Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 126
Filtrar
1.
Neurosci Lett ; 771: 136467, 2022 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-35063502

RESUMEN

The inflammatory response related to surgery is considered surgical inflammation. Most anesthetic agents directly or indirectly suppress the immune response. However, the intravenous anesthetics pentobarbital and ketamine were reported to inhibit the lipopolysaccharide-induced inflammatory response such as cytokines formation. Neurogenic inflammation is inflammation originating from the local release of inflammatory mediators, such as substance P (SP), by primary afferent neurons after noxious stimuli like surgery. Thus, in this study, we examined whether pentobarbital and ketamine suppress SP release from cultured dorsal root ganglion (DRG) neurons. DRG cells were dissected from male Wistar rats. Released SP was measured by radioimmunoassay. We demonstrated that higher concentrations of pentobarbital (100-1,000 µM) significantly inhibited capsaicin (100 nM)-induced, but not high K+ (50 mM)-induced, SP release from DRG cells, although a high concentration of ketamine (1 mM) did not. This study revealed that pentobarbital functions between the activation of vanilloid receptor subtype 1 (TRPV1) receptors, to which capsaicin selectively binds, and the opening of voltage-operated Ca2+ channels (VOCC) in the nerve endings. Therefore, the anti-inflammatory action of pentobarbital is mediated through different mechanisms than those of ketamine. Thus, the inhibitory effect of pentobarbital on SP release from peripheral terminals may protect against neurogenic inflammation after surgery.


Asunto(s)
Antiinflamatorios/uso terapéutico , Inflamación Neurogénica/tratamiento farmacológico , Pentobarbital/uso terapéutico , Nervios Periféricos/metabolismo , Sustancia P/metabolismo , Analgésicos/farmacología , Animales , Antiinflamatorios/farmacología , Canales de Calcio/metabolismo , Capsaicina/farmacología , Células Cultivadas , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/metabolismo , Ketamina/farmacología , Masculino , Inflamación Neurogénica/metabolismo , Pentobarbital/farmacología , Nervios Periféricos/efectos de los fármacos , Ratas , Ratas Wistar , Fármacos del Sistema Sensorial/farmacología , Canales Catiónicos TRPV/metabolismo
2.
Int Immunopharmacol ; 98: 107868, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34153665

RESUMEN

Microglial overactivation-mediated neuroinflammation contributes greatly to the pathogenesis of neurodegenerative diseases, such as Parkinson's disease. Macrophage migration inhibitory factor (MIF) is a pleiotropic proinflammatory cytokine that is involved in the pathophysiology of various inflammatory diseases by inducing various proinflammatory cytokines. Compound 3-({[4-(4-methoxyphenyl)-6-methyl-2-pyrimidinyl]thio}methyl)benzoic acid (Z-312) is a novel small -molecule inhibitor of MIF tautomeric activity. In this study, we investigated the anti-inflammatory effects of Z-312 on liposaccharide (LPS)-induced neuroinflammation in vitro and in vivo. The results showed that Z-312 significantly decreased the production of nitric oxide (NO), interleukin (IL)-1ß, tumor necrosis factor (TNF)-α and IL-6 in LPS-stimulated microglial cells. Mechanistically, nuclear translocation of the p65 subunit of nuclear factor (NF)-κB, degradation and phosphorylation of IκBα, NF-κB transcriptional activity and phosphorylation of p38 mitogen-activated protein kinase (MAPK) and JNK were markedly attenuated by pretreatment with Z-312 in BV-2 microglial cells. In addition, Z-312 suppressed the neurotoxic effects of cell culture medium of LPS-activated BV-2 microglia on cocultured mouse HT22 neuroblastoma cells. An in vivo study demonstrated that Z-312 markedly ameliorated microglial activation and subsequent DA neuron loss in an LPS-induced Parkinson's disease (PD) mouse model. These results suggest that MIF inhibitor Z-312 may be a promising neuroprotective agent for the treatment of neuroinflammation-mediated neurological diseases.


Asunto(s)
Antiinflamatorios/uso terapéutico , Ácido Benzoico/uso terapéutico , Factores Inhibidores de la Migración de Macrófagos/antagonistas & inhibidores , Microglía/metabolismo , Inflamación Neurogénica/tratamiento farmacológico , Enfermedad de Parkinson/tratamiento farmacológico , Animales , Ácido Benzoico/química , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Lipopolisacáridos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/patología , FN-kappa B/metabolismo , Óxido Nítrico/metabolismo , Transducción de Señal
3.
Bull Exp Biol Med ; 171(2): 212-217, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-34173095

RESUMEN

Using rat and mouse models of neurogenic, ethanol-induced, and indometacin-induced damage to the gastric mucosa we demonstrated that course preventive treatment with flavonoid complex from aerial parts of Lychnis chalcedonica L. increased the resistance of gastric mucosa to ulcerogenic factors of different etiology. The gastroprotective effect of the phytocomplex in a dose range of 16-1600 µg/kg was comparable with that of the reference drug plantaglucide and was superior to that of the reference drugs eleutherococcus extract and methyluracil in the therapeutic doses. The antiulcerogenic activity of Lychnis chalcedonica flavonoid complex considerably exceeded activity of Lychnis chalcedonica L. extract demonstrated in our previous experiments.


Asunto(s)
Antiulcerosos/uso terapéutico , Extractos Vegetales/uso terapéutico , Úlcera Gástrica/tratamiento farmacológico , Animales , Animales no Consanguíneos , Antiulcerosos/aislamiento & purificación , Antiulcerosos/farmacología , Citoprotección/efectos de los fármacos , Modelos Animales de Enfermedad , Etanol , Flavonoides/aislamiento & purificación , Flavonoides/farmacología , Flavonoides/uso terapéutico , Mucosa Gástrica/efectos de los fármacos , Mucosa Gástrica/patología , Indometacina , Lychnis/química , Masculino , Ratones , Inflamación Neurogénica/tratamiento farmacológico , Inflamación Neurogénica/patología , Fitoterapia , Extractos Vegetales/farmacología , Ratas , Ratas Wistar , Silene , Úlcera Gástrica/inducido químicamente , Úlcera Gástrica/patología
4.
J Exp Med ; 218(4)2021 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-33620419

RESUMEN

Despite the development of effective therapies, a substantial proportion of asthmatics continue to have uncontrolled symptoms, airflow limitation, and exacerbations. Transient receptor potential cation channel member A1 (TRPA1) agonists are elevated in human asthmatic airways, and in rodents, TRPA1 is involved in the induction of airway inflammation and hyperreactivity. Here, the discovery and early clinical development of GDC-0334, a highly potent, selective, and orally bioavailable TRPA1 antagonist, is described. GDC-0334 inhibited TRPA1 function on airway smooth muscle and sensory neurons, decreasing edema, dermal blood flow (DBF), cough, and allergic airway inflammation in several preclinical species. In a healthy volunteer Phase 1 study, treatment with GDC-0334 reduced TRPA1 agonist-induced DBF, pain, and itch, demonstrating GDC-0334 target engagement in humans. These data provide therapeutic rationale for evaluating TRPA1 inhibition as a clinical therapy for asthma.


Asunto(s)
Asma/tratamiento farmacológico , Inflamación Neurogénica/tratamiento farmacológico , Dolor/tratamiento farmacológico , Prurito/tratamiento farmacológico , Piridinas/farmacología , Piridinas/uso terapéutico , Pirimidinas/farmacología , Pirimidinas/uso terapéutico , Canal Catiónico TRPA1/antagonistas & inhibidores , Adolescente , Adulto , Animales , Estudios de Cohortes , Modelos Animales de Enfermedad , Perros , Método Doble Ciego , Femenino , Cobayas , Voluntarios Sanos , Humanos , Isotiocianatos/administración & dosificación , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Dolor/inducido químicamente , Prurito/inducido químicamente , Ratas , Ratas Sprague-Dawley , Canal Catiónico TRPA1/deficiencia , Resultado del Tratamiento , Adulto Joven
5.
Br J Pharmacol ; 177(19): 4386-4399, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33289534

RESUMEN

A significant number of cannabinoids are known to have analgesic and anti-inflammatory properties in various diseases. Due to their presynaptic/terminal location, cannabinoid receptors can inhibit synaptic transmission and have the potential to regulate neurogenic inflammation. Neurogenic inflammation occurs when a noxious signal is detected in the periphery initiating an antidromic axon reflex in the same sensory neurone leading to depolarization of the afferent terminal. Neuropeptides are subsequently released and contribute to vasodilation, plasma extravasation and modulation of immune cells. Endocannabinoids, synthetic cannabinoids and phytocannabinoids can reduce neuroinflammation by inhibiting afferent firing and inflammatory neuropeptide release. Thus, in addition to a direct effect on vascular smooth muscle and inflammatory cells, cannabinoids can reduce inflammation by silencing small diameter neurones. This review examines the neuropharmacological processes involved in regulating antidromic depolarization of afferent nerve terminals by cannabinoids and the control of neurogenic inflammation in different diseases.


Asunto(s)
Cannabinoides , Cannabinoides/farmacología , Endocannabinoides , Humanos , Inflamación/tratamiento farmacológico , Inflamación Neurogénica/tratamiento farmacológico , Neuronas Aferentes , Receptores de Cannabinoides
6.
Front Immunol ; 11: 571844, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33193354

RESUMEN

Multiple sclerosis (MS) is an inflammatory and demyelinating disease of the central nervous system (CNS). The persistent inflammation is being mainly attributed to local oxidative stress and inflammasome activation implicated in the ensuing demyelination and axonal damage. Since new control measures remain necessary, we evaluated the preventive and therapeutic potential of a beta-selenium-lactic acid derivative (LAD-ßSe), which is a source of organic selenium under development, to control experimental autoimmune encephalomyelitis (EAE) that is an animal model for MS. Two EAE murine models: C57BL/6 and SJL/J immunized with myelin oligodendrocyte glycoprotein and proteolipid protein, respectively, and a model of neurodegeneration induced by LPS in male C57BL/6 mice were used. The preventive potential of LAD-ßSe was initially tested in C57BL/6 mice, the chronic MS model, by three different protocols that were started 14 days before or 1 or 7 days after EAE induction and were extended until the acute disease phase. These three procedures were denominated preventive therapy -14 days, 1 day, and 7 days, respectively. LAD-ßSe administration significantly controlled clinical EAE development without triggering overt hepatic and renal dysfunction. In addition of a tolerogenic profile in dendritic cells from the mesenteric lymph nodes, LAD-ßSe also downregulated cell amount, activation status of macrophages and microglia, NLRP3 (NOD-like receptors) inflammasome activation and other pro-inflammatory parameters in the CNS. The high Se levels found in the CNS suggested that the product crossed the blood-brain barrier having a possible local effect. The hypothesis that LAD-ßSe was acting locally was then confirmed by using the LPS-induced neurodegeneration model that also displayed Se accumulation and downmodulation of pro-inflammatory parameters in the CNS. Remarkably, therapy with LAD-ßSe soon after the first remitting episode in SJL/J mice, also significantly downmodulated local inflammation and clinical disease severity. This study indicates that LAD-ßSe, and possibly other derivatives containing Se, are able to reach the CNS and have the potential to be used as preventive and therapeutic measures in distinct clinical forms of MS.


Asunto(s)
Antiinflamatorios/uso terapéutico , Sistema Nervioso Central/efectos de los fármacos , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Inflamasomas/metabolismo , Microglía/patología , Esclerosis Múltiple/tratamiento farmacológico , Inflamación Neurogénica/tratamiento farmacológico , Selenio/uso terapéutico , Animales , Sistema Nervioso Central/patología , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inmunología , Humanos , Ácido Láctico/química , Masculino , Ratones , Ratones Endogámicos C57BL , Esclerosis Múltiple/inmunología , Glicoproteína Mielina-Oligodendrócito/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Inflamación Neurogénica/inmunología , Selenio/química
7.
J Tradit Chin Med ; 40(5): 792-802, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33000580

RESUMEN

OBJECTIVE: To investigate the efficacy of active compounds of Chanqin (CQ) granules on PM2.5-induced airway neurogenic inflammation in vivo, and to elucidate the underlying mechanisms of action. METHODS: The Traditional Chinese Medicine systems pharmacology (TCMSP) database was searched, and the results were combined with oral bioavailability and drug analysis to identify the compounds in CQ granules. The pharmacophore modeling approach was used to predict the compound targets, and the diseases corresponding to the targets were obtained by searching the therapeutic target database (TTD), pharmacogenomics knowledgebase (PharmGKB) and DrugBank databases. Cytoscape software was used to construct the network pharmacological charts for Component-Target and Target-Disease interactions of the CQ granules. Then, the mechanisms of action and effectiveness of CQ granules for the treatment of PM2.5-induced airway neurogenic inflammation were analyzed. RESULTS: A total of 195 compounds and 171 targets were obtained from the analyses. A total of 569 corresponding diseases were identified for these targets. Component-target and target-disease networks were constructed. The possible mechanisms and effective components in CQ granules for treating airway neurogenic inflammation were analyzed. Quercetin, kaempferol and isorhamnetin, beta-sitosterol and sitosterol, which are typically found in the formulation, have extensive pharmacological activities, including anti-inflammatory, antioxidant and antiviral actions and neuroprotective properties. Among these targets, androgen receptor, estrogen receptor, prostaglandin G/H synthase 2, and inducible nitric oxide synthase play important pathological roles, including the induction of neurogenic inflammation. CQ granules may have therapeutic effectiveness for numerous diseases in addition to respiratory diseases, including neoplasms, digestive system diseases, cardiovascular diseases, respiratory tract diseases and nervous system diseases. In vivo, CQ granules are effective in treating pulmonary inflammation and downregulate neuropeptides in the bronchoalveolar lavage fluid after PM2.5 exposure. CQ granules significantly decreased the levels of neurokinin A, neurokinin B and calcitonin gene-related peptide in the lung and dorsal root ganglia. CQ also significantly suppressed the upregulation of p-extracellular regulated protein kinase 1/2 and p-methyl ethyl ketone 1/2 induced by PM2.5 exposure. CONCLUSION: CQ granules have potential for the treatment of neurogenic inflammation induced by PM2.5 in vivo, and the mechanism might involve downregulation of neuropeptides in the BALF, lung and dorsal root ganglia.


Asunto(s)
Medicamentos Herbarios Chinos/administración & dosificación , Medicamentos Herbarios Chinos/química , Inflamación Neurogénica/tratamiento farmacológico , Material Particulado/toxicidad , Animales , Humanos , Masculino , Inflamación Neurogénica/etiología , Inflamación Neurogénica/genética , Inflamación Neurogénica/metabolismo , Neuropéptidos/genética , Neuropéptidos/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Neumonía/tratamiento farmacológico , Neumonía/genética , Neumonía/metabolismo , Ratas , Ratas Sprague-Dawley
8.
Nat Immunol ; 21(5): 513-524, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32284594

RESUMEN

Oxidative stress is a central part of innate immune-induced neurodegeneration. However, the transcriptomic landscape of central nervous system (CNS) innate immune cells contributing to oxidative stress is unknown, and therapies to target their neurotoxic functions are not widely available. Here, we provide the oxidative stress innate immune cell atlas in neuroinflammatory disease and report the discovery of new druggable pathways. Transcriptional profiling of oxidative stress-producing CNS innate immune cells identified a core oxidative stress gene signature coupled to coagulation and glutathione-pathway genes shared between a microglia cluster and infiltrating macrophages. Tox-seq followed by a microglia high-throughput screen and oxidative stress gene network analysis identified the glutathione-regulating compound acivicin, with potent therapeutic effects that decrease oxidative stress and axonal damage in chronic and relapsing multiple sclerosis models. Thus, oxidative stress transcriptomics identified neurotoxic CNS innate immune populations and may enable discovery of selective neuroprotective strategies.


Asunto(s)
Encefalomielitis Autoinmune Experimental/genética , Perfilación de la Expresión Génica/métodos , Microglía/fisiología , Esclerosis Múltiple/genética , Inflamación Neurogénica/genética , Animales , Antioxidantes/uso terapéutico , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Femenino , Redes Reguladoras de Genes , Ensayos Analíticos de Alto Rendimiento , Humanos , Inmunidad Innata , Isoxazoles/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Esclerosis Múltiple/tratamiento farmacológico , Inflamación Neurogénica/tratamiento farmacológico , Estrés Oxidativo , Análisis de Secuencia de ARN , Análisis de la Célula Individual
9.
Int Immunopharmacol ; 83: 106491, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32279045

RESUMEN

Oxidative stress acts as the major causative factor for various age-associated neurodegenerative diseases, triggering cognitive and memory impairments. In the present study, the underlying neuroprotective mechanism governing how shikonin acts against D-galactose (D-gal)-induced memory impairment, neuroinflammation and neuron damage was examined. The results revealed that chronic administration of D-gal [150 mg/kg intraperitoneally (i.p.)] in mice caused cognitive and memory impairments, as determined by Morris water-maze test. Shikonin treatment, however, alleviated D-gal-induced memory impairment and reversed the D-gal-induced neural damage and apoptosis. Furthermore, western blotting and the results of morphological analysis revealed that shikonin treatments markedly reduced D-gal induced neuroinflammation through inhibition of astrocytosis as determined by glial fibrillary acidic protein (GFAP) detection, and downregulating other inflammatory mediators, including tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß), and IL-6. Moreover, shikonin treatment led to inhibition of the activation of nuclear factor-κB (NF-κB) and the phosphorylation of mitogen-activated protein kinases (MAPKs), preventing neurodegeneration. Hence, taken together, the results of the present study suggested that shikonin attenuated D-gal-induced memory impairment, neuroinflammation and neurodegeneration, possibly via the NF-κB/mitogen-activated protein kinase (MAPK) pathway. Our data suggest that shikonin could be a promising, endogenous and compatible antioxidant candidate for age-associated neurodegenerative diseases, including Alzheimer's disease.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Naftoquinonas/uso terapéutico , Enfermedades Neurodegenerativas/tratamiento farmacológico , Inflamación Neurogénica/tratamiento farmacológico , Animales , Células Cultivadas , Disfunción Cognitiva , Citocinas/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Galactosa/metabolismo , Humanos , Mediadores de Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Estrés Oxidativo , Transducción de Señal
10.
J Med Chem ; 63(7): 3665-3677, 2020 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-32162512

RESUMEN

TWIK-related K+ (TREK) channels are potential analgesic targets. However, selective activators for TREK with both defined action mechanism and analgesic ability for chronic pain have been lacking. Here, we report (1S,3R)-3-((4-(6-methylbenzo[d]thiazol-2-yl)phenyl)carbamoyl)cyclopentane-1-carboxylic acid (C3001a), a selective activator for TREK, against other two-pore domain K+ (K2P) channels. C3001a binds to the cryptic binding site formed by P1 and TM4 in TREK-1, as suggested by computational modeling and experimental analysis. Furthermore, we identify the carboxyl group of C3001a as a structural determinant for binding to TREK-1/2 and the key residue that defines the subtype selectivity of C3001a. C3001a targets TREK channels in the peripheral nervous system to reduce the excitability of nociceptive neurons. In neuropathic pain, C3001a alleviated spontaneous pain and cold hyperalgesia. In a mouse model of acute pancreatitis, C3001a alleviated mechanical allodynia and inflammation. Together, C3001a represents a lead compound which could advance the rational design of peripherally acting analgesics targeting K2P channels without opioid-like adverse effects.


Asunto(s)
Analgésicos/uso terapéutico , Antiinflamatorios/uso terapéutico , Benzotiazoles/uso terapéutico , Inflamación Neurogénica/tratamiento farmacológico , Dolor/tratamiento farmacológico , Canales de Potasio de Dominio Poro en Tándem/agonistas , Analgésicos/metabolismo , Analgésicos/farmacocinética , Animales , Antiinflamatorios/metabolismo , Antiinflamatorios/farmacocinética , Benzotiazoles/metabolismo , Benzotiazoles/farmacocinética , Sitios de Unión , Ganglios Espinales/efectos de los fármacos , Células HEK293 , Humanos , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Simulación del Acoplamiento Molecular , Estructura Molecular , Pancreatitis/tratamiento farmacológico , Canales de Potasio de Dominio Poro en Tándem/química , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Unión Proteica , Ratas Sprague-Dawley , Relación Estructura-Actividad
11.
Int Immunopharmacol ; 81: 106300, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32070922

RESUMEN

Oxidative stress and neuroinflammation are critically involved in amyloid beta (Aß) induced cognitive impairments. ß-Lapachone (ß-LAP) is a natural activator of NAD(P)H quinone oxidoreductase 1 (NQO1) which has antioxidant and anti-inflammatory properties.This study investigated the effect of ß-LAP administration on Aß-induced memory deficit, oxidative stress, neuroinflammation, and apoptosis cell death in the hippocampus. Forty BALB/c mice were allocated into control, sham, ß-LAP (ßL), Aß, and Aß + ßL groups. Intracerebroventricular injection of Aß1-42 was used to induce Alzheimer's disease (AD) model. Mice in the ßL and Aß + ßL groups were treated with ß-LAP (10 mg/kg, i.p) for 4 days. Results revealed that ß-LAP attenuated memory impairment in the Aß-received mice, as measured in the novel object recognition (NOR) and Barnes maze tests. Moreover, Aß resulted in inflammasome activation evident by enhanced caspase-1 immunoreactivity and interleukin-1 beta (IL-1ß) protein levels. However, ß-LAP could markedly reduce reactive oxygen species (ROS) production and down-regulate mRNA expression of NLRP3 inflammasome and protein levels of cleaved caspase 1 and IL-1ß. Additionally, ß-LAP-treated mice showed increased SIRT1 levels and NAD+/NADH ratio in the hippocampus. These results were followed by fewer number of TUNEL-positive cell, reduced hippocampal atrophy and neuronal loss in the hippocampal dentate gyrus (DG). These results indicated that the protective effect of ß-LAP against AD-associated cognitive deficits is partially through its strong antioxidant and anti-inflammatory actions.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Antiinflamatorios/uso terapéutico , Disfunción Cognitiva/tratamiento farmacológico , Inflamasomas/metabolismo , Naftoquinonas/uso terapéutico , Inflamación Neurogénica/tratamiento farmacológico , Péptidos beta-Amiloides/inmunología , Animales , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Estrés Oxidativo
12.
Int Immunopharmacol ; 81: 106263, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32028243

RESUMEN

Pathological ß-amyloid (Aß)-induced microglial activation could cause chronic neuroinflammation in the brain of Alzheimer's disease (AD) patients, and has been considered as one of the main pathological events of this disease. Chicago sky blue 6B (CSB6B), a pigment used in biochemical staining, has been reported to produce analgesic effects in neuroinflammatory-associated pain models. We have previously found that CSB6B could directly inhibit Aß aggregation and prevent Aß toxicity in neurons. However, it remains unclear whether this compound could prevent Aß-induced neuroinflammation and impairments of learning and memory in the AD models. In this study, CSB6B was found to effectively inhibit the production of pro-inflammatory cytokines, including tumor necrosis factor-α and interleukin-1ß, without affecting cell viability in BV2 microglia cells stimulated by Aß oligomer and lipopolysaccharide. Moreover, CSB6B significantly reduced mRNA expression of inducible nitric oxide synthase and increased mRNA expression of arginase-1, suggesting that CSB6B might promote the polarization of BV2 cells into M2 phenotype. In Aß oligomer-treated mice, hippocampal injection of CSB6B prevented cognitive impairments, and attenuated pro-inflammatory cytokines production. In addition, CSB6B inhibited nuclear transcription factor-κB (NF-κB), and restrainedthe activation of NOD-like receptor pyrin domain containing-3 (NLRP3) both in vitro and in vivo. According to our results, CSB6B may counteract Aß-induced cognitive impairments and neuroinflammation by inhibiting NF-κB and NLRP3. Combined with previous studies, we anticipated that CSB6B may further develop into a potential anti-AD drug with multiple functions on neurons and microglia cells, concurrently.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Antiinflamatorios/uso terapéutico , Disfunción Cognitiva/tratamiento farmacológico , Microglía/metabolismo , Inflamación Neurogénica/tratamiento farmacológico , Neuronas/metabolismo , Azul de Tripano/uso terapéutico , Péptidos beta-Amiloides/inmunología , Animales , Línea Celular , Citocinas/metabolismo , Humanos , Mediadores de Inflamación/metabolismo , Ratones , Ratones Endogámicos ICR , Microglía/patología , FN-kappa B/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Neuronas/patología , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo
13.
Int Immunopharmacol ; 81: 106281, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32058930

RESUMEN

Cerebral infarction causes severe social and economic burdens to patients due to its high morbidity and mortality rates, and the available treatments are limited. RO27-3225 is a highly selective melanocortin receptor 4 agonist that alleviates damage in many nervous system diseases, such as cerebral hemorrhage, traumatic brain injury and chronic neurodegenerative diseases. However, the effect of RO27-3225 on cerebral infarction remains unclear. In this study, we used a mouse model of transient middle cerebral artery occlusion (tMCAO) and administered RO27-3225 or saline to the mice through intraperitoneal injection. RO27-3225 increased the number of Nestin+/BrdU+ cells and doublecortin (DCX)+/BrdU+ cells in the subventricular zone (SVZ) and the number of DCX+/BrdU+ cells in the peri-infarct area on day 7 after tMCAO. Furthermore, RO27-3225 decreased the number of activated microglia (Iba1+ cells with a specific morphology) and the expression levels of Iba1, TNFα, IL6, and iNOS proteins and increased the number of PDGFRß+ cells in the peri-infarct region on day 3 after tMCAO. Finally, RO27-3225-treated mice exhibited significantly decreased infarct volumes, brain water contents, and neurological deficits after cerebral infarction. Thus, RO27-3225 can improve the outcomes following cerebral infarction, partially by regulating neurogenesis in the SVZ, PDGFRß+ cell survival and neuroinflammation in the peri-infarct zone. Our research reveals that RO27-3225 is a potential new treatment for cerebral infarction.


Asunto(s)
Antiinflamatorios/uso terapéutico , Infarto Cerebral/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Inflamación Neurogénica/tratamiento farmacológico , Neuronas/fisiología , Péptidos/uso terapéutico , Animales , Células Cultivadas , Proteína Doblecortina , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Neurogénesis , Neuronas/efectos de los fármacos , Receptor de Melanocortina Tipo 4/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo
15.
J Cereb Blood Flow Metab ; 40(12): 2505-2520, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-31910709

RESUMEN

Lipopolysaccharide (LPS) is a major component of the outer membrane of Gram-negative bacteria and a potent inflammatory stimulus for the innate immune response via toll-like receptor (TLR) 4 activation. Type 2 diabetes is associated with changes in gut microbiota and impaired intestinal barrier functions, leading to translocation of microbiota-derived LPS into the circulatory system, a condition referred to as metabolic endotoxemia. We investigated the effects of metabolic endotoxemia after experimental stroke with transient middle cerebral artery occlusion (MCAO) in a murine model of type 2 diabetes (db/db) and phenotypically normal littermates (db/+). Compared to db/+ mice, db/db mice exhibited an altered gut microbial composition, increased intestinal permeability, and higher plasma LPS levels. In addition, db/db mice presented increased infarct volumes and higher expression levels of LPS, TLR4, and inflammatory cytokines in the ischemic brain, as well as more severe neurological impairments and reduced survival rates after MCAO. Oral administration of a non-absorbable antibiotic modulated the gut microbiota and improved metabolic endotoxemia and stroke outcomes in db/db mice; these effects were associated with reduction of LPS levels and neuroinflammation in the ischemic brain. These data suggest that targeting metabolic endotoxemia may be a novel potential therapeutic strategy to improve stroke outcomes.


Asunto(s)
Isquemia Encefálica/metabolismo , Endotoxemia/metabolismo , Lipopolisacáridos/metabolismo , Inflamación Neurogénica/metabolismo , Administración Oral , Animales , Antibacterianos/administración & dosificación , Antibacterianos/uso terapéutico , Isquemia Encefálica/patología , Estudios de Casos y Controles , Citocinas/metabolismo , Diabetes Mellitus Tipo 2/complicaciones , Endotoxemia/tratamiento farmacológico , Endotoxemia/fisiopatología , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/fisiología , Bacterias Gramnegativas/metabolismo , Inmunidad Innata/efectos de los fármacos , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/patología , Infarto de la Arteria Cerebral Media/veterinaria , Lipopolisacáridos/sangre , Lipopolisacáridos/farmacología , Masculino , Ratones , Modelos Animales , Inflamación Neurogénica/tratamiento farmacológico , Inflamación Neurogénica/fisiopatología , Polimixina B/administración & dosificación , Polimixina B/uso terapéutico , Accidente Cerebrovascular/metabolismo , Tasa de Supervivencia , Receptor Toll-Like 4/efectos de los fármacos , Receptor Toll-Like 4/metabolismo
16.
Int Immunopharmacol ; 79: 106092, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31863920

RESUMEN

Baicalein (5,6,7-trihydroxyflavone), isolated from the root of traditional Chinese herb Scutellaria baicalensis Georgi, has anti-inflammatory and anti-oxidative activities. This study explored the protective and modulatory mechanisms of baicalein on neuroinflammation, oxidative stress and metabolic abnormality in lipopolysaccharide (LPS)-activated BV-2 cells. Our results demonstrated that treatment with baicalein remarkably restrained the production of pro-inflammatory factors including nitric oxide (NO), interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) in LPS-activated BV-2 cells. Moreover, baicalein significantly inhibited reactive oxygen species (ROS) production, decreased cyclooxygenase-2 (COX-2) and nuclear factor-b (NF-κB)/p65 expression. 1H NMR metabolomics analysis revealed that 12 differential metabolites were regulated by baicalein, implicated in alanine, aspartate and glutamate metabolism, glutathione metabolism, arginine and proline metabolism, D-glutamine and D-glutamate metabolism. In conclusion, these results indicated that baicalein has protective and modulatory effects on neuroinflammation and oxidative stress in LPS-activated BV-2 cells.


Asunto(s)
Antiinflamatorios/farmacología , Flavanonas/farmacología , Enfermedades Metabólicas/tratamiento farmacológico , Microglía/metabolismo , Inflamación Neurogénica/tratamiento farmacológico , Animales , Línea Celular , Ciclooxigenasa 2/metabolismo , Citocinas/metabolismo , Regulación de la Expresión Génica , Mediadores de Inflamación/metabolismo , Lipopolisacáridos/metabolismo , Ratones , Microglía/patología , FN-kappa B/metabolismo , Transducción de Señal
17.
Inflammation ; 43(1): 264-273, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31707574

RESUMEN

Two main contributors of sterile neurogenic inflammation underlying migraine pain, calcitonin gene-related peptide (CGRP), and meningeal mast cells (MMCs) play a key role in the activation of the inflammatory cascade resulting in the sensitization of trigeminal nociceptors. It is well established that phytochemical agent thymoquinone exhibits multiple anti-inflammatory effects in different in vitro and in vivo models of neuroinflammation. But its effects on the CGRP release and meningeal mast cells are unknown. In the present study, we investigated the effects of thymoquinone on the CGRP release in migraine-related strategic structures which are crucial targets for anti-migraine drugs, and on the MMCs in glyceryl trinitrate (GTN)-induced in vivo migraine model as well as in the ex vivo meningeal preparations in rats. Anti-inflammatory thymoquinone ameliorated GTN-stimulated CGRP levels in plasma, and migraine-related structures including trigeminal ganglion and brainstem; moreover, thymoquinone inhibited degranulation of MMCs and prevented the increase in the number of MMCs in GTN-induced in vivo migraine model. However, in the ex vivo meningeal preparations, thymoquinone did not inhibit the GTN-induced CGRP release from trigeminal meningeal afferents. Our findings suggest that thymoquinone mediates modulation of CGRP release in trigeminal ganglion neurons and brainstem, and stabilization of MMCs. Thus, thymoquinone may be a promising candidate to prevent the meningeal neurogenic inflammation and consequently migraine.


Asunto(s)
Antiinflamatorios/farmacología , Benzoquinonas/farmacología , Péptido Relacionado con Gen de Calcitonina/metabolismo , Mastocitos/metabolismo , Trastornos Migrañosos/tratamiento farmacológico , Inflamación Neurogénica/prevención & control , Animales , Péptido Relacionado con Gen de Calcitonina/sangre , Modelos Animales de Enfermedad , Masculino , Meninges/citología , Trastornos Migrañosos/inducido químicamente , Inflamación Neurogénica/tratamiento farmacológico , Nitroglicerina/toxicidad , Fitoquímicos/farmacología , Ratas , Ratas Wistar
18.
Front Immunol ; 10: 2707, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31849936

RESUMEN

Background: Accumulating evidence suggests inhibiting neuroinflammation as a potential target in therapeutic or preventive strategies for Alzheimer's disease (AD). MAPK-activated protein kinase II (MK2), downstream kinase of p38 mitogen activated protein kinase (MAPK) p38 MAPK, was unveiled as a promising option for the treatment of AD. Increasing evidence points at MK2 as involved in neuroinflammatory responses. MMI-0100, a cell-penetrating peptide inhibitor of MK2, exhibits anti-inflammatory effects and is in current clinical trials for the treatment of pulmonary fibrosis. Therefore, it is important to understand the actions of MMI-0100 in neuroinflammation. Methods: The mouse memory function was evaluated using novel object recognition (NOR) and object location recognition (OLR) tasks. Brain hippocampus tissue samples were analyzed by quantitative PCR, Western blotting, and immunostaining. Near-infrared fluorescent and confocal microscopy experiments were used to detect the brain uptake and distribution after intranasal MMI-0100 application. Results: Central MMI-0100 was able to ameliorate the memory deficit induced by Aß1-42 or LPS in novel object and location memory tasks. MMI-0100 suppressed LPS-induced activation of astrocytes and microglia, and dramatically decreased a series of pro-inflammatory cytokines such as TNF-α, IL-6, IL-1ß, COX-2, and iNOS via inhibiting phosphorylation of MK2, but not ERK, JNK, and p38 in vivo and in vitro. Importantly, one of the reasons for the failure of macromolecular protein or peptide drugs in the treatment of AD is that they cannot cross the blood-brain barrier. Our data showed that intranasal administration of MMI-0100 significantly ameliorates the memory deficit induced by Aß1-42 or LPS. Near-infrared fluorescent and confocal microscopy experiment results showed that a strong fluorescent signal, coming from mouse brains, was observed at 2 h after nasal applications of Cy7.5-MMI-0100. However, brains from control mice treated with saline or Cy7.5 alone displayed no significant signal. Conclusions: MMI-0100 attenuates Aß1-42- and LPS-induced neuroinflammation and memory impairments via the MK2 signaling pathway. Meanwhile, these data suggest that the MMI-0100/MK2 system may provide a new potential target for treatment of AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Astrocitos/fisiología , Hipocampo/metabolismo , MAP Quinasa Quinasa 2/metabolismo , Trastornos de la Memoria/tratamiento farmacológico , Inflamación Neurogénica/tratamiento farmacológico , Péptidos/uso terapéutico , Administración Intranasal , Péptidos beta-Amiloides/inmunología , Animales , Autoantígenos/inmunología , Células Cultivadas , Citocinas/metabolismo , Modelos Animales de Enfermedad , Hipocampo/patología , Humanos , Mediadores de Inflamación/metabolismo , MAP Quinasa Quinasa 2/antagonistas & inhibidores , Masculino , Ratones , Ratones Endogámicos , Fragmentos de Péptidos/inmunología , Péptidos/farmacología , Transducción de Señal
19.
Elife ; 82019 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-31765298

RESUMEN

Voltage-dependent sodium and calcium channels in pain-initiating nociceptor neurons are attractive targets for new analgesics. We made a permanently charged cationic derivative of an N-type calcium channel-inhibitor. Unlike cationic derivatives of local anesthetic sodium channel blockers like QX-314, this cationic compound inhibited N-type calcium channels more effectively with extracellular than intracellular application. Surprisingly, the compound is also a highly effective sodium channel inhibitor when applied extracellularly, producing more potent inhibition than lidocaine or bupivacaine. The charged inhibitor produced potent and long-lasting analgesia in mouse models of incisional wound and inflammatory pain, inhibited release of the neuropeptide calcitonin gene-related peptide (CGRP) from dorsal root ganglion neurons, and reduced inflammation in a mouse model of allergic asthma, which has a strong neurogenic component. The results show that some cationic molecules applied extracellularly can powerfully inhibit both sodium channels and calcium channels, thereby blocking both nociceptor excitability and pro-inflammatory peptide release.


Asunto(s)
Canales de Calcio Tipo N/genética , Inflamación Neurogénica/tratamiento farmacológico , Dolor/tratamiento farmacológico , Canales de Sodio/genética , Animales , Bupivacaína/farmacología , Calcio/metabolismo , Bloqueadores de los Canales de Calcio/farmacología , Señalización del Calcio/efectos de los fármacos , Modelos Animales de Enfermedad , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/patología , Humanos , Lidocaína/análogos & derivados , Lidocaína/farmacología , Ratones , Inflamación Neurogénica/genética , Inflamación Neurogénica/patología , Nociceptores , Dolor/genética , Dolor/patología , Bloqueadores de los Canales de Sodio/farmacología
20.
Biochemistry (Mosc) ; 84(2): 101-118, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31216970

RESUMEN

TRPA1 is a cation channel located on the plasma membrane of many types of human and animal cells, including skin sensory neurons and epithelial cells of the intestine, lungs, urinary bladder, etc. TRPA1 is the major chemosensor that also responds to thermal and mechanical stimuli. Substances that activate TRPA1, e.g., allyl isothiocyanates (pungent components of mustard, horseradish, and wasabi), cinnamaldehyde from cinnamon, organosulfur compounds from garlic and onion, tear gas, acrolein and crotonaldehyde from cigarette smoke, etc., cause burning, mechanical and thermal hypersensitivity, cough, eye irritation, sneezing, mucus secretion, and neurogenic inflammation. An increased activity of TRPA1 leads to the emergence of chronic pruritus and allergic dermatitis and is associated with episodic pain syndrome, a hereditary disease characterized by episodes of debilitating pain triggered by stress. TRPA1 is now considered as one of the targets for developing new anti-inflammatory and analgesic drugs. This review summarizes information on the structure, function, and physiological role of this channel, as well as describes known TRPA1 ligands and their significance as therapeutic agents in the treatment of inflammation-associated pain.


Asunto(s)
Analgésicos/farmacología , Antiinflamatorios no Esteroideos/farmacología , Inflamación Neurogénica/tratamiento farmacológico , Dolor/tratamiento farmacológico , Canal Catiónico TRPA1/antagonistas & inhibidores , Analgésicos/química , Animales , Antiinflamatorios no Esteroideos/química , Humanos , Ligandos , Estructura Molecular , Inflamación Neurogénica/metabolismo , Dolor/metabolismo , Canal Catiónico TRPA1/química , Canal Catiónico TRPA1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA