Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
J Neurochem ; 158(1): 14-24, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33025576

RESUMEN

Metabolism, the umbrella term for complex biochemical pathways that sustain the basic functions of life, has garnered attention in recent years for its role in immune activation. Indeed, metabolic pathways and their intricate and complex connections with immune mechanisms constitute a new area of immunology termed 'immunometabolism'. One highlight is the existence of a switch in the key metabolic programs in immune cells, which executes their effector functions. 'Metabolic reprogramming' is observed in conditions of both peripheral diseases as well as in neurodegenerative conditions associated with inflammation such as multiple sclerosis. Moreover metabolic reprogramming occurs for almost every immune cell type. Whether metabolic changes are cause or effect of immune activation, however, remains to be fully understood. Being central to cellular activation, metabolism has become very topical in terms of exploring therapeutic targets. This review covers the major metabolic programs in immune cells, discuss metabolites as regulators of immune cell functions, and consider metabolic enzymes or pathways as therapeutic targets using examples from multiple sclerosis and its animal models.


Asunto(s)
Leucocitos/inmunología , Leucocitos/metabolismo , Redes y Vías Metabólicas/inmunología , Microglía/inmunología , Microglía/metabolismo , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/metabolismo , Animales , Humanos , Inflamación
2.
J Neuroinflammation ; 18(1): 301, 2021 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-34952601

RESUMEN

BACKGROUND: Multiple sclerosis (MS) is a neurodegenerative condition of the central nervous system (CNS). It is associated with blood-brain barrier (BBB) breakdown and intravasation of leukocytes, particularly monocyte-derived macrophages, into the CNS. Pericytes are mural cells that are encased within the basement membrane of vasculature, and they contribute functionally to the neurovascular unit. These cells play an important role in maintaining BBB integrity and CNS homeostasis. However, the critical role of pericytes in mediating inflammation in MS or its models is unclear. Whether pericytes infiltrate into the CNS parenchyma in MS also needs clarification. METHODS: CNS samples from the experimental autoimmune encephalomyelitis (EAE) mouse model of MS were collected at different time points for immunohistochemical analysis of pericytes along the inflamed vasculature. These findings were validated using MS brain specimens, and further analysis of pericyte involvement in inflammation was carried out by culturing primary pericytes and macrophages. Multiplex ELISA, transmigration assay and real-time PCR were used to study the inflammatory potential of pericytes in cultures. RESULTS: We found that pericytes exhibit a heterogenous morphology, with notable elongation in the inflamed perivascular cuffs of EAE. This was manifested by a decrease in pericyte density but an increase in the coverage by pericytes along the vasculature. Chondroitin sulfate proteoglycans (CSPGs), a family of extracellular matrix proteins enriched within inflamed perivascular cuffs, elevated levels of pro-inflammatory chemokines/cytokines in pericytes in culture. Importantly, pericytes stimulated with CSPGs enhanced macrophage migration. We did not detect pericytes in the CNS parenchyma during EAE, and this was corroborated in MS brain samples. CONCLUSIONS: Our data suggest that pericytes seek to restore the BBB through increased coverage, but that their exposure to CSPGs prompt their facilitation of macrophages to enter the CNS to elevate neuroinflammation in EAE and MS.


Asunto(s)
Encefalomielitis Autoinmune Experimental/patología , Macrófagos/patología , Esclerosis Múltiple/patología , Pericitos/patología , Animales , Barrera Hematoencefálica/citología , Barrera Hematoencefálica/patología , Encéfalo/patología , Quimiocinas/metabolismo , Citocinas/metabolismo , Encefalitis/patología , Femenino , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Pericitos/ultraestructura , Cultivo Primario de Células
3.
J Neurosci Res ; 98(12): 2390-2405, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32815569

RESUMEN

Pericytes are contractile cells that extend along the vasculature to mediate key homeostatic functions of endothelial barriers within the body. In the central nervous system (CNS), pericytes are important contributors to the structure and function of the neurovascular unit, which includes endothelial cells, astrocytes and neurons. The understanding of pericytes has been marred by an inability to accurately distinguish pericytes from other stromal cells with similar expression of identifying markers. Evidence is now growing in favor of pericytes being actively involved in both CNS homeostasis and pathology of neurological diseases, including multiple sclerosis, spinal cord injury, and Alzheimer's disease among others. In this review, we discuss the current understanding on the characterization of pericytes, their roles in maintaining the integrity of the blood-brain barrier, and their contributions to neuroinflammation and neurorepair. Owing to its plethora of surface receptors, pericytes respond to inflammatory mediators such as CCL2 (monocyte chemoattractant protein-1) and tumor necrosis factor-α, in turn secreting CCL2, nitric oxide, and several cytokines. Pericytes can therefore act as promoters of both the innate and adaptive arms of the immune system. Much like professional phagocytes, pericytes also have the ability to clear up cellular debris and macromolecular plaques. Moreover, pericytes promote the activities of CNS glia, including in maturation of oligodendrocyte lineage cells for myelination. Conversely, pericytes can impair regenerative processes by contributing to scar formation. A better characterization of CNS pericytes and their functions would bode well for therapeutics aimed at alleviating their undesirable properties and enhancing their benefits.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Homeostasis/fisiología , Mediadores de Inflamación/metabolismo , Enfermedades del Sistema Nervioso/metabolismo , Pericitos/metabolismo , Animales , Barrera Hematoencefálica/inmunología , Encéfalo/inmunología , Endocitosis/fisiología , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Humanos , Mediadores de Inflamación/inmunología , Enfermedades del Sistema Nervioso/inmunología , Pericitos/inmunología
4.
J Virol ; 88(9): 4798-810, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24522920

RESUMEN

UNLABELLED: MicroRNAs (miRNAs) are single-stranded small RNA molecules that regulate various cellular processes. miRNA 155 (miR-155) regulates various aspects of innate and adaptive immune responses and plays a key role in various viral infections and the resulting neuroinflammation. The present study evaluated the involvement of miR-155 in modulating Japanese encephalitis virus (JEV)-induced neuroinflammation. We observed that miR-155 expression was upregulated during JEV infection of mouse primary microglia, the BV-2 microglia cell line, and in both mouse and human brains. In vitro and in vivo knockdown of miR-155 minimized JEV-induced inflammatory responses. In the present study, we confirmed targeting of the Src homology 2-containing inositol phosphatase 1 (SHIP1) 3' untranslated region (UTR) by miR-155 in the context of JEV infection. Inhibition of SHIP1 by miR-155 resulted in higher beta interferon (IFN-ß) and proinflammatory cytokine production through activation of TANK-binding kinase 1 (TBK-1). Based on these observations, we conclude that miR-155 modulates the neuroinflammatory response during JEV infection via negative regulation of SHIP1 expression. Thus, modulation of miR-155 could be a novel strategy to regulate JEV-induced neuroinflammation. IMPORTANCE: Japanese encephalitis virus (JEV), a member of the family Flaviviridae that causes Japanese encephalitis (JE), is the most common mosquito-borne encephalitis virus in the Asia-Pacific region. The disease is feared, as currently there are no specific antiviral drugs available. JEV targets the central nervous system, leading to high mortality and neurological and psychiatric sequelae in some of those who survive. The level of inflammation correlates well with the clinical outcome in patients. Recently, microRNA (miRNA), a single-stranded noncoding RNA, has been implicated in various brain disorders. The present study investigates the role of miRNA in JEV-induced neuroinflammation. Our results show that miRNA 155 (miR-155) targets the Src homology 2-containing inositol phosphatase 1 (SHIP1) protein and promotes inflammation by regulating the NF-κB pathway, increasing the expression of various proinflammatory cytokines and the antiviral response. Thus, miR-155 is a potential therapeutic target to develop antivirals in JE and other brain disorders where inflammation plays a significant role in disease progression.


Asunto(s)
Virus de la Encefalitis Japonesa (Especie)/inmunología , Encefalitis Japonesa/inmunología , Encefalitis Japonesa/patología , Interacciones Huésped-Patógeno , MicroARNs/metabolismo , Monoéster Fosfórico Hidrolasas/biosíntesis , Animales , Encéfalo/patología , Células Cultivadas , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Inositol Polifosfato 5-Fosfatasas , Ratones , Ratones Endogámicos BALB C , Microglía/inmunología , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas
5.
J Neurochem ; 129(1): 143-54, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24236890

RESUMEN

Japanese encephalitis virus (JEV), a single-stranded RNA (ssRNA) virus, is the leading cause of encephalitis in Asia. Microglial activation is one of the key events in JEV-induced neuroinflammation. Although the various microRNAs (miRNAs) has been shown to regulate microglia activation during pathological conditions including neuroviral infections, till date, the involvement of miRNAs in JEV infection has not been evaluated. Hence, we sought to evaluate the possible role of miRNAs in mediating JEV-induced microglia activation. Initial screening revealed significant up-regulation of miR-29b in JEV-infected mouse microglial cell line (BV-2) and primary microglial cells. Furthermore, using bioinformatics tools, we identified tumor necrosis factor alpha-induced protein 3, a negative regulator of nuclear factor-kappa B signaling as a potential target of miR-29b. Interestingly, in vitro knockdown of miR-29b resulted in significant over-expression of tumor necrosis factor alpha-induced protein 3, and subsequent decrease in nuclear translocation of pNF-κB. JEV infection in BV-2 cell line elevated inducible nitric oxide synthase, cyclooxygenase-2, and pro-inflammatory cytokine expression levels, which diminished after miR-29b knockdown. Collectively, our study demonstrates involvement of miR-29b in regulating JEV- induced microglial activation.


Asunto(s)
Cisteína Endopeptidasas/biosíntesis , Virus de la Encefalitis Japonesa (Especie) , Encefalitis Japonesa/metabolismo , Péptidos y Proteínas de Señalización Intracelular/biosíntesis , MicroARNs/fisiología , Microglía/metabolismo , Microglía/virología , Animales , Animales Recién Nacidos , Encefalitis Japonesa/genética , Femenino , Masculino , Ratones , Ratones Endogámicos BALB C , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa
6.
Inflamm Res ; 63(8): 657-65, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24781802

RESUMEN

OBJECTIVE AND DESIGN: The present study was aimed to evaluate the anti-inflammatory potentials of Vespa tropica (VT) venom and its isolated peptides. Effects of whole venom and its two peptides (Vt1512 and Vt1386) on lipopolysaccharide (LPS) challenged BV-2 murine microglial cells was evaluated. MATERIALS: Mouse microglial cell line, BV-2 and crude venom extract as well as purified peptides from VT along with LPS from Salmonella enterica were used for the studies. TREATMENT: BV-2 cells were treated with 500 ng/ml of LPS and different doses of crude wasp venom as well as purified peptides. METHODS: We used immunoblotting, cytokine bead arrays and fluorescence activated cell sorter (FACS) to evaluate the levels of various proteins, cytokines and reactive oxygen species (ROS). RESULTS: Our studies suggest that treatment with whole venom significantly reduces oxidative stress and LPS-stimulated activation of microglia. Also, purified peptides from crude venom exhibited potential anti-inflammatory properties. Further, whole venom was found to be targeting Akt and p38 MAPK pathways, leading to suppressed NF-κB phosphorylation in LPS challenged BV-2 cells. CONCLUSIONS: VT venom possesses anti-inflammatory properties and can be further explored for their therapeutic potential in treating various inflammatory conditions of the central nervous system (CNS).


Asunto(s)
Antiinflamatorios/farmacología , Citocinas/metabolismo , Microglía/efectos de los fármacos , FN-kappa B/metabolismo , Venenos de Avispas/farmacología , Animales , Línea Celular , Ciclooxigenasa 2/metabolismo , Lipopolisacáridos , Ratones , Microglía/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Avispas
7.
J Neuroinflammation ; 9: 57, 2012 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-22429472

RESUMEN

BACKGROUND: Neuroinflammation occurs as a result of microglial activation in response to invading micro-organisms or other inflammatory stimuli within the central nervous system. According to our earlier findings, Krüppel-like factor 4 (Klf4), a zinc finger transcription factor, is involved in microglial activation and subsequent release of proinflammatory cytokines, tumor necrosis factor alpha, macrophage chemoattractant protein-1 and interleukin-6 as well as proinflammatory enzymes, inducible nitric oxide synthase and cyclooxygenase-2 in lipopolysaccharide-treated microglial cells. Our current study focuses on finding the molecular mechanism of the anti-inflammatory activities of honokiol in lipopolysaccharide-treated microglia with emphasis on the regulation of Klf4. METHODS: For in vitro studies, mouse microglial BV-2 cell lines as well as primary microglia were treated with 500 ng/mL lipopolysaccharide as well as 1 µM and 10 µM of honokiol. We cloned full-length Klf4 cDNA in pcDNA3.1 expression vector and transfected BV-2 cells with this construct using lipofectamine for overexpression studies. For in vivo studies, brain tissues were isolated from BALB/c mice treated with 5 mg/kg body weight of lipopolysaccharide either with or without 2.5 or 5 mg/kg body weight of honokiol. Expression of Klf4, cyclooxygenase-2, inducible nitric oxide synthase and phospho-nuclear factor-kappa B was measured using immunoblotting. We also measured the levels of cytokines, reactive oxygen species and nitric oxide in different conditions. RESULTS: Our findings suggest that honokiol can substantially downregulate the production of proinflammatory cytokines and inflammatory enzymes in lipopolysaccharide-stimulated microglia. In addition, honokiol downregulates lipopolysaccharide-induced upregulation of both Klf4 and phospho-nuclear factor-kappa B in these cells. We also found that overexpression of Klf4 in BV-2 cells suppresses the anti-inflammatory action of honokiol. CONCLUSIONS: Honokiol potentially reduces inflammation in activated microglia in a Klf4-dependent manner.


Asunto(s)
Inflamación/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Microglía/efectos de los fármacos , Animales , Línea Celular Transformada , Ciclooxigenasa 2/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Inflamación/inducido químicamente , Factor 4 Similar a Kruppel , Ratones , Ratones Endogámicos BALB C , FN-kappa B/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Polisacáridos/efectos adversos , Polisacáridos/farmacología , Especies Reactivas de Oxígeno/metabolismo
8.
J Neurosci Res ; 88(8): 1615-31, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20127816

RESUMEN

Neuroinflammation is a complex innate response of neural tissue against harmful effects of diverse stimuli viz., pathogens, damaged cells and irritants within the Central Nervous System (CNS). Studies show that multiple inflammatory mediators including cytokines, chemokines and prostaglandins are elevated in the Cerebrospinal Fluid (CSF) and in post-mortem brain tissues of patients with history of neuroinflammatory conditions as well as neurodegenerative disorders like Alzheimer's disease, Parkinson's disease and Multiple Sclerosis. The innate immunity mediators in the brain, namely microglia and astrocytes, express certain Pattern Recognition Receptors (PRRs), which are always on 'high-alert' for pathogens or other inflammatory triggers and participate in the assembly and activation of the inflammasome. The inflammasome orchestrates the activation of the precursors of proinflammatory caspases, which in turn, cleave the precursor forms of interleukin-1beta, IL-18 and IL-33 into their active forms; the secretion of which leads to a potent inflammatory response, and/or influences the release of toxins from glial and endothelial cells. Altered expression of inflammasome mediators can either promote or inhibit neurodegenerative processes. Therefore, modulating the inflammasome machinery seems a better combat strategy than summarily suppressing all inflammation in most neuroinflammatory conditions. In the current review we have surveyed the identified triggers and pathways of inflammasome activation and the following events which ultimately accomplish the innate inflammatory response in the CNS, with a goal to provide an analytical insight into disease pathogenesis that might provide cues for devising novel therapeutic strategies.


Asunto(s)
Enfermedades del Sistema Nervioso Central , Inflamación , Complejos Multiproteicos/metabolismo , Transducción de Señal/fisiología , Animales , Enfermedades del Sistema Nervioso Central/inmunología , Enfermedades del Sistema Nervioso Central/metabolismo , Enfermedades del Sistema Nervioso Central/patología , Humanos , Sistema Inmunológico/fisiología , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Modelos Biológicos
9.
J Neurochem ; 110(3): 1070-81, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19500213

RESUMEN

4-Methylnitrosamino-1-(3-pyridyl)-1-butanone (NNK) is a tobacco-specific procarcinogen. We have investigated whether NNK causes inflammatory upheaval in the brain by activation of resident microglia and astrocyte and result in bystander neuronal damage. We have carried out the work in both in vitro and in vivo models. We have found that treatment with NNK causes significant activation of mouse microglial (BV2) cell line as evident by increase in reactive oxygen species and nitric oxide level. Western blot analysis has showed increase in proinflammatory signaling proteins, proinflammatory effector proteins, and other stress-related proteins. Interestingly, increased levels of proinflammatory cytokines like interleukin (IL)-6, tumor necrosis factor-alpha, monocyte chemoattractant protein 1 (MCP1), and IL-12p70 are also detected. Work from our in vivo studies has demonstrated similar increase in proinflammatory signaling and effector molecules along with the proinflammatory cytokine levels, following NNK treatment. Immunohistochemical staining of the brain sections of NNK-treated mice reveals massive microglial and astrocyte activation along with distinct foci of neuronal damage. Both in vitro and in vivo results provide strong indication that NNK causes significant upheaval of the inflammatory condition of brain and inflicts subsequent neuronal damage.


Asunto(s)
Carcinógenos/toxicidad , Microglía/efectos de los fármacos , Microglía/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Nicotiana/toxicidad , Animales , Línea Celular , Línea Celular Tumoral , Femenino , Masculino , Ratones , Ratones Endogámicos BALB C , Microglía/patología , Neuronas/patología , Nitrosaminas/toxicidad
10.
J Clin Invest ; 129(8): 3277-3292, 2019 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-31112527

RESUMEN

The migration of leukocytes into the CNS drives the neuropathology of multiple sclerosis (MS). This penetration likely utilizes energy resources that remain to be defined. Using the experimental autoimmune encephalomyelitis (EAE) model of MS, we determined that macrophages within the perivascular cuff of post-capillary venules are highly glycolytic as manifested by strong expression of lactate dehydrogenase A (LDHA) that converts pyruvate to lactate. These macrophages expressed prominent levels of monocarboxylate transporter-4 (MCT-4) specialized in secreting lactate from glycolytic cells. The functional relevance of glycolysis was confirmed by siRNA-mediated knockdown of LDHA and MCT-4, which decreased lactate secretion and macrophage transmigration. MCT-4 was in turn regulated by EMMPRIN (CD147) as determined through co-expression/co-immunoprecipitation studies, and siRNA-mediated EMMPRIN silencing. The functional relevance of MCT-4/EMMPRIN interaction was affirmed by lower macrophage transmigration in culture using the MCT-4 inhibitor, α-cyano-4-hydroxy-cinnamic acid (CHCA), a cinnamon derivative. CHCA also reduced leukocyte infiltration and the clinical severity of EAE. Relevance to MS was corroborated by the strong expression of MCT-4, EMMPRIN and LDHA in perivascular macrophages in MS brains. These results detail the metabolism of macrophages for transmigration from perivascular cuffs into the CNS parenchyma and identifies CHCA and diet as potential modulators of neuro-inflammation in MS.


Asunto(s)
Encéfalo/metabolismo , Movimiento Celular , Encefalomielitis Autoinmune Experimental/metabolismo , Glucólisis , Macrófagos/metabolismo , Esclerosis Múltiple/metabolismo , Animales , Basigina/metabolismo , Encéfalo/patología , Femenino , L-Lactato Deshidrogenasa/metabolismo , Macrófagos/patología , Ratones , Transportadores de Ácidos Monocarboxílicos/metabolismo , Esclerosis Múltiple/patología , Proteínas Musculares/metabolismo
11.
J Leukoc Biol ; 98(1): 33-48, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25977287

RESUMEN

EMMPRIN (CD147), originally described as an inducer of the expression of MMPs, has gained attention in its involvement in various immunologic diseases, such that anti-EMMPRIN antibodies are considered as potential therapeutic medications. Given that MMPs are involved in the pathogenesis of various disease states, it is relevant that targeting an upstream inducer would make for an effective therapeutic strategy. Additionally, EMMPRIN is now appreciated to have multiple roles apart from MMP induction, including in cellular functions, such as migration, adhesion, invasion, energy metabolism, as well as T cell activation and proliferation. Here, we review what is known about EMMPRIN in numerous immunologic/inflammatory disease conditions with a particular focus on its complex roles in T cell biology.


Asunto(s)
Basigina/fisiología , Enfermedades del Sistema Inmune/fisiopatología , Linfocitos T/fisiología , Animales , Basigina/genética , Humanos , Enfermedades del Sistema Inmune/inmunología , Ratones , Ratones Noqueados
12.
Matrix Biol ; 44-46: 138-46, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25644103

RESUMEN

Matrix metalloproteinases (MMPs) are engaged in pathologies associated with infections, tumors, autoimmune disorders and neurological dysfunctions. With the identification of an upstream regulator of MMPs, EMMPRIN (Extracellular matrix metalloproteinase inducer, CD147), it is relevant to address if EMMPRIN plays a role in the pathology of central nervous system (CNS) diseases. This would enable the possibility of a more upstream and effective therapeutic target. Indeed, conditions including gliomas, Alzheimer's disease (AD), multiple sclerosis (MS), and other insults such as hypoxia/ischemia show elevated levels of EMMPRIN which correlate with MMP production. In contrast, given EMMPRIN's role in CNS homeostasis with respect to regulation of monocarboxylate transporters (MCTs) and interactions with adhesion molecules including integrins, we need to consider that EMMPRIN may also serve important regulatory or protective functions. This review summarizes the current understanding of EMMPRIN's involvement in CNS homeostasis, its possible roles in escalating or reducing neural injury, and the mechanisms of EMMPRIN including and apart from MMP induction.


Asunto(s)
Basigina/metabolismo , Sistema Nervioso Central/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Basigina/efectos de los fármacos , Moléculas de Adhesión Celular/metabolismo , Sistema Nervioso Central/patología , Regulación Enzimológica de la Expresión Génica , Homeostasis , Humanos , Terapia Molecular Dirigida , Transportadores de Ácidos Monocarboxílicos/metabolismo
13.
Neurotoxicology ; 41: 143-53, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24530660

RESUMEN

Lead is one of the major pollutants of environment and is highly toxic to the functioning of central nervous system (CNS). The chronic exposure of this heavy metal is debilitating to the functional behavior of an organism. Studies have shown that acute exposure to Pb can lead to glial activation and secretion of cyto-chemokines in both in vitro and in vivo models. However, the cellular source of secretion of these cyto-chemokines remains to be identified. Microglia are monocytes of the brain, and are primary source of cytokine secretion in the CNS. We hypothesized that microglia exposed to Pb can secrete cyto-chemokines, thereby resulting in subsequent neuronal death. Our studies show that stimulation of BV-2 mouse microglia with 10µÐœ dose of Pb resulted in up-regulation of extracellular signal-regulated kinase (ERK) and protein kinase B (Akt) pathways, along with activation of an important transcription factor, nuclear factor-κB (NF-κB). Further, we found that the levels of tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6) and monocyte chemoattractant protein-1 (MCP-1), and cyclooxygenase-2 (COX-2) pro-inflammatory enzyme were increased in response to Pb exposure. Furthermore, treatment with conditioned media from Pb treated BV-2 cells lead to neuronal death in neuroblastoma cells, which potentially involved the activation of caspase-3 enzyme. In all, the current study brings forth critical involvement of microglial activation in mediating the neurotoxicity associated with lead exposure.


Asunto(s)
Microglía/efectos de los fármacos , Neuronas/fisiología , Compuestos Organometálicos/farmacología , Análisis de Varianza , Animales , Caspasa 3/metabolismo , Muerte Celular/efectos de los fármacos , Línea Celular Transformada , Supervivencia Celular/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Ciclooxigenasa 2/metabolismo , Citocinas/metabolismo , Relación Dosis-Respuesta a Droga , MAP Quinasa Quinasa Quinasa 5/metabolismo , Ratones , Peso Molecular , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
14.
CNS Neurol Disord Drug Targets ; 12(6): 726-40, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24070095

RESUMEN

Inflammation plays a critical role in the progression of neurodegenerative diseases. Microglia are the resident macrophages of the central nervous system (CNS) which actively take part in the neuronal development of CNS and are involved in clearance of pathogens as well as cellular debris from the system upon insult to this organization. Chronic activation of microglia in neurodegenerative disorders such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), amyotrophic lateral sclerosis (ALS) as well as inflammatory conditions of CNS such as multiple sclerosis (MS) results in overall upregulation of pro-inflammatory cytokines and chemokines in the brain parenchyma. This compromises the neuronal health which further activates microglia by releasing death associated molecules such as neuromelanin, Aß peptides and cellular debris at the lesion site thereby forming a vicious cycle of disease advancement. Targeting microglial activation has proven to be a viable option in the treatment of inflammation related neurodegenerative diseases. This review will discuss the central position of inflammation and therapeutic strategies aiming to alleviate disease progression in some of the important inflammatory conditions of CNS.


Asunto(s)
Encéfalo/patología , Microglía/patología , Microglía/fisiología , Enfermedades Neurodegenerativas/patología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Encéfalo/metabolismo , Humanos , Mediadores de Inflamación/fisiología , Microglía/metabolismo , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/patología , Enfermedades Neurodegenerativas/metabolismo , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología
15.
Methods Mol Biol ; 1041: 71-82, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23813371

RESUMEN

Cytokine measurement is a prerequisite to understand the inflammatory state of the body. Quantitative analysis of cytokines by Western blotting and ELISA is a daunting task as these are time-consuming and error-prone protocols. With the advent of flow cytometry, the estimation of cytokines using the classical antigen-antibody reaction has become a popular choice with researchers/clinicians. Here, we describe a protocol for multiple cytokine analysis using flow cytometry.


Asunto(s)
Citocinas/metabolismo , Microglía/metabolismo , Animales , Western Blotting , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Ratones , Microglía/citología
16.
PLoS One ; 7(2): e32270, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22393394

RESUMEN

BACKGROUND: Japanese Encephalitis virus (JEV) is a common cause of acute and epidemic viral encephalitis. JEV infection is associated with microglial activation resulting in the production of pro-inflammatory cytokines including Interleukin-1 ß (IL-1ß) and Interleukin-18 (IL-18). The Pattern Recognition Receptors (PRRs) and the underlying mechanism by which microglia identify the viral particle leading to the production of these cytokines is unknown. METHODOLOGY/PRINCIPAL FINDINGS: For our studies, we have used murine model of JEV infection as well as BV-2 mouse microglia cell line. In this study, we have identified a signalling pathway which leads to the activation of caspase-1 as the key enzyme responsible for the maturation of both IL-1ß and IL-18 in NACHT, LRR and PYD domains-containing protein-3 (NLRP3) dependent manner. Depletion of NLRP3 results in the reduction of caspase-1 activity and subsequent production of these cytokines. CONCLUSION/SIGNIFICANCE: Our results identify a mechanism mediated by Reactive Oxygen Species (ROS) production and potassium efflux as the two danger signals that link JEV infection to caspase-1 activation resulting in subsequent IL-1ß and IL-18 maturation.


Asunto(s)
Proteínas Portadoras/genética , Proteínas Portadoras/fisiología , Virus de la Encefalitis Japonesa (Especie)/metabolismo , Encefalitis Japonesa/genética , Encefalitis Japonesa/virología , Animales , Caspasa 1/metabolismo , Línea Celular , Citocinas/metabolismo , Inflamación , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Ratones , Ratones Endogámicos BALB C , Microglía/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR , Estructura Terciaria de Proteína , Especies Reactivas de Oxígeno , Transducción de Señal
17.
Front Biosci (Landmark Ed) ; 16(6): 2187-205, 2011 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-21622170

RESUMEN

Microglia, the resident macrophages of the Central Nervous System (CNS) mediate key innate immune responses against foreign invasions within the CNS and clear the debris after any damage to the nearby tissue. Blood Brain Barrier (BBB) segregates the CNS from the rest of the lymphatic system and prevents the entry of foreign molecules into the brain. Pathogens still cross the BBB via different mechanisms and can cause severe infections of the CNS. Viral encephalitis is the most common form of brain infection and the causative agents include Japanese Encephalitis Virus (JEV), West Nile Virus (WNV), Murray Valley Encephalitis Virus (MVEV), Herpes Simplex Virus (HSV), Human Immunodeficiency Virus (HIV), Cytomegalovirus (CMV) and Hepatitis C Virus (HCV) among several others. Microglia expresses various Pattern Recognition Receptors (PRRs) to identify viral signatures called Pathogen Associated Molecular Patterns (PAMPs) to which microglia respond by releasing several pro and anti-inflammatory cytokines like MCP1, IL-1beta, Type I IFN, IFN-gamma, TNF-alpha etc. This review discusses the various viral infections of the brain and strategies employed by microglia to detect them.


Asunto(s)
Encefalitis Viral/virología , Microglía/inmunología , Microglía/virología , Complejo SIDA Demencia/inmunología , Complejo SIDA Demencia/virología , Barrera Hematoencefálica/inmunología , Barrera Hematoencefálica/virología , Citocinas/biosíntesis , Infecciones por Citomegalovirus/inmunología , Infecciones por Citomegalovirus/virología , Virus de la Encefalitis del Valle Murray , Encefalitis por Arbovirus/inmunología , Encefalitis por Arbovirus/virología , Encefalitis por Herpes Simple/inmunología , Encefalitis por Herpes Simple/virología , Encefalitis Japonesa/inmunología , Encefalitis Japonesa/virología , Encefalitis Viral/inmunología , VIH-1 , Hepatitis C/inmunología , Hepatitis C/virología , Herpesvirus Humano 1 , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Innata , Modelos Neurológicos , Fiebre del Nilo Occidental/inmunología , Fiebre del Nilo Occidental/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA