LPS-induced inflammatory activation was measured. Interestingly, the LPS-induced expression of monocyte chemoattractant protein (MCP)-1 was suppressed by NaN3 without affecting the expression of IL-8 and TNF-α. Further analysis of cellular signaling mediators involved in the expression of these cytokines revealed that NaN3 suppressed the LPS-induced activation of signal transducers and activator of transcription (STAT)1 and inhibitor of κB (IκB) ς, which are involved in the LPS-induced expression of MCP-1, while the LPS-induced activation of nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) was not affected. The LPS-induced expression of MCP-2 and CXCL10, which are also regulated by STAT1, was suppressed by NaN3. Similarly, the LPS-induced expression of IL-6, which is regulated by IκBζ, was suppressed by NaN3. These results demonstrate that NaN3 selectively suppresses the LPS-induced expression of pro-inflammatory mediators through the suppression of STAT1 and IκBζ activation. These new findings about the activity of NaN3 may contribute to the development of specific regulators of macrophage activity during acute and chronic inflammation.


Assuntos
Quimiocina CCL2/biossíntese , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas I-kappa B/antagonistas & inibidores , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Proteínas Nucleares/antagonistas & inibidores , Fator de Transcrição STAT1/antagonistas & inibidores , Azida Sódica/farmacologia , Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Animais , Linhagem Celular Tumoral , Quimiocina CCL2/genética , Proteínas I-kappa B/biossíntese , Proteínas I-kappa B/genética , Mediadores da Inflamação/metabolismo , Interleucina-8/biossíntese , Interleucina-8/genética , Leucemia Monocítica Aguda/patologia , Macrófagos/metabolismo , Camundongos , NF-kappa B/metabolismo , Proteínas Nucleares/biossíntese , Proteínas Nucleares/genética , Fosforilação/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Células RAW 264.7 , Fator de Transcrição STAT1/metabolismo , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/genética
14.
J Neurosci Res ; 94(9): 837-49, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-26931482

RESUMO

Pyruvate dehydrogenase (PDH) kinases (PDKs) 1-4, expressed in peripheral and central tissues, regulate the activity of the PDH complex (PDC). The PDC is an important mitochondrial gatekeeping enzyme that controls cellular metabolism. The role of PDKs in diverse neurological disorders, including neurometabolic aberrations and neurodegeneration, has been described. Implications for a role of PDKs in inflammation and neurometabolic coupling led us to investigate the effect of genetic ablation of PDK2/4 on nociception in a mouse model of acute inflammatory pain. Deficiency in Pdk2 and/or Pdk4 in mice led to attenuation of formalin-induced nociceptive behaviors (flinching, licking, biting, or lifting of the injected paw). Likewise, the pharmacological inhibition of PDKs substantially diminished the nociceptive responses in the second phase of the formalin test. Furthermore, formalin-provoked paw edema formation and mechanical and thermal hypersensitivities were significantly reduced in Pdk2/4-deficient mice. Formalin-driven neutrophil recruitment at the site of inflammation, spinal glial activation, and neuronal sensitization were substantially lessened in the second or late phase of the formalin test in Pdk2/4-deficient animals. Overall, our results suggest that PDK2/4 can be a potential target for the development of pharmacotherapy for the treatment of acute inflammatory pain. © 2016 Wiley Periodicals, Inc.


Assuntos
Dor Aguda/genética , Dor Aguda/psicologia , Comportamento Animal , Inflamação/genética , Inflamação/psicologia , Nociceptividade , Proteínas Serina-Treonina Quinases/genética , Animais , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Ativação de Macrófagos/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neuroglia , Infiltração de Neutrófilos/genética , Medição da Dor , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Piruvato Desidrogenase Quinase de Transferência de Acetil , Medula Espinal/patologia
15.
J Biol Chem ; 289(24): 16773-89, 2014 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-24808182

RESUMO

Lipocalin-2 (LCN2) plays an important role in cellular processes as diverse as cell growth, migration/invasion, differentiation, and death/survival. Furthermore, recent studies indicate that LCN2 expression and secretion by glial cells are induced by inflammatory stimuli in the central nervous system. The present study was undertaken to examine the regulation of LCN2 expression in experimental autoimmune encephalomyelitis (EAE) and to determine the role of LCN2 in the disease process. LCN2 expression was found to be strongly increased in spinal cord and secondary lymphoid tissues after EAE induction. In spinal cords astrocytes and microglia were the major cell types expressing LCN2 and its receptor 24p3R, respectively, whereas in spleens, LCN2 and 24p3R were highly expressed in neutrophils and dendritic cells, respectively. Furthermore, disease severity, inflammatory infiltration, demyelination, glial activation, the expression of inflammatory mediators, and the proliferation of MOG-specific T cells were significantly attenuated in Lcn2-deficient mice as compared with wild-type animals. Myelin oligodendrocyte glycoprotein-specific T cells in culture exhibited an increased expression of Il17a, Ifng, Rorc, and Tbet after treatment with recombinant LCN2 protein. Moreover, LCN2-treated glial cells expressed higher levels of proinflammatory cytokines, chemokines, and MMP-9. Adoptive transfer and recombinant LCN2 protein injection experiments suggested that LCN2 expression in spinal cord and peripheral immune organs contributes to EAE development. Taken together, these results imply LCN2 is a critical mediator of autoimmune inflammation and disease development in EAE and suggest that LCN2 be regarded a potential therapeutic target in multiple sclerosis.


Assuntos
Proteínas de Fase Aguda/metabolismo , Encefalomielite Autoimune Experimental/metabolismo , Lipocalinas/metabolismo , Tecido Linfoide/metabolismo , Proteínas Oncogênicas/metabolismo , Medula Espinal/metabolismo , Proteínas de Fase Aguda/genética , Animais , Astrócitos/metabolismo , Células Cultivadas , Células Dendríticas/metabolismo , Deleção de Genes , Interferon gama/genética , Interferon gama/metabolismo , Interleucina-17/genética , Interleucina-17/metabolismo , Lipocalina-2 , Lipocalinas/genética , Tecido Linfoide/patologia , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Bainha de Mielina/genética , Bainha de Mielina/metabolismo , Neutrófilos/metabolismo , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Proteínas Oncogênicas/genética , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Medula Espinal/patologia , Linfócitos T/metabolismo
16.
Cell Immunol ; 296(2): 115-21, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25929183

RESUMO

MARCKS, a substrate of protein kinase C, is involved in various processes associated with cytoskeletal movement. Although the expression of MARCKS is highly induced in macrophages, its role in macrophage function has not been studied in detail. Notably, the suppression of MARCKS expression in macrophage cell lines blocked LPS-induced expression of TNF-α at the transcriptional level. Treatment of macrophages with MARCKS N-terminus sequence (MANS) and effector domain (ED) peptides, which mimic functional domains and block the phosphorylation of MARCKS, suppressed the LPS-induced expression of TNF-α through suppression of p38 and JNK MAPKs and NF-κB. Treatment of mice with MANS peptide reduced serum TNF-α and IL-6 levels and resulted in 40% survival of mice after the administration of a lethal dose of LPS. These data demonstrate that MARCKS is involved in the regulation of proinflammatory cytokine expression in macrophages and that MARCKS-derived peptides can be used to suppress inflammatory responses.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/genética , MAP Quinase Quinase 4/genética , Macrófagos Peritoneais/imunologia , Proteínas de Membrana/genética , NF-kappa B/genética , Peptídeos/farmacologia , Sepse/genética , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Animais , Linhagem Celular , Regulação da Expressão Gênica , Humanos , Interleucina-6/antagonistas & inibidores , Interleucina-6/genética , Interleucina-6/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Lipopolissacarídeos/antagonistas & inibidores , Lipopolissacarídeos/farmacologia , MAP Quinase Quinase 4/imunologia , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/patologia , Masculino , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/imunologia , Camundongos , Substrato Quinase C Rico em Alanina Miristoilada , NF-kappa B/imunologia , Cultura Primária de Células , Proteína Quinase C/genética , Proteína Quinase C/imunologia , Estrutura Terciária de Proteína , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/imunologia , Sepse/tratamento farmacológico , Sepse/imunologia , Sepse/mortalidade , Transdução de Sinais , Análise de Sobrevida , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia , Proteínas Quinases p38 Ativadas por Mitógeno/imunologia
17.
Immunol Invest ; 44(3): 309-20, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25831081

RESUMO

Fascin is a well-known cytoskeletal regulatory protein that, as a substrate of protein kinase C (PKC), is involved in PKC-mediated translational regulation of TNF-α in macrophages stimulated with lipopolysaccharide (LPS). The regulatory effects of fascin targeted the 3'-untraslated region (UTR) of the TNF-α mRNA, and suppression of PKC activity or fascin expression resulted in specific blockage of the LPS-induced translational activation of the mRNA. In an effort to identify the molecular mechanism of this fascin-mediated translational regulation, the expression levels of micro-RNA (miRNA) after stimulation of the toll-like receptor 4 (TLR4) signaling pathways were analyzed in cells with down-regulation of fascin. The LPS-induced translation of TNF-α is known to be regulated by miR-155 and miR-125b, which have positive and negative effects, respectively. Interestingly, suppression of fascin expression reversed LPS-induced down-regulation of miR-125b and abolished the LPS-induced increase in miR-155. Furthermore, introduction of miR-155 precursor, blocking of miR-125b activity, or introduction of a mutation into the miR-125b binding site of the TNF-α 3'-UTR restored translational activation in cells with suppressed fascin expression. These data indicate that fascin regulates translation through miR-155 and miR-125b, which target 3' UTR in TNF-α mRNA.


Assuntos
Proteínas de Transporte/metabolismo , Macrófagos/fisiologia , MicroRNAs/metabolismo , Proteínas dos Microfilamentos/metabolismo , Fator de Necrose Tumoral alfa/genética , Regiões 3' não Traduzidas/genética , Sítios de Ligação/genética , Proteínas de Transporte/genética , Regulação da Expressão Gênica/genética , Células HEK293 , Humanos , Lipopolissacarídeos/imunologia , MicroRNAs/genética , Proteínas dos Microfilamentos/genética , Mutação/genética , Biossíntese de Proteínas/genética , Proteína Quinase C/metabolismo , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Transdução de Sinais/genética , Receptor 4 Toll-Like/metabolismo
18.
J Immunol ; 191(10): 5204-19, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-24089194

RESUMO

Astrocytes provide structural and functional support for neurons, as well as display neurotoxic or neuroprotective phenotypes depending upon the presence of an immune or inflammatory microenvironment. This study was undertaken to characterize multiple phenotypes of activated astrocytes and to investigate the regulatory mechanisms involved. We report that activated astrocytes in culture exhibit two functional phenotypes with respect to pro- or anti-inflammatory gene expression, glial fibrillary acidic protein expression, and neurotoxic or neuroprotective activities. The two distinct functional phenotypes of astrocytes were also demonstrated in a mouse neuroinflammation model, which showed pro- or anti-inflammatory gene expression in astrocytes following challenge with classical or alternative activation stimuli; similar results were obtained in the absence of microglia. Subsequent studies involving recombinant lipocalin-2 (LCN2) protein treatment or Lcn2-deficient mice indicated that the pro- or anti-inflammatory functionally polarized phenotypes of astrocytes and their intracellular signaling pathway were critically regulated by LCN2 under in vitro and in vivo conditions. Astrocyte-derived LCN2 promoted classical proinflammatory activation of astrocytes but inhibited IL-4-STAT6 signaling, a canonical pathway involved in alternative anti-inflammatory activation. Our results suggest that the secreted protein LCN2 is an autocrine modulator of the functional polarization of astrocytes in the presence of immune or inflammatory stimuli and that LCN2 could be targeted therapeutically to dampen proinflammatory astrocytic activation and related pathologies in the CNS.


Assuntos
Proteínas de Fase Aguda/metabolismo , Astrócitos/metabolismo , Encéfalo/imunologia , Lipocalinas/metabolismo , Proteínas Oncogênicas/metabolismo , Proteínas de Fase Aguda/deficiência , Proteínas de Fase Aguda/genética , Animais , Astrócitos/citologia , Astrócitos/imunologia , Polaridade Celular , Células Cultivadas , Inflamação/imunologia , Interleucina-4/metabolismo , Lipocalina-2 , Lipocalinas/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/citologia , Proteínas Oncogênicas/deficiência , Proteínas Oncogênicas/genética , Fator de Transcrição STAT6/metabolismo , Transdução de Sinais/imunologia
19.
J Biol Chem ; 288(33): 24116-27, 2013 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-23836894

RESUMO

Lipocalin 2 (LCN2), which is also known as 24p3 and neutrophil gelatinase-associated lipocalin (NGAL), binds small, hydrophobic ligands and interacts with cell surface receptor 24p3R to regulate diverse cellular processes. In the present study, we examined the role of LCN2 in the pathogenesis of neuropathic pain using a mouse model of spared nerve injury (SNI). Lcn2 mRNA levels were significantly increased in the dorsal horn of the spinal cord after SNI, and LCN2 protein was mainly localized in neurons of the dorsal and ventral horns. LCN2 receptor 24p3R was expressed in spinal neurons and microglia after SNI. Lcn2-deficient mice exhibited significantly less mechanical pain hypersensitivity during the early phase after SNI, and an intrathecal injection of recombinant LCN2 protein elicited mechanical pain hypersensitivity in naive animals. Lcn2 deficiency, however, did not affect acute nociceptive pain. Lcn2-deficient mice showed significantly less microglial activation and proalgesic chemokine (CCL2 and CXCL1) production in the spinal cord after SNI than wild-type mice, and recombinant LCN2 protein induced the expression of these chemokines in cultured neurons. Furthermore, the expression of LCN2 and its receptor was detected in neutrophils and macrophages in the sciatic nerve following SNI, suggesting the potential role of peripheral LCN2 in neuropathic pain. Taken together, our results indicate that LCN2 plays a critical role in the development of pain hypersensitivity following peripheral nerve injury and suggest that LCN2 mediates neuropathic pain by inducing chemokine expression and subsequent microglial activation.


Assuntos
Proteínas de Fase Aguda/metabolismo , Quimiocinas/metabolismo , Lipocalinas/metabolismo , Neuralgia/etiologia , Neuralgia/metabolismo , Proteínas Oncogênicas/metabolismo , Traumatismos dos Nervos Periféricos/complicações , Traumatismos dos Nervos Periféricos/metabolismo , Transdução de Sinais , Proteínas de Fase Aguda/genética , Proteínas de Fase Aguda/farmacologia , Animais , Células Cultivadas , Córtex Cerebral/patologia , Quimiocinas/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Hiperalgesia/complicações , Hiperalgesia/metabolismo , Hiperalgesia/patologia , Lipocalina-2 , Lipocalinas/genética , Lipocalinas/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Microglia/enzimologia , Microglia/patologia , Modelos Biológicos , Neuralgia/enzimologia , Neuralgia/patologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Nociceptividade/efeitos dos fármacos , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/farmacologia , Traumatismos dos Nervos Periféricos/enzimologia , Traumatismos dos Nervos Periféricos/patologia , Fosforilação/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Superfície Celular/metabolismo , Nervo Isquiático/efeitos dos fármacos , Nervo Isquiático/metabolismo , Nervo Isquiático/patologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Medula Espinal/patologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
20.
FASEB J ; 27(3): 1176-90, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23207546

RESUMO

Activated macrophages are classified into two different forms: classically activated (M1) or alternatively activated (M2) macrophages. The presence of M1/M2 phenotypic polarization has also been suggested for microglia. Here, we report that the secreted protein lipocalin 2 (LCN2) amplifies M1 polarization of activated microglia. LCN2 protein (EC 1 µg/ml), but not glutathione S-transferase used as a control, increased the M1-related gene expression in cultured mouse microglial cells after 8-24 h. LCN2 was secreted from M1-polarized, but not M2-polarized, microglia. LCN2 inhibited phosphorylation of STAT6 in IL-4-stimulated microglia, suggesting LCN2 suppression of the canonical M2 signaling. In the lipopolysaccharide (LPS)-induced mouse neuroinflammation model, the expression of LCN2 was notably increased in microglia. Primary microglial cultures derived from LCN2-deficient mice showed a suppressed M1 response and enhanced M2 response. Mice lacking LCN2 showed a markedly reduced M1-related gene expression in microglia after LPS injection, which was consistent with the results of histological analysis. Neuroinflammation-associated impairment in motor behavior and cognitive function was also attenuated in the LCN2-deficient mice, as determined by the rotarod performance test, fatigue test, open-field test, and object recognition task. These findings suggest that LCN2 is an M1-amplifier in brain microglial cells.


Assuntos
Proteínas de Fase Aguda/metabolismo , Polaridade Celular/fisiologia , Lipocalinas/metabolismo , Microglia/metabolismo , Proteínas Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas de Fase Aguda/genética , Animais , Polaridade Celular/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Humanos , Inflamação/induzido quimicamente , Inflamação/genética , Inflamação/metabolismo , Células Jurkat , Lipocalina-2 , Lipocalinas/genética , Lipopolissacarídeos/toxicidade , Camundongos , Camundongos Mutantes , Microglia/citologia , Proteínas Oncogênicas/genética , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Proteínas Proto-Oncogênicas/genética , Fator de Transcrição STAT6/genética , Fator de Transcrição STAT6/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA