Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 176
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Adv Sci (Weinh) ; 11(19): e2308031, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38493498

RESUMEN

Activated microglia in the retina are essential for the development of autoimmune uveitis. Yin-Yang 1 (YY1) is an important transcription factor that participates in multiple inflammatory and immune-mediated diseases. Here, an increased YY1 lactylation in retinal microglia within in the experimental autoimmune uveitis (EAU) group is observed. YY1 lactylation contributed to boosting microglial activation and promoting their proliferation and migration abilities. Inhibition of lactylation suppressed microglial activation and attenuated inflammation in EAU. Mechanistically, cleavage under targets & tagmentation ï¼ˆCUT&Tag) analysis revealed that YY1 lactylation promoted microglial activation by regulating the transcription of a set of inflammatory genes, including STAT3, CCL5, IRF1, IDO1, and SEMA4D. In addition, p300 is identified as the writer of YY1 lactylation. Inhibition of p300 decreased YY1 lactylation and suppressed microglial inflammation in vivo and in vitro. Collectively, the results showed that YY1 lactylation promoted microglial dysfunction in autoimmune uveitis by upregulating inflammatory cytokine secretion and boosting cell migration and proliferation. Therapeutic effects can be achieved by targeting the lactate/p300/YY1 lactylation/inflammatory genes axis.


Asunto(s)
Enfermedades Autoinmunes , Modelos Animales de Enfermedad , Microglía , Uveítis , Factor de Transcripción YY1 , Animales , Femenino , Humanos , Ratones , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/metabolismo , Proliferación Celular/genética , Inflamación/genética , Inflamación/metabolismo , Ratones Endogámicos C57BL , Microglía/metabolismo , Microglía/inmunología , Uveítis/genética , Uveítis/inmunología , Uveítis/metabolismo , Factor de Transcripción YY1/genética , Factor de Transcripción YY1/metabolismo
2.
Front Immunol ; 12: 689453, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34616393

RESUMEN

Evidence concerning the role of alcohol-induced neuroinflammation in alcohol intake and relapse has increased in the last few years. It is also proven that mu-opioid receptors (MORs) mediate the reinforcing properties of alcohol and, interestingly, previous research suggests that neuroinflammation and MORs could be related. Our objective is to study neuroinflammatory states and microglial activation, together with adaptations on MOR expression in the mesocorticolimbic system (MCLS) during the abstinence and relapse phases. To do so, we have used a sex-dependent rat model of complete Freund's adjuvant (CFA)-induced alcohol deprivation effect (ADE). Firstly, our results confirm that only CFA-treated female rats, the only experimental group that showed relapse-like behavior, exhibited specific alterations in the expression of phosphorylated NFκB, iNOS, and COX2 in the PFC and VTA. More interestingly, the analysis of the IBA1 expression revealed a decrease of the microglial activation in PFC during abstinence and an increase of its expression in the relapse phase, together with an augmentation of this activation in the NAc in both phases that only occur in female CFA-treated rats. Additionally, the expression of IL1ß also evidenced these dynamic changes through these two phases following similar expression patterns in both areas. Furthermore, the expression of the cytokine IL10 showed a different profile than that of IL1ß, indicating anti-inflammatory processes occurring only during abstinence in the PFC of CFA-female rats but neither during the reintroduction phase in PFC nor in the NAc. These data indicate a downregulation of microglial activation and pro-inflammatory processes during abstinence in the PFC, whereas an upregulation can be observed in the NAc during abstinence that is maintained during the reintroduction phase only in CFA-female rats. Secondly, our data reveal a correlation between the alterations observed in IL1ß, IBA1 levels, and MOR levels in the PFC and NAc of CFA-treated female rats. Although premature, our data suggest that neuroinflammatory processes, together with neural adaptations involving MOR, might play an important role in alcohol relapse in female rats, so further investigations are warranted.


Asunto(s)
Alcoholismo/metabolismo , Sistema Límbico/metabolismo , Microglía/metabolismo , Neuroinmunomodulación , Dolor/metabolismo , Corteza Prefrontal/metabolismo , Receptores Opioides mu/metabolismo , Abstinencia de Alcohol , Alcoholismo/inmunología , Alcoholismo/fisiopatología , Animales , Proteínas de Unión al Calcio/metabolismo , Ciclooxigenasa 2/metabolismo , Citocinas/metabolismo , Femenino , Adyuvante de Freund , Mediadores de Inflamación/metabolismo , Sistema Límbico/inmunología , Sistema Límbico/fisiopatología , Masculino , Proteínas de Microfilamentos/metabolismo , Microglía/inmunología , FN-kappa B/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Dolor/inducido químicamente , Dolor/inmunología , Dolor/fisiopatología , Fosforilación , Corteza Prefrontal/inmunología , Corteza Prefrontal/fisiopatología , Ratas Sprague-Dawley , Recurrencia , Factores Sexuales
3.
Eur J Pharmacol ; 909: 174362, 2021 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-34297968

RESUMEN

The functions and mechanisms of GPR40 receptor to ameliorating the Alzheimer's disease (AD) by external treatment of encephalopathy remain unknown. In present study, the typical Aß1-42 induced mice model was applied to explore the functions and mechanisms of GPR40 receptor by external treatment of encephalopathy in AD. GPR40 agonist GW9508 and antagonist GW1100 were given by i.g injection to activate/inhibit the GPR40 receptor respectively in the gut of AD mouse which illustrated the function and mechanism of GPR40 receptor in ameliorating AD symptoms by external treatment of encephalopathy. A series of behavioral experiments were used to investigate the cognitive function and memory ability of mice, while molecular biology experiments such as Western blot, ELISA, flow cytometry were used to detect the corresponding changes of signaling pathways. The results revealed that intragastric administrated GW9508 could significantly ameliorate cognitive deficits of AD mouse, up-regulate the expression levels of gut-brain peptides both in blood circulation and hypothalamus thus up-regulate the expression levels of α-MSH in hypothalamus, while the negative autophagy-related proteins and inflammation-related proteins were down-regulated correspondingly. Meanwhile, GW9508 could also inhibit the pathological process of neuroinflammation in microglia. GW1100 reversed the effects of GW9508 significantly. These results suggested that GPR40 was an underlying therapeutic target for the external treatment of encephalopathy related to AD and GPR40 agonist could be explored as the emerging AD therapeutic drug.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Disfunción Cognitiva/tratamiento farmacológico , Metilaminas/administración & dosificación , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Propionatos/administración & dosificación , Receptores Acoplados a Proteínas G/agonistas , Administración Oral , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/administración & dosificación , Péptidos beta-Amiloides/toxicidad , Animales , Técnicas de Observación Conductual , Barrera Hematoencefálica/metabolismo , Disfunción Cognitiva/diagnóstico , Disfunción Cognitiva/inmunología , Disfunción Cognitiva/patología , Regulación hacia Abajo/efectos de los fármacos , Humanos , Hipotálamo/efectos de los fármacos , Hipotálamo/inmunología , Hipotálamo/patología , Masculino , Metilaminas/farmacocinética , Ratones , Microglía/efectos de los fármacos , Microglía/inmunología , Microglía/patología , Enfermedades Neuroinflamatorias/diagnóstico , Enfermedades Neuroinflamatorias/inmunología , Enfermedades Neuroinflamatorias/patología , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/toxicidad , Propionatos/farmacocinética , Receptores Acoplados a Proteínas G/metabolismo , Distribución Tisular
4.
J Tradit Chin Med ; 41(2): 185-193, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33825397

RESUMEN

OBJECTIVE: To investigate the anti-neuroinflammatory properties of Panax ginseng (P. ginseng) root by measuring the levels of nitric oxide (NO), tumour necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and interleukin-10 (IL-10) in lipopolysaccharide (LPS)-stimulated BV2 microglia cells. METHODS: Maximal non-toxic dose (MNTD) of methanol extract of P. ginseng root culture on BV2 microglia cells was first determined via 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, followed by treatment and LPS stimulation of cells, and the measurement of NO using Griess assay and TNF-α, IL-6, and IL-10 using ELISA assay. RESULTS: The MNTD of P. ginseng root extract was determined to be (587 ± 57) µg/mL. Following that, NO and IL-6 levels were found to be insignificantly reduced by 6.88% and 0.14% respectively in stimulated cells upon treatment with MNTD. Treatment with MNTD yielded similar insignificant result, with only a reduction of 3.58% and 0.08% in NO and IL-6 levels respectively. However, TNF-α and IL-10 levels were significantly downregulated by 15.64% and 34.96% respectively upon treatment with P. ginseng root extract at MNTD. CONCLUSION: Methanol extract of P. ginseng root culture did not show any significant anti-inflammatory effects on NO and IL-6 levels, but might potentially possess both anti-neuroinflammatory and pro-neuroinflammatory properties through the downregulation of TNF-α and IL-10 respectively.


Asunto(s)
Antiinflamatorios/farmacología , Medicamentos Herbarios Chinos/farmacología , Inflamación/tratamiento farmacológico , Microglía/efectos de los fármacos , Panax/química , Animales , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/inmunología , Humanos , Inflamación/genética , Inflamación/inmunología , Lipopolisacáridos/efectos adversos , Microglía/inmunología , FN-kappa B/genética , FN-kappa B/inmunología , Óxido Nítrico/inmunología , Raíces de Plantas/química
5.
Nature ; 593(7858): 255-260, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33911285

RESUMEN

Alzheimer's disease (AD) is the most prevalent cause of dementia1. Although there is no effective treatment for AD, passive immunotherapy with monoclonal antibodies against amyloid beta (Aß) is a promising therapeutic strategy2,3. Meningeal lymphatic drainage has an important role in the accumulation of Aß in the brain4, but it is not known whether modulation of meningeal lymphatic function can influence the outcome of immunotherapy in AD. Here we show that ablation of meningeal lymphatic vessels in 5xFAD mice (a mouse model of amyloid deposition that expresses five mutations found in familial AD) worsened the outcome of mice treated with anti-Aß passive immunotherapy by exacerbating the deposition of Aß, microgliosis, neurovascular dysfunction, and behavioural deficits. By contrast, therapeutic delivery of vascular endothelial growth factor C improved clearance of Aß by monoclonal antibodies. Notably, there was a substantial overlap between the gene signature of microglia from 5xFAD mice with impaired meningeal lymphatic function and the transcriptional profile of activated microglia from the brains of individuals with AD. Overall, our data demonstrate that impaired meningeal lymphatic drainage exacerbates the microglial inflammatory response in AD and that enhancement of meningeal lymphatic function combined with immunotherapies could lead to better clinical outcomes.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Péptidos beta-Amiloides/inmunología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Inmunoterapia , Vasos Linfáticos/inmunología , Meninges/inmunología , Microglía/inmunología , Envejecimiento/efectos de los fármacos , Envejecimiento/inmunología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/efectos de los fármacos , Animales , Anticuerpos Monoclonales Humanizados/inmunología , Encéfalo/irrigación sanguínea , Encéfalo/citología , Encéfalo/efectos de los fármacos , Encéfalo/inmunología , Modelos Animales de Enfermedad , Hipocampo/citología , Hipocampo/efectos de los fármacos , Hipocampo/inmunología , Humanos , Inflamación/tratamiento farmacológico , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Masculino , Meninges/irrigación sanguínea , Meninges/citología , Ratones , Microglía/citología , Microglía/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Factor C de Crecimiento Endotelial Vascular/metabolismo , Factor C de Crecimiento Endotelial Vascular/farmacología
6.
Int J Mol Sci ; 22(5)2021 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-33806610

RESUMEN

Microglia, the innate immune cells of the CNS, exhibit long-term response changes indicative of innate immune memory (IIM). Our previous studies revealed IIM patterns of microglia with opposing immune phenotypes: trained immunity after a low dose and immune tolerance after a high dose challenge with pathogen-associated molecular patterns (PAMP). Compelling evidence shows that innate immune cells adopt features of IIM via immunometabolic control. However, immunometabolic reprogramming involved in the regulation of IIM in microglia has not been fully addressed. Here, we evaluated the impact of dose-dependent microglial priming with ultra-low (ULP, 1 fg/mL) and high (HP, 100 ng/mL) lipopolysaccharide (LPS) doses on immunometabolic rewiring. Furthermore, we addressed the role of PI3Kγ on immunometabolic control using naïve primary microglia derived from newborn wild-type mice, PI3Kγ-deficient mice and mice carrying a targeted mutation causing loss of lipid kinase activity. We found that ULP-induced IIM triggered an enhancement of oxygen consumption and ATP production. In contrast, HP was followed by suppressed oxygen consumption and glycolytic activity indicative of immune tolerance. PI3Kγ inhibited glycolysis due to modulation of cAMP-dependent pathways. However, no impact of specific PI3Kγ signaling on immunometabolic rewiring due to dose-dependent LPS priming was detected. In conclusion, immunometabolic reprogramming of microglia is involved in IIM in a dose-dependent manner via the glycolytic pathway, oxygen consumption and ATP production: ULP (ultra-low-dose priming) increases it, while HP reduces it.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase Ib/inmunología , Inmunidad Innata/inmunología , Memoria Inmunológica/inmunología , Adenosina Trifosfato/inmunología , Animales , Glucólisis/inmunología , Tolerancia Inmunológica/inmunología , Lipopolisacáridos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/inmunología , Consumo de Oxígeno/inmunología , Moléculas de Patrón Molecular Asociado a Patógenos/inmunología , Transducción de Señal/inmunología
7.
Int J Mol Sci ; 22(3)2021 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-33573368

RESUMEN

Neurodegeneration is the pathological condition, in which the nervous system or neuron loses its structure, function, or both, leading to progressive degeneration or the death of neurons, and well-defined associations of tissue system, resulting in clinical manifestations. Neuroinflammation has been shown to precede neurodegeneration in several neurodegenerative diseases (NDs). No drug is yet known to delay or treat neurodegeneration. Although the etiology and potential causes of NDs remain widely indefinable, matrix metalloproteinases (MMPs) evidently have a crucial role in the progression of NDs. MMPs, a protein family of zinc (Zn2+)-containing endopeptidases, are pivotal agents that are involved in various biological and pathological processes in the central nervous system (CNS). The current review delineates the several emerging evidence demonstrating the effects of MMPs in the progression of NDs, wherein they regulate several processes, such as (neuro)inflammation, microglial activation, amyloid peptide degradation, blood brain barrier (BBB) disruption, dopaminergic apoptosis, and α-synuclein modulation, leading to neurotoxicity and neuron death. Published papers to date were searched via PubMed, MEDLINE, etc., while using selective keywords highlighted in our manuscript. We also aim to shed a light on pathophysiological effect of MMPs in the CNS and focus our attention on its detrimental and beneficial effects in NDs, with a special focus on Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), Alzheimer's disease (AD), multiple sclerosis (MS), and Huntington's disease (HD), and discussed various therapeutic strategies targeting MMPs, which could serve as potential modulators in NDs. Over time, several agents have been developed in order to overcome challenges and open up the possibilities for making selective modulators of MMPs to decipher the multifaceted functions of MMPs in NDs. There is still a greater need to explore them in clinics.


Asunto(s)
Barrera Hematoencefálica/patología , Inhibidores de la Metaloproteinasa de la Matriz/uso terapéutico , Metaloproteinasas de la Matriz/metabolismo , Enfermedades Neurodegenerativas/patología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/inmunología , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/enzimología , Barrera Hematoencefálica/inmunología , Ensayos Clínicos como Asunto , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Humanos , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Inflamación/patología , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Microglía/inmunología , Microglía/metabolismo , Microglía/patología , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/inmunología , Neuronas/inmunología , Neuronas/patología , Resultado del Tratamiento
8.
Neuroimmunomodulation ; 28(1): 22-28, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33530091

RESUMEN

COVID-19, a disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) betacoronavirus, affects children in a different way than it does in adults, with milder symptoms. However, several cases of neurological symptoms with neuroinflammatory syndromes, such as the multisystem inflammatory syndrome (MIS-C), following mild cases, have been reported. As with other viral infections, such as rubella, influenza, and cytomegalovirus, SARS-CoV-2 induces a surge of proinflammatory cytokines that affect microglial function, which can be harmful to brain development. Along with the viral induction of neuroinflammation, other noninfectious conditions may interact to produce additional inflammation, such as the nutritional imbalance of fatty acids and polyunsaturated fatty acids and alcohol consumption during pregnancy. Additionally, transient thyrotoxicosis induced by SARS-CoV-2 with secondary autoimmune hypothyroidism has been reported, which could go undetected during pregnancy. Together, those factors may pose additional risk factors for SARS-CoV-2 infection impacting mechanisms of neural development such as synaptic pruning and neural circuitry formation. The present review discusses those conditions in the perspective of the understanding of risk factors that should be considered and the possible emergence of neurodevelopmental disorders in COVID-19-infected children.


Asunto(s)
Encéfalo/crecimiento & desarrollo , COVID-19/inmunología , Inflamación/inmunología , Microglía/inmunología , Trastornos del Neurodesarrollo/inmunología , Encéfalo/inmunología , Encéfalo/fisiopatología , COVID-19/fisiopatología , Dieta , Grasas Insaturadas en la Dieta , Ácidos Grasos Insaturados , Trastornos del Espectro Alcohólico Fetal/inmunología , Trastornos del Espectro Alcohólico Fetal/fisiopatología , Humanos , Inflamación/fisiopatología , Trastornos del Neurodesarrollo/fisiopatología , Plasticidad Neuronal , Factores de Riesgo , SARS-CoV-2 , Índice de Severidad de la Enfermedad , Síndrome de Respuesta Inflamatoria Sistémica
9.
Int J Mol Sci ; 22(3)2021 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-33494411

RESUMEN

Parkinson's disease (PD) is a common neurodegenerative disorder characterized by the loss of dopaminergic (DAergic) neurons and the presence of α-synuclein-containing Lewy bodies. The unstructured α-synuclein forms insoluble fibrils and aggregates that result in increased reactive oxygen species (ROS) and cellular toxicity in PD. Neuroinflammation engaged by microglia actively contributes to the pathogenesis of PD. In this study, we showed that VB-037 (a quinoline compound), glycyrrhetic acid (a pentacyclic triterpenoid), Glycyrrhiza inflata (G. inflata, a Chinese herbal medicine), and Shaoyao Gancao Tang (SG-Tang, a formulated Chinese medicine) suppressed the nitric oxide (NO) production and interleukin (IL)-1ß maturation in α-synuclein-stimulated BV-2 cells. Mouse inflammation antibody array further revealed increased IL-1α, IL-1ß, tumor necrosis factor (TNF)-α, interferon (IFN)-γ, IL-6, granulocyte-macrophage colony-stimulating factor (GM-CSF) and granulocyte colony-stimulating factor (G-CSF) expression in α-synuclein-inflamed BV-2 cells and compound pretreatment effectively reduced the expression and release of these pro-inflammatory mediators. The test compounds and herbal medicines further reduced α-synuclein aggregation and associated oxidative stress, and protected cells against α-synuclein-induced neurotoxicity by downregulating NLR family pyrin domain containing 1 (NLRP1) and 3 (NLRP3), caspase 1, IL-1ß, IL-6, and associated nuclear factor (NF)-κB inhibitor alpha (IκBα)/NF-κB P65 subunit (P65), c-Jun N-terminal kinase (JNK)/proto-oncogene c-Jun (JUN), mitogen-activated protein kinase 14 (P38)/signal transducer and activator of transcription 1 (STAT1) and Janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) pathways in dopaminergic neurons derived from α-synuclein-expressing SH-SY5Y cells. Our findings indicate the potential of VB-037, glycyrrhetic acid, G. inflata, and SG-Tang through mitigating α-synuclein-stimulated neuroinflammation in PD, as new drug candidates for PD treatment.


Asunto(s)
Susceptibilidad a Enfermedades , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/metabolismo , Animales , Antiinflamatorios/química , Antiinflamatorios/farmacología , Proteínas Bacterianas/metabolismo , Biomarcadores , Línea Celular Tumoral , Citocinas/genética , Citocinas/metabolismo , Genes Reporteros , Inflamasomas/metabolismo , Mediadores de Inflamación/metabolismo , Ratones , Microglía/efectos de los fármacos , Microglía/inmunología , Microglía/metabolismo , Estructura Molecular , Terapia Molecular Dirigida/métodos , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/patología , Factor de Transcripción STAT3/metabolismo , alfa-Sinucleína/metabolismo
10.
J Cell Mol Med ; 25(3): 1371-1382, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33369103

RESUMEN

Inflammation and neuronal apoptosis aggravate the secondary damage after spinal cord injury (SCI). Rehmannioside A (Rea) is a bioactive herbal extract isolated from Rehmanniae radix with low toxicity and neuroprotection effects. Rea treatment inhibited the release of pro-inflammatory mediators from microglial cells, and promoted M2 polarization in vitro, which in turn protected the co-cultured neurons from apoptosis via suppression of the NF-κB and MAPK signalling pathways. Furthermore, daily intraperitoneal injections of 80 mg/kg Rea into a rat model of SCI significantly improved the behavioural and histological indices, promoted M2 microglial polarization, alleviated neuronal apoptosis, and increased motor function recovery. Therefore, Rea is a promising therapeutic option for SCI and should be clinically explored.


Asunto(s)
Apoptosis/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Microglía/efectos de los fármacos , Microglía/metabolismo , Neuronas/metabolismo , Fármacos Neuroprotectores/farmacología , Traumatismos de la Médula Espinal/metabolismo , Animales , Biomarcadores , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Mediadores de Inflamación/metabolismo , Microglía/inmunología , Modelos Biológicos , Actividad Motora , FN-kappa B/metabolismo , Extractos Vegetales/química , Extractos Vegetales/farmacología , Ratas , Rehmannia/química , Transducción de Señal , Traumatismos de la Médula Espinal/tratamiento farmacológico , Traumatismos de la Médula Espinal/etiología , Traumatismos de la Médula Espinal/rehabilitación
11.
Biosci Trends ; 14(4): 271-278, 2020 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-32741856

RESUMEN

Tourette's syndrome (TS) is an inherited neurologic disorder characterized by involuntary stereotyped motor and vocal tics. Its pathogenesis is still unclear and its treatment remains limited. Recent research has suggested the involvement of immune mechanisms in the pathophysiology of TS. Microglia are the brain's resident innate immune cells. They can mediate neuroinflammation and regulate brain development and homeostasis. A traditional Chinese medicine (TCM), Ningdong granule (NDG), has been found to be efficacious in the treatment of TS while causing few adverse reactions. In the current study, a rat model of 3,3'-iminodipropionitrile (IDPN)-induced TS was used to explore the regulating effects and mechanisms of NDG on microglia-mediated neuroinflammation. IDNP led to robust pathological changes and neurobehavioral complications, with activation of microglia in the striatum of rats with TS. After activation by IDNP, microglia strongly responded to this specific injury, and TNF-α, IL-6, and MCP-1 were released in the striatum and/or serum of rats with TS. Interestingly, NDG inhibited the activation of microglia and decreased the abnormal expression of TNF-α, IL-6, and MCP-1 in the striatum and/or serum of rats with TS, thus controlling tics. However, there were no significant changes in the striatum and/or serum of rats with TS after treatment with haloperidol. The anti-TS action of haloperidol might occur not through microglial activation and neuroinflammation but through the DAT system, thus controlling tics. In conclusion, microglia might play key roles in mediating neuroinflammatory responses in TS, triggering the release of TNF-α, IL-6, and MCP-1.NDG inhibited tics in rats with TS, and this mechanism may be associated with a reduction in the increased number of activated microglia and a decrease in the expression of pro-inflammatory cytokines and chemokines in the striatum and/or serum.


Asunto(s)
Cuerpo Estriado/efectos de los fármacos , Medicamentos Herbarios Chinos/farmacología , Microglía/efectos de los fármacos , Síndrome de Tourette/tratamiento farmacológico , Animales , Quimiocina CCL2/sangre , Quimiocina CCL2/metabolismo , Cuerpo Estriado/citología , Cuerpo Estriado/inmunología , Cuerpo Estriado/patología , Modelos Animales de Enfermedad , Medicamentos Herbarios Chinos/uso terapéutico , Haloperidol/farmacología , Haloperidol/uso terapéutico , Humanos , Interleucina-6/sangre , Interleucina-6/metabolismo , Masculino , Microglía/inmunología , Microglía/metabolismo , Nitrilos/toxicidad , Ratas , Ratas Wistar , Síndrome de Tourette/inducido químicamente , Síndrome de Tourette/inmunología , Síndrome de Tourette/patología , Factor de Necrosis Tumoral alfa/sangre , Factor de Necrosis Tumoral alfa/metabolismo
12.
Ann Clin Transl Neurol ; 7(9): 1513-1523, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32762033

RESUMEN

OBJECTIVE: Neuroinflammation is considered a key driver for neurodegeneration in several neurological diseases, including amyotrophic lateral sclerosis (ALS). SOD1 mutations cause about 20% of familial ALS, and related pathology might generate microglial activation triggering neurodegeneration. 11 C-PK11195 is the prototypical and most validated PET radiotracer, targeting the 18-kDa translocator protein which is overexpressed in activated microglia. In this study, we investigated microglia activation in asymptomatic (ASYM) and symptomatic (SYM) SOD1 mutated carriers, by using 11 C-PK11195 and PET imaging. METHODS: We included 20 subjects: 4 ASYM-carriers, neurologically normal, 6 SYM-carriers with probable ALS, and 10 healthy controls. A receptor parametric mapping procedure estimated 11 C-PK11195 binding potentials and voxel-wise statistical comparisons were performed at group and single-subject levels. RESULTS: Both the SYM- and ASYM-carriers showed significant microglia activation in cortical and subcortical structures, with variable patterns at individual level. Clusters of activation were present in occipital and temporal regions, cerebellum, thalamus, and medulla oblongata. Notably, SYM-carriers showed microglia activation also in supplementary and primary motor cortices and in the somatosensory regions. INTERPRETATION: In vivo neuroinflammation occurred in all SOD1 mutated cases since the presymptomatic stages, as shown by a significant cortical and subcortical microglia activation. The involvement of sensorimotor cortex became evident at the symptomatic disease stage. Although our data indicate the role of in vivo PET imaging for assessing resident microglia in the investigation of SOD1-ALS pathophysiology, further studies are needed to clarify the temporal and spatial dynamics of microglia activation and its relationship with neurodegeneration.


Asunto(s)
Amidas , Esclerosis Amiotrófica Lateral , Encéfalo , Inflamación , Isoquinolinas , Microglía , Tomografía de Emisión de Positrones , Superóxido Dismutasa-1/genética , Adulto , Anciano , Esclerosis Amiotrófica Lateral/diagnóstico por imagen , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/inmunología , Esclerosis Amiotrófica Lateral/metabolismo , Encéfalo/diagnóstico por imagen , Encéfalo/inmunología , Encéfalo/metabolismo , Femenino , Heterocigoto , Humanos , Inflamación/diagnóstico por imagen , Inflamación/inmunología , Inflamación/metabolismo , Masculino , Microglía/inmunología , Microglía/metabolismo , Persona de Mediana Edad , Síntomas Prodrómicos
13.
Molecules ; 25(16)2020 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-32784835

RESUMEN

An effective and previously demonstrated screening method for active constituents in natural products using LC-MS coupled with a bioassay was reported in our earlier studies. With this, the current investigation attempted to identify bioactive constituents of Scutellaria baicalensis through LC-MS coupled with a bioassay. Peaks at broadly 17-20 and 24-25 min on the MS chromatogram displayed an inhibitory effect on NO production in lipopolysaccharide-induced BV2 microglia cells. Similarly, peaks at roughly 17-19 and 22 min showed antioxidant activity with an 2,2'-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid) (ABTS)/2,2-diphenyl-1- picrylhydrazyl (DPPH) assay. For confirmation of LC-MS coupled with a bioassay, nine compounds (1-9) were isolated from an MeOH extract of S. baicalensis. As we predicted, compounds 1, 8, and 9 significantly reduced lipopolysaccharide (LPS)-induced NO production in BV2 cells. Likewise, compounds 5, 6, and 8 exhibited free radical-scavenging activities with the ABTS/DPPH assay. In addition, the structural similarity of the main components was confirmed by analyzing the total extract and EtOAc fractions through molecular networking. Overall, the results suggest that the method comprised of LC-MS coupled with a bioassay can effectively predict active compounds without an isolation process, and the results of molecular networking predicted that other components around the active compound node may also be active.


Asunto(s)
Antiinflamatorios/farmacología , Antioxidantes/farmacología , Cromatografía Liquida/métodos , Microglía/efectos de los fármacos , Extractos Vegetales/farmacología , Scutellaria baicalensis/química , Espectrometría de Masas en Tándem/métodos , Animales , Bioensayo , Lipopolisacáridos/farmacología , Ratones , Microglía/citología , Microglía/inmunología
14.
Mol Cells ; 43(5): 431-437, 2020 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-32392909

RESUMEN

The hypothalamus is a crucial organ for the maintenance of appropriate body fat storage. Neurons in the hypothalamic arcuate nucleus (ARH) detect energy shortage or surplus via the circulating concentrations of metabolic hormones and nutrients, and then coordinate energy intake and expenditure to maintain energy homeostasis. Malfunction or loss of hypothalamic ARH neurons results in obesity. Accumulated evidence suggests that hypothalamic inflammation is a key pathological mechanism that links chronic overconsumption of a high-fat diet (HFD) with the development of obesity and related metabolic complications. Interestingly, overnutrition-induced hypothalamic inflammation occurs specifically in the ARH, where microglia initiate an inflammatory response by releasing proinflammatory cytokines and chemokines in response to excessive fatty acid flux. Upon more prolonged HFD consumption, astrocytes and perivascular macrophages become involved and sustain hypothalamic inflammation. ARH neurons are victims of hypothalamic inflammation, but they may actively participate in hypothalamic inflammation by sending quiescence or stress signals to surrounding glia. In this mini-review, we describe the current state of knowledge regarding the contributions of neurons and glia, and their interactions, to HFD-induced hypothalamic inflammation.


Asunto(s)
Tejido Adiposo/inmunología , Hipotálamo/inmunología , Inflamación/metabolismo , Macrófagos/inmunología , Microglía/inmunología , Neuronas/inmunología , Obesidad/inmunología , Animales , Citocinas/metabolismo , Dieta Alta en Grasa , Metabolismo Energético , Humanos , Inmunidad Celular , Inflamación Neurogénica
15.
Acupunct Med ; 38(6): 426-434, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32310010

RESUMEN

OBJECTIVE: To investigate the regulatory mechanism of manual acupuncture (MA) on microglial polarization-mediated neuroinflammation after traumatic brain injury (TBI), focusing on the RhoA/Rho-associated coiled coil-forming protein kinase (ROCK2) pathway. METHODS: Sprague Dawley (SD) rats were used to generate a TBI model using Feeney's freefall epidural impact method. MA was performed on half of the TBI model rats, while the others remained untreated. Acupuncture was administered at GV15, GV16, GV20, GV26, and LI4. At the end of the intervention, rat brain tissue samples were collected, and the microglial M1 polarization status was observed by immunofluorescence labeling of CD86, an M1 microglia-specific protein. RhoA/ROCK2 signaling components were detected by quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting. An enzyme-linked immunosorbent assay (ELISA) was used to detect the expression levels of inflammatory factors. RESULTS: Compared with normal rats, the CD86 expression density in the untreated TBI model rats was high and showed an aggregated expression pattern. The genes and proteins of the RhoA/ROCK2 signaling pathway were highly expressed, and inflammatory factors were significantly increased. The CD86 expression density in TBI rats after MA was reduced compared to that in untreated TBI rats and showed a scattered distribution. The expression of RhoA/ROCK2 signaling pathway genes and proteins was also significantly reduced, and inflammatory factors were decreased. CONCLUSION: These results show that MA may inhibit M1 polarization of microglia by regulating the RhoA/ROCK2 signaling pathway, thereby reducing neuroinflammation in TBI.


Asunto(s)
Terapia por Acupuntura , Lesiones Traumáticas del Encéfalo/inmunología , Lesiones Traumáticas del Encéfalo/terapia , Microglía/inmunología , Proteínas de Unión al GTP rho/inmunología , Quinasas Asociadas a rho/inmunología , Animales , Lesiones Traumáticas del Encéfalo/enzimología , Lesiones Traumáticas del Encéfalo/genética , Modelos Animales de Enfermedad , Humanos , Masculino , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Proteínas de Unión al GTP rho/genética , Quinasas Asociadas a rho/genética
16.
J Agric Food Chem ; 68(12): 3750-3757, 2020 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-32125844

RESUMEN

Increasing health-promoting effects of resveratrol and its molecular structural analogues have been discovered, and the acting mechanism has been explored. However, the activity comparison of such compounds in targeting macrophage-related inflammation associated with neurodegenerative diseases remains untouched. In this study, we evaluated the activation and polarization transition of lipopolysaccharide (LPS)-stimulated BV-2 mouse microglial macrophages exposed to resveratrol (RES) and its analogues pterostilbene (PTE), oxyresveratrol (ORES), acetyl-trans-resveratrol (ARES), and trans-2,3,5,4'-tetrahydroxystilbene-2-O-glucopyranoside (TSG). At 10 µM, all of the five stilbene compounds have effectively suppressed the LPS-stimulated BV-2 cell release of proinflammatory mediators such as NO, TNF-α, iNOS, IL-1ß, and IL-6. Mechanism study elucidated that they exert anti-inflammatory effects through MAPKs (ERK1/2, JNK, and p38) and NF-κB signaling pathways. Further investigation in treating BV-2 cells with resveratrol and its analogues revealed the reversal of LPS-induced phenotype molecules from M1 (iNOS, IL-1ß, IL-6, and CD86) to M2 (Arg1, CD163, and IL-10) subtypes, manifesting that these five stilbenes suppressed inflammation through modulating the polarized phenotypes of BV-2 microglia. Most importantly, PTE demonstrated the most potent inhibitory activity among these five stilbene compounds. Therefore, this study not only highlights microglia-induced inflammatory responses as a potential therapeutic target but also suggests future insights in considering the options of nutraceutical development for resveratrol and its analogues.


Asunto(s)
Antiinflamatorios/química , Antiinflamatorios/farmacología , Macrófagos/efectos de los fármacos , Microglía/efectos de los fármacos , Resveratrol/análogos & derivados , Resveratrol/farmacología , Animales , Línea Celular , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Lipopolisacáridos/inmunología , Activación de Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones , Microglía/inmunología , Extractos Vegetales/farmacología , Estilbenos/farmacología
17.
Int Immunopharmacol ; 83: 106396, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32193103

RESUMEN

OBJECTIVE: To elucidate the effects of ISO-α-acids (IAAs), a PPAR-γ agonist, on ICH rats and its potential mechanism. MATERIAL AND METHODS: The Sprague Dawley rats ICH model was induced by stereotactic injecting of 100 µl autologous artery blood. Ninety male rats were randomly allocated to five groups: autologous blood and IAAs (IAA); received autologous blood, IAAs and PPAR-γ inhibitor (IAA + GW9662); autologous blood and normal Saline (Saline); only autologous blood (Mock); and only needle injection (Sham). Neurological functions were assessed by mNSS. Hematoma volume, brain water content, surface proteins and inflammatory factors were detected. The microglia anti-inflammatory abilities were also evaluated. RESULTS: IAAs were able to significantly decrease ICH rat's mNSS scores, alleviate brain water content, improve hematoma resolution than Saline, Mock (p < 0.05). More "M2" microglial/macrophage can be induced by IAAs. The expression of CD 36 was statistically higher in IAA than other groups (p < 0.05). Injection of IAAs led to a greatly increasing in CD 11b and CD 206 double-positive anti-inflammatory type microglial/macrophage, moreover, a reduction of inflammatory cytokines expression (p < 0.05). Such protective effects can be relieved by GW9662. CONCLUSIONS: This is the first study to elucidate the relationship between IAAs and ICH. IAAs were able to accelerate hematoma absorption, alleviate brain edema, suppress peri-hematoma inflammations and finally improved the outcome of ICH rats. The phenotype was due to the IAAs induction of "M2" microglial/macrophage via activating of PPAR-γ and increasing CD 36 expression.


Asunto(s)
Edema Encefálico/tratamiento farmacológico , Hemorragia Cerebral/tratamiento farmacológico , Hematoma/tratamiento farmacológico , Ácidos Indolacéticos/uso terapéutico , Microglía/inmunología , Extractos Vegetales/uso terapéutico , Animales , Antiinflamatorios/uso terapéutico , Antígenos CD36/genética , Antígenos CD36/metabolismo , Diferenciación Celular , Citocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Humulus/inmunología , Ácidos Indolacéticos/farmacología , PPAR gamma/agonistas , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Células Th2/inmunología , Regulación hacia Arriba
18.
PLoS One ; 15(3): e0230142, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32210464

RESUMEN

Neuroinflammation is a major risk factor associated with the pathogenesis of neurodegenerative diseases. Conventional non-steroidal anti-inflammatory drugs are prescribed but their long term use is associated with adverse effects. Thus, herbal based medicines are attracting major attraction worldwide as potential therapeutic candidates. Tylophora indica (Burm. f) Merrill is a valuable medicinal plant well known in Ayurvedic practices for its immunomodulatory, anti-oxidant, anti-asthmatic and antirheumatic activities. The present study aimed to elucidate the anti-neuroinflammatory potential of water and hydroalcoholic leaf extracts of micropropagated plants of T. indica using BV-2 microglia activated with lipopolysaccharide as an in vitro model system and development of an efficient reproducible protocol for its in vitro cloning. Non cytotoxic doses of the water and hydroalcoholic extracts (0.2µg/ml and 20µg/ml, respectively) were selected using MTT assay. α-Tubulin, Iba-1 and inflammatory cascade proteins like NFκB, AP1 expression was studied using immunostaining to ascertain the anti-neuroinflammatory potential of these extracts. Further, anti-migratory activity was also analyzed by Wound Scratch Assay. Both extracts effectively attenuated lipopolysaccharide induced microglial activation, migration and the production of nitrite via regulation of the expression of NFκB and AP1 as the possible underlying target molecules. An efficient and reproducible protocol for in vitro cloning of T. indica through multiple shoot proliferation from nodal segments was established on both solid and liquid Murashige and Skoog's (MS) media supplemented with 15µM and 10µM of Benzyl Amino Purine respectively. Regenerated shoots were rooted on both solid and liquid MS media supplemented with Indole-3-butyric acid (5-15µM) and the rooted plantlets were successfully acclimatized and transferred to open field conditions showing 90% survivability. The present study suggests that T. indica may prove to be a potential anti-neuroinflammatory agent and may be further explored as a potential therapeutic candidate for the management of neurodegenerative diseases. Further, the current study will expedite the conservation of T. indica ensuring ample supply of this threatened medicinal plant to fulfill its increasing demand in herbal industry.


Asunto(s)
Microglía/efectos de los fármacos , Extractos Vegetales/uso terapéutico , Plantas Medicinales/crecimiento & desarrollo , Tylophora/crecimiento & desarrollo , Complejo 1 de Proteína Adaptadora/efectos de los fármacos , Complejo 1 de Proteína Adaptadora/metabolismo , Línea Celular , Humanos , Técnicas In Vitro , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Lipopolisacáridos/inmunología , Microglía/inmunología , FN-kappa B/efectos de los fármacos , FN-kappa B/metabolismo , Enfermedades Neurodegenerativas/tratamiento farmacológico
19.
Biomed Pharmacother ; 125: 110017, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32106384

RESUMEN

Despite the growing knowledge of the mechanisms of chronic pain, the treatment of this disorder in the clinic remains a major challenge. Src-family protein tyrosine kinases (SFKs), a group of non-receptor protein tyrosine kinases, have been implicated in neuronal development and synaptic plasticity. SFKs are critical for the regulate of N-methyl-D-aspartic acid receptor (NMDAR) 2B subunit phosphorylation by various transmembrane receptors, e.g., G-protein coupled receptors (GPCRs), EphB receptors (EphBRs), increased intracellular calcium, epidermal growth factor (EGF) and other growth factors, and thus contribute to the development of chronic pain. SFKs have also been regarded as important points of convergence of intracellular signalling components for the regulation of microglial functions and the immune response. Additionally, the intrathecal administration of SFK inhibitors significantly alleviates mechanical allodynia in different chronic pain models. Here, we reviewed the current evidence for the role of SFKs in the development of chronic pain caused by complete Freund's adjuvant (CFA) injection, peripheral nerve injury (PNI), streptozotocin (STZ) injection and bone metastasis. Moreover, the role of SFKs in the development of morphine tolerance is also discussed. The regulation of SFKs therefore has emerged as a potential therapeutic target for the treatment of chronic pain in terms of safety and efficacy.


Asunto(s)
Dolor Crónico/metabolismo , Familia-src Quinasas/metabolismo , Animales , Biomarcadores , Proteínas Portadoras/metabolismo , Dolor Crónico/tratamiento farmacológico , Dolor Crónico/etiología , Susceptibilidad a Enfermedades , Tolerancia a Medicamentos , Humanos , Inmunomodulación , Microglía/inmunología , Microglía/metabolismo , Terapia Molecular Dirigida , Morfina/metabolismo , Morfina/farmacología , Morfina/uso terapéutico , Plasticidad Neuronal/genética , Unión Proteica , Receptores de N-Metil-D-Aspartato/metabolismo , Transducción de Señal , Familia-src Quinasas/antagonistas & inhibidores
20.
Int Immunopharmacol ; 79: 106083, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31923823

RESUMEN

The neuroinflammatory response induced by microglia plays a vital role in causing secondary brain damage after traumatic brain injury (TBI). Previous studies have found that the improved regulation of activated microglia could reduce neurological damage post-TBI. Phillyrin (Phi) is one of the main active ingredients extracted from the fruits of the medicinal plant Forsythia suspensa (Thunb.) with anti-inflammatory effects. Our study attempted to investigate the effects of phillyrin on microglial activation and neuron damage after TBI. The TBI model was applied to induce brain injury in mice, and neurological scores, brain water content, hematoxylin and eosin staining and Nissl staining were employed to determine the neuroprotective effects of phillyrin. Immunofluorescent staining and western blot analysis were used to detect nuclear factor-kappa B (NF-κB) and peroxisome proliferator-activated receptor gamma (PPARγ) expression and nuclear translocation, and the inflammation-related proteins and mRNAs were assessed by western blot analysis and quantitative real-time PCR. The results revealed that phillyrin not only inhibited the proinflammatory response induced by activated microglia but also attenuated neurological impairment and brain edema in vivo in a mouse TBI model. Additionally, phillyrin suppressed the phosphorylation of NF-κB in microglia after TBI insult. These effects of phillyrin were mostly abolished by the antagonist of PPARγ. Our results reveal that phillyrin could prominently inhibit the inflammation of microglia via the PPARγ signaling pathway, thus leading to potential neuroprotective treatment after traumatic brain injury.


Asunto(s)
Antiinflamatorios/uso terapéutico , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Encéfalo/metabolismo , Glucósidos/uso terapéutico , Inflamación/tratamiento farmacológico , Microglía/inmunología , PPAR gamma/metabolismo , Animales , Encéfalo/patología , Células Cultivadas , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Fármacos Neuroprotectores , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA