Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Neurobiol ; 60(10): 5578-5591, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37326902

RESUMEN

Persistent reprogramming of epigenetic pattern leads to changes in gene expression observed in many neurological disorders. Transient receptor potential cation channel subfamily A member 1 (TRPA1), a member of the TRP channels superfamily, is activated by many migraine triggers and expressed in trigeminal neurons and brain regions that are important in migraine pathogenesis. TRP channels change noxious stimuli into pain signals with the involvement of epigenetic regulation. The expression of the TRPA1 encoding gene, TRPA1, is modulated in pain-related syndromes by epigenetic alterations, including DNA methylation, histone modifications, and effects of non-coding RNAs: micro RNAs (miRNAs), long non-coding RNAs, and circular RNAs. TRPA1 may change epigenetic profile of many pain-related genes as it may modify enzymes responsible for epigenetic modifications and expression of non-coding RNAs. TRPA1 may induce the release of calcitonin gene related peptide (CGRP), from trigeminal neurons and dural tissue. Therefore, epigenetic regulation of TRPA1 may play a role in efficacy and safety of anti-migraine therapies targeting TRP channels and CGRP. TRPA1 is also involved in neurogenic inflammation, important in migraine pathogenesis. The fundamental role of TRPA1 in inflammatory pain transmission may be epigenetically regulated. In conclusion, epigenetic connections of TRPA1 may play a role in efficacy and safety of anti-migraine therapy targeting TRP channels or CGRP and they should be further explored for efficient and safe antimigraine treatment. This narrative/perspective review presents information on the structure and functions of TRPA1 as well as role of its epigenetic connections in pain transmission and potential in migraine therapy.


Asunto(s)
Trastornos Migrañosos , Canales de Potencial de Receptor Transitorio , Humanos , Canal Catiónico TRPA1/metabolismo , Péptido Relacionado con Gen de Calcitonina/genética , Inflamación Neurogénica/genética , Epigénesis Genética , Canales de Potencial de Receptor Transitorio/genética , Canales de Potencial de Receptor Transitorio/metabolismo , Dolor/tratamiento farmacológico , Dolor/genética , Trastornos Migrañosos/genética , Trastornos Migrañosos/metabolismo
2.
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
3.
Front Immunol ; 11: 456, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32296418

RESUMEN

Neuroinflammation commences decades before Alzheimer's disease (AD) clinical onset and represents one of the earliest pathomechanistic alterations throughout the AD continuum. Large-scale genome-wide association studies point out several genetic variants-TREM2, CD33, PILRA, CR1, MS4A, CLU, ABCA7, EPHA1, and HLA-DRB5-HLA-DRB1-potentially linked to neuroinflammation. Most of these genes are involved in proinflammatory intracellular signaling, cytokines/interleukins/cell turnover, synaptic activity, lipid metabolism, and vesicle trafficking. Proteomic studies indicate that a plethora of interconnected aberrant molecular pathways, set off and perpetuated by TNF-α, TGF-ß, IL-1ß, and the receptor protein TREM2, are involved in neuroinflammation. Microglia and astrocytes are key cellular drivers and regulators of neuroinflammation. Under physiological conditions, they are important for neurotransmission and synaptic homeostasis. In AD, there is a turning point throughout its pathophysiological evolution where glial cells sustain an overexpressed inflammatory response that synergizes with amyloid-ß and tau accumulation, and drives synaptotoxicity and neurodegeneration in a self-reinforcing manner. Despite a strong therapeutic rationale, previous clinical trials investigating compounds with anti-inflammatory properties, including non-steroidal anti-inflammatory drugs (NSAIDs), did not achieve primary efficacy endpoints. It is conceivable that study design issues, including the lack of diagnostic accuracy and biomarkers for target population identification and proof of mechanism, may partially explain the negative outcomes. However, a recent meta-analysis indicates a potential biological effect of NSAIDs. In this regard, candidate fluid biomarkers of neuroinflammation are under analytical/clinical validation, i.e., TREM2, IL-1ß, MCP-1, IL-6, TNF-α receptor complexes, TGF-ß, and YKL-40. PET radio-ligands are investigated to accomplish in vivo and longitudinal regional exploration of neuroinflammation. Biomarkers tracking different molecular pathways (body fluid matrixes) along with brain neuroinflammatory endophenotypes (neuroimaging markers), can untangle temporal-spatial dynamics between neuroinflammation and other AD pathophysiological mechanisms. Robust biomarker-drug codevelopment pipelines are expected to enrich large-scale clinical trials testing new-generation compounds active, directly or indirectly, on neuroinflammatory targets and displaying putative disease-modifying effects: novel NSAIDs, AL002 (anti-TREM2 antibody), anti-Aß protofibrils (BAN2401), and AL003 (anti-CD33 antibody). As a next step, taking advantage of breakthrough and multimodal techniques coupled with a systems biology approach is the path to pursue for developing individualized therapeutic strategies targeting neuroinflammation under the framework of precision medicine.


Asunto(s)
Enfermedad de Alzheimer/genética , Citocinas/genética , Inflamación Neurogénica/genética , Medicina de Precisión/métodos , Animales , Autorrenovación de las Células/genética , Estudio de Asociación del Genoma Completo , Humanos , Metabolismo de los Lípidos/genética , Medicina de Precisión/tendencias , Proteómica , Transducción de Señal/genética , Biología de Sistemas
4.
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
5.
J Immunol Res ; 2020: 9469210, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32258178

RESUMEN

Alzheimer's disease (AD) has been clinically characterized by a progressive degeneration of neurons which resulted in a gradual and irreversible cognitive impairment. The accumulation of Aß and τ proteins in the brain contribute to the severity of the disease. Recently, vitexin compound has been the talk amongst researchers due to its pharmacological properties as anti-inflammation and anti-AD. However, the epigenetic mechanism of the compound in regulating the neuroinflammation activity is yet to be fully elucidated. Hence, this review discusses the potential of vitexin compound to have the pharmacoepigenetic property in regulating the neuroinflammation activity in relation to AD. It is with hope that the review would unveil the potential of vitexin as the candidate in treating AD.


Asunto(s)
Enfermedad de Alzheimer/inmunología , Antiinflamatorios/metabolismo , Apigenina/metabolismo , Inflamación Neurogénica/inmunología , Neuronas/patología , Enfermedad de Alzheimer/genética , Animales , Epigénesis Genética , Humanos , Inflamación Neurogénica/genética
6.
Int Immunopharmacol ; 79: 106091, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31896511

RESUMEN

BACKGROUND: Neuroinflammation is a common therapeutic target for traumatic brain injury (TBI) due to its contribution to delayed secondary cell death and has the potential to occur for years after the initial insult. Previous studies demonstrate that miR-429 is up-regulated in the brain lesions of TBI mice, while its role in regulating neuroinflammation and brain injury remains largely unknown. METHOD: The expression of miR-429 in LPS-activated microglia and microglia in TBI model was detected by RT-PCR. The effects of miR-429 inhibitors on LPS-activated microglia in vitro as well as neurological recovery and post-traumatic neuroinflammatory response in TBI model mice were detected in vivo. RESULTS: LPS and TBI significantly induce the up-expression of miR-429, inflammatory cytokines, MAPK-p38 and phosphorylated NF-κB in microglia, which were all inhibited by miR-429 inhibitors. Meanwhile, miR-429 inhibitors also attenuated the neurological impairment in TBI mice. Bioinformatics analysis showed that miR-429 could target and inhibit the expression of dual specificity protein phosphatase 1 (DUSP1), thus inhibiting the expression of MAPK-p38 and phosphorylated NF-κB. CONCLUSION: miR-429 plays a pro-inflammatory role in activated microglia by targeting DUSP1 signaling pathway. Inhibiting miR-429 can attenuate the inflammatory response of microglia and TBI-mediated brain damage.


Asunto(s)
Lesiones Traumáticas del Encéfalo/metabolismo , MicroARNs/genética , Microglía/metabolismo , Microglía/patología , Inflamación Neurogénica/genética , Neuronas/fisiología , Animales , Lesiones Traumáticas del Encéfalo/genética , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Regeneración , Transducción de Señal
7.
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
8.
EMBO J ; 38(10)2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30944096

RESUMEN

Astrocytes are critical regulators of neuroinflammation in multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis (EAE). Growing evidence indicates that ubiquitination of signaling molecules is an important cell-intrinsic mechanism governing astrocyte function during MS and EAE Here, we identified an upregulation of the deubiquitinase OTU domain, ubiquitin aldehyde binding 1 (OTUB1) in astrocytes during MS and EAE Mice with astrocyte-specific OTUB1 ablation developed more severe EAE due to increased leukocyte accumulation, proinflammatory gene transcription, and demyelination in the spinal cord as compared to control mice. OTUB1-deficient astrocytes were hyperactivated in response to IFN-γ, a fingerprint cytokine of encephalitogenic T cells, and produced more proinflammatory cytokines and chemokines than control astrocytes. Mechanistically, OTUB1 inhibited IFN-γ-induced Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signaling by K48 deubiquitination and stabilization of the JAK2 inhibitor suppressor of cytokine signaling 1 (SOCS1). Thus, astrocyte-specific OTUB1 is a critical inhibitor of neuroinflammation in CNS autoimmunity.


Asunto(s)
Astrocitos/inmunología , Astrocitos/patología , Autoinmunidad/genética , Cisteína Endopeptidasas/fisiología , Interferón gamma/fisiología , Inflamación Neurogénica/genética , Animales , Animales Recién Nacidos , Astrocitos/metabolismo , Células Cultivadas , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/patología , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Femenino , Interferón gamma/antagonistas & inhibidores , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Inflamación Neurogénica/patología , Neuroinmunomodulación/genética
9.
Neurotherapeutics ; 16(1): 216-230, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30225790

RESUMEN

Micro-RNAs (miRs) are short, noncoding RNAs that negatively regulate gene expression at the post-transcriptional level and have been implicated in the pathophysiology of secondary damage after traumatic brain injury (TBI). Among miRs linked to inflammation, miR-155 has been implicated as a pro-inflammatory factor in a variety of organ systems. We examined the expression profile of miR-155, following experimental TBI (controlled cortical impact) in adult male C57Bl/6 mice, as well as the effects of acute or delayed administration of a miR-155 antagomir on post-traumatic neuroinflammatory responses and neurological recovery. Trauma robustly increased miR-155 expression in the injured cortex over 7 days. Similar TBI-induced miR-155 expression changes were also found in microglia/macrophages isolated from the injured cortex at 7 days post-injury. A miR-155 hairpin inhibitor (antagomir; 0.5 nmol), administered intracerebroventricularly (ICV) immediately after injury, attenuated neuroinflammatory markers at both 1 day and 7 days post-injury and reduced impairments in spatial working memory. Delayed ICV infusion of the miR-155 antagomir (0.5 nmol/day), beginning 24 h post-injury and continuing for 6 days, attenuated neuroinflammatory markers at 7 days post-injury and improved motor, but not cognitive, function through 28 days. The latter treatment limited NADPH oxidase 2 expression changes in microglia/macrophages in the injured cortex and reduced cortical lesion volume. In summary, TBI causes a robust and persistent neuroinflammatory response that is associated with increased miR-155 expression in microglia/macrophages, and miR-155 inhibition reduces post-traumatic neuroinflammatory responses and improves neurological recovery. Thus, miR-155 may be a therapeutic target for TBI-related neuroinflammation.


Asunto(s)
Antagomirs/administración & dosificación , Lesiones Traumáticas del Encéfalo , MicroARNs/antagonistas & inhibidores , Inflamación Neurogénica , Animales , Lesiones Traumáticas del Encéfalo/genética , Lesiones Traumáticas del Encéfalo/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Inflamación Neurogénica/genética , Recuperación de la Función/efectos de los fármacos , Recuperación de la Función/genética
10.
Int Immunopharmacol ; 65: 413-421, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30388515

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a rapidly progressing neurodegenerative disease characterized by motor neuron loss in the brain and spinal cord. Mutations in Cu-Zn superoxide dismutase (SOD1) are the first identified genetic mutations that are causative for familial ALS. Soluble SOD1 oligomers are considered the most toxic species and play a key role in the pathologic process of ALS. Here we present a therapeutic strategy for ALS with an oligomer-specific antibody (W20) targeting toxic SOD1 oligomers. Our study showed that W20 significantly improved motor neuron survival and motor performance in SOD1-G93A mouse model of ALS when administrated even at low dose within short time. Further investigation demonstrated that the beneficial effects of W20 resulted from the reduction of SOD1 oligomer levels and the inhibition of gliosis and neuroinflammation in the spinal cords and brain stems of ALS model mice. These findings for the first time suggest that an oligomer-specific antibody has promising therapeutic potential for ALS and open a new way for ALS treatment.


Asunto(s)
Esclerosis Amiotrófica Lateral/terapia , Anticuerpos/uso terapéutico , Gliosis/terapia , Inmunoterapia/métodos , Inflamación Neurogénica/terapia , Médula Espinal/patología , Superóxido Dismutasa/genética , Esclerosis Amiotrófica Lateral/genética , Animales , Modelos Animales de Enfermedad , Gliosis/genética , Humanos , Masculino , Ratones , Ratones Transgénicos , Actividad Motora/efectos de los fármacos , Neuronas Motoras/patología , Inflamación Neurogénica/genética , Superóxido Dismutasa/inmunología
11.
Int J Mol Sci ; 18(8)2017 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-28817088

RESUMEN

Acute central nervous system (CNS) injury, encompassing traumatic brain injury (TBI) and stroke, accounts for a significant burden of morbidity and mortality worldwide, largely attributable to the development of cerebral oedema and elevated intracranial pressure (ICP). Despite this, clinical treatments are limited and new therapies are urgently required to improve patient outcomes and survival. Originally characterised in peripheral tissues, such as the skin and lungs as a neurally-elicited inflammatory process that contributes to increased microvascular permeability and tissue swelling, neurogenic inflammation has now been described in acute injury to the brain where it may play a key role in the secondary injury cascades that evolve following both TBI and stroke. In particular, release of the neuropeptides substance P (SP) and calcitonin gene-related peptide (CGRP) appear to be critically involved. In particular, increased SP expression is observed in perivascular tissue following acute CNS injury, with the magnitude of SP release being related to both the frequency and degree of the insult. SP release is associated with profound blood-brain barrier disruption and the subsequent development of vasogenic oedema, as well as neuronal injury and poor functional outcomes. Inhibition of SP through use of a neurokinin 1 (NK1) antagonist is highly beneficial following both TBI and ischaemic stroke in pre-clinical models. The role of CGRP is more unclear, especially with respect to TBI, with both elevations and reductions in CGRP levels reported following trauma. However, a beneficial role has been delineated in stroke, given its potent vasodilatory effects. Thus, modulating neuropeptides represents a novel therapeutic target in the treatment of cerebral oedema following acute CNS injury.


Asunto(s)
Lesiones Traumáticas del Encéfalo/genética , Péptido Relacionado con Gen de Calcitonina/genética , Inflamación Neurogénica/genética , Sustancia P/genética , Animales , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/fisiopatología , Edema Encefálico/genética , Edema Encefálico/fisiopatología , Lesiones Traumáticas del Encéfalo/fisiopatología , Permeabilidad Capilar/genética , Regulación de la Expresión Génica , Humanos , Inflamación Neurogénica/fisiopatología , Neuronas/patología
12.
J Recept Signal Transduct Res ; 37(5): 447-452, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28553881

RESUMEN

Migraine is one of the most common types of pain associated with sterile inflammatory conditions. Soluble urokinase plasminogen activator receptor (suPAR) is a potential novel inflammatory marker. We aim to determine the association between serum values of suPAR, procalcitonin, fibrinogen, and high-sensitivity C-reactive protein (hs-CRP) and migraine disease characteristics. The study involved a total of 60 migraine patients (33 patients in the interictal period, 27 patients in the attack period) and 30 healthy individuals. The serum values of suPAR were found to be significantly higher in migraine patients in the attack period than in migraine patients in the interictal period, and in healthy individuals (p < .01 for both). In addition, levels of suPAR were determined to be higher in migraine with aura patients than in migraine without aura patients. When we subdivided migraine patients according to frequency of attack (attacks/month), significant differences were found between the suPAR and procalcitonin levels (measured during the attack period) of those in the frequent-attack group (4-5 or more) versus those in the less frequent attack group (less than 4). Serum levels of procalcitonin were shown to be significantly higher in migraine patients during the attack period compared with migraine patients in the interictal period and in control subjects (p = .001 for both). Significant differences were found between plasma levels of fibrinogen in migraine patients versus control subjects (p < .01). No statistically significant difference was found between levels of hs-CRP in migraine patients versus the control group. These findings may show that presenting a high level of suPAR in migraine patients with attack and aura results to predisposition to occurring on the symptoms and that high levels of suPAR, procalcitonin and fibrinogen in patients with migraine result in neurogenic inflammation during migraine headaches.


Asunto(s)
Trastornos Migrañosos/sangre , Migraña con Aura/sangre , Inflamación Neurogénica/sangre , Receptores del Activador de Plasminógeno Tipo Uroquinasa/sangre , Proteína C-Reactiva/metabolismo , Calcitonina/sangre , Femenino , Fibrinógeno/metabolismo , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Masculino , Trastornos Migrañosos/genética , Trastornos Migrañosos/fisiopatología , Migraña con Aura/genética , Migraña con Aura/fisiopatología , Inflamación Neurogénica/genética , Inflamación Neurogénica/fisiopatología , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética
13.
Exp Mol Pathol ; 102(3): 505-514, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28533125

RESUMEN

Due to the limited data on diabetic ketoacidosis and brain edema (DKA/BE) in children/adolescents and the lack of recent data on adults with type 1 diabetes (T1D), we addressed the question of whether neuroinflammation was present in the fatal DKA of adults. We performed immunohistochemistry (IHC) studies on the brains of two young adults with T1D and fatal DKA and compared them with two teenagers with poorly controlled diabetes and fatal DKA. C5b-9, the membrane attack complex (MAC) had significantly greater deposits in the grey and white matter of the teenagers than the young adults (p=0.03). CD59, a MAC assembly inhibitory protein was absent, possibly suppressed by the hyperglycemia in the teenagers but was expressed in the young adults despite comparable average levels of hyperglycemia. The receptor for advanced glycation end products (RAGE) had an average expression in the young adults significantly greater than in the teenagers (p=0.02). The autophagy marker Light Chain 3 (LC3) A/B was the predominant form of programmed cell death (PCD) in the teenage brains. The young adults had high expressions of both LC3A/B and TUNEL, an apoptotic cell marker for DNA fragmentation. BE was present in the newly diagnosed young adult with hyperglycemic hyperosmolar DKA and also in the two teenagers. Our data indicate that significant differences in neuroinflammatory components, initiated by the dysregulation of DKA and interrelated metabolic and immunologic milieu, are likely present in the brains of fatal DKA of teenagers when compared with young adults.


Asunto(s)
Biomarcadores/metabolismo , Diabetes Mellitus Tipo 1/genética , Cetoacidosis Diabética/genética , Inflamación Neurogénica/genética , Adolescente , Adulto , Autofagia , Encéfalo/fisiopatología , Edema Encefálico/diagnóstico , Edema Encefálico/etiología , Edema Encefálico/genética , Antígenos CD59/genética , Antígenos CD59/metabolismo , Fragmentación del ADN , Diabetes Mellitus Tipo 1/complicaciones , Cetoacidosis Diabética/complicaciones , Regulación de la Expresión Génica , Humanos , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Mediadores de Inflamación/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Inflamación Neurogénica/etiología , Adulto Joven
14.
Transl Psychiatry ; 7(5): e1126, 2017 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-28485729

RESUMEN

The histaminergic system (HS) has a critical role in cognition, sleep and other behaviors. Although not well studied in autism spectrum disorder (ASD), the HS is implicated in many neurological disorders, some of which share comorbidity with ASD, including Tourette syndrome (TS). Preliminary studies suggest that antagonism of histamine receptors 1-3 reduces symptoms and specific behaviors in ASD patients and relevant animal models. In addition, the HS mediates neuroinflammation, which may be heightened in ASD. Together, this suggests that the HS may also be altered in ASD. Using RNA sequencing (RNA-seq), we investigated genome-wide expression, as well as a focused gene set analysis of key HS genes (HDC, HNMT, HRH1, HRH2, HRH3 and HRH4) in postmortem dorsolateral prefrontal cortex (DLPFC) initially in 13 subjects with ASD and 39 matched controls. At the genome level, eight transcripts were differentially expressed (false discovery rate <0.05), six of which were small nucleolar RNAs (snoRNAs). There was no significant diagnosis effect on any of the individual HS genes but expression of the gene set of HNMT, HRH1, HRH2 and HRH3 was significantly altered. Curated HS gene sets were also significantly differentially expressed. Differential expression analysis of these gene sets in an independent RNA-seq ASD data set from DLPFC of 47 additional subjects confirmed these findings. Understanding the physiological relevance of an altered HS may suggest new therapeutic options for the treatment of ASD.


Asunto(s)
Trastorno del Espectro Autista/genética , Histamina/genética , Receptores Histamínicos/efectos de los fármacos , Análisis de Secuencia de ARN/métodos , Síndrome de Tourette/genética , Adolescente , Adulto , Anciano , Trastorno del Espectro Autista/metabolismo , Trastorno del Espectro Autista/fisiopatología , Niño , Preescolar , Cognición/fisiología , Diagnóstico , Femenino , Estudio de Asociación del Genoma Completo/métodos , Histamina/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Inflamación Neurogénica/genética , Corteza Prefrontal/metabolismo , Corteza Prefrontal/patología , Sueño/fisiología , Síndrome de Tourette/metabolismo , Síndrome de Tourette/fisiopatología , Transcriptoma/genética , Adulto Joven
15.
Exp Neurol ; 293: 62-73, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28359740

RESUMEN

Vasoactive intestinal peptide (VIP) is one of the neuropeptides showing the strongest up-regulation in the nociceptive pathway after peripheral nerve injury and has been proposed to support neuropathic pain. Nevertheless, the story may be more complicated considering the known suppressive effects of the peptide on the immune reactivity of microglial cells, which have been heavily implicated in the onset and maintenance of pain after nerve injury. We here used mice deficient in VIP and the model of spared nerve injury, characterized by persistent tactile hypersensitivity. While tactile hypersensitivity developed similarly to wild type mice for the ipsilateral hindpaw, only transgenic mice showed a mirror-image tactile hypersensitivity in the contralateral hindpaw. This exacerbated neuropathic pain phenotype appeared to be mediated through a local mechanism acting at the level of the lumbar spinal cord as a distant nerve lesion in the front limb did not lead to hindpaw hypersensitivity in VIP-deficient mice. Innocuous tactile hindpaw stimulation was found to increase a neuronal activation marker in the bilateral superficial laminae of the lumbar dorsal horn of VIP-deficient, but not wild type mice, after SNI. A deeper study into the immune responsiveness to the nerve lesion also proved that VIP-deficient mice had a stronger early pro-inflammatory cytokine response and a more pronounced microglial reactivity compared to wild type controls. The latter was also observed at four weeks after spared nerve injury, a time at which bilateral tactile hypersensitivity persisted in VIP-deficient mice. These data suggest an action of VIP in neuropathic states that is more complicated than previously assumed. Future research is now needed for a deeper understanding of the relative contribution of receptors and fiber populations involved in the VIP-neuropathic pain link.


Asunto(s)
Hiperalgesia/etiología , Hiperalgesia/genética , Inflamación Neurogénica/etiología , Inflamación Neurogénica/genética , Traumatismos de los Nervios Periféricos/complicaciones , Péptido Intestinal Vasoactivo/deficiencia , Animales , Proteínas de Unión al Calcio/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Lateralidad Funcional/genética , Regulación de la Expresión Génica/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Hiperalgesia/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de Microfilamentos/metabolismo , Neuralgia/etiología , Inflamación Neurogénica/metabolismo , Dimensión del Dolor , Proteínas Proto-Oncogénicas c-fos/metabolismo , Factores de Tiempo , Péptido Intestinal Vasoactivo/genética
16.
Int Immunopharmacol ; 46: 156-162, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28284149

RESUMEN

Emerging evidence suggests that microRNAs (miRNAs) play a critical role in the pathogenesis of neuropathic pain. However, the exact role of miRNAs in regulating neuropathic pain remains largely unknown. In this study, we aimed to investigate the potential role of miR-93 in a rat model of neuropathic pain induced by chronic constriction sciatic nerve injury (CCI). We found a significant decrease of miR-93 in the spinal cord of CCI rats compared with sham rats. Overexpression of miR-93 significantly alleviated neuropathic pain development and reduced inflammatory cytokine expression, including interleukin (IL)-1ß, tumor necrosis factor (TNF)-α, and IL-6 in CCI rats. By bioinformatic analysis and dual-luciferase reporter assay, we found that miR-93 directly targeted the 3'-untranslated region (UTR) of signal transducer and activator of transcription 3 (STAT3), an important regulator of inflammation. Overexpression of miR-93 markedly suppressed the expression of STAT3 in vitro and in vivo. Furthermore, overexpression of STAT3 significantly reversed the miR-93 overexpression-induced suppressive effects on neuropathic pain development and neuroinflammation. Taken together, our study suggests that miR-93 inhibits neuropathic pain development of CCI rats possibly through inhibiting STAT3-mediated neuroinflammation. Our findings indicate that miR-93 may serve as a novel therapeutic target for neuropathic pain intervention.


Asunto(s)
Constricción Patológica/genética , MicroARNs/genética , Neuralgia/genética , Inflamación Neurogénica , Factor de Transcripción STAT3/genética , Neuropatía Ciática/genética , Médula Espinal/fisiología , Regiones no Traducidas 3'/genética , Animales , Células Cultivadas , Biología Computacional , Constricción Patológica/terapia , Citocinas/metabolismo , Regulación hacia Abajo , Femenino , Humanos , Mediadores de Inflamación/metabolismo , Neuralgia/terapia , Inflamación Neurogénica/genética , Ratas , Ratas Sprague-Dawley , Factor de Transcripción STAT3/metabolismo , Neuropatía Ciática/terapia
17.
Biosci Rep ; 36(6)2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27852797

RESUMEN

Increasing evidence supports the involvement of inflammatory and immune processes in temporal lobe epilepsy (TLE). miRNAs represent small regulatory RNA molecules that have been shown to act as negative regulators of gene expression controlling different biological processes, including immune system homoeostasis and function. We investigated the expression and cellular distribution of miRNA-146a (miR-146a) in a rat model of TLE. Prominent up-regulation of miR-146a activation was evident in 1 week after status epilepticus (SE) and persisted in the chronic phase. The predicted miR-146a's target complement factor H (CFH) mRNA and protein expression was also down-regulated in TLE rat model. Furthermore, transfection of miR-146a mimics in neuronal and glial cells down-regulated CFH mRNA and protein levels respectively. Luciferase reporter assays demonstrated that miR-146a down-regulated CFH mRNA expression via 3'-UTR pairing. Down-regulating miR-146a by intracerebroventricular injection of antagomir-146a enhanced the hippocampal expression of CFH in TLE model and decreased seizure susceptibility. These findings suggest that immunopathological deficits associated with TLE can in part be explained by a generalized miR-146a-mediated down-regulation of CFH that may contribute to epileptogenesis in a rat model of TLE.


Asunto(s)
Factor H de Complemento/metabolismo , Epilepsia del Lóbulo Temporal/metabolismo , Hipocampo/metabolismo , MicroARNs/metabolismo , Inflamación Neurogénica/metabolismo , Animales , Antagomirs/administración & dosificación , Factor H de Complemento/genética , Modelos Animales de Enfermedad , Regulación hacia Abajo , Epilepsia del Lóbulo Temporal/genética , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Humanos , Inyecciones Intraventriculares , Masculino , MicroARNs/genética , Inflamación Neurogénica/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Regulación hacia Arriba
18.
Am J Chin Med ; 44(1): 119-32, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26916918

RESUMEN

Dendropanax morbifera (D. morbifera), a species endemic to Korea, is largely distributed throughout the southern part of the country. Its leaves, stems, roots, and seeds have been used as a form of alternative medicine for various diseases and neurological disorders including paralysis, stroke, and migraine. However, the molecular mechanisms that underlie the remedial effects of D. morbifera remain largely unknown. In this paper, extracts from D. morbifera leaves were prepared using ethyl acetate as a solvent (abbreviated as DMLE). The modulatory effects of DMLE on neuroinflammation were studied in a lipopolysaccharide (LPS)-stimulated BV2 murine microglial cell line. Production of pro-inflammatory cytokines, activation of mitogen-activated protein kinases (MAPKs) and nuclear factor-kappa B (NF-[Formula: see text]B), and different M1/M2 activation states of microglia were examined. DMLE treatment suppressed the production of pro-inflammatory cytokines including tumor necrosis factor-[Formula: see text] (TNF-[Formula: see text]), interleukin-6 (IL-6), and nitric oxide (NO) in LPS-stimulated BV2 cells. DMLE treatment also attenuated the activation of MAPKs and NF-[Formula: see text]B. In a novel discovery, we found that DMLE up-regulated the marker genes representing an alternative, anti-inflammatory M2 polarization, while suppressing the expression of the classical, pro-inflammatory M1 activation state genes. Here, we uncovered the cellular mechanisms underlying the beneficial effects of D. morbifera against neuroinflammation using BV2 microglia cells. These results strongly suggest that DMLE was able to counter the effects of LPS on BV2 cells via control of microglia polarization states. Additionally, study results indicated that DMLE may have therapeutic potential as a neuroinflammation-suppressing treatment for neurodegenerative diseases.


Asunto(s)
Araliaceae/química , Microglía/patología , Inflamación Neurogénica/tratamiento farmacológico , Fitoterapia , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico , Animales , Polaridad Celular/efectos de los fármacos , Polaridad Celular/genética , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Citocinas/metabolismo , Mediadores de Inflamación/metabolismo , Interleucina-6/metabolismo , Lipopolisacáridos/efectos adversos , Ratones , Microglía/metabolismo , Terapia Molecular Dirigida , Enfermedades Neurodegenerativas/tratamiento farmacológico , Inflamación Neurogénica/inducido químicamente , Inflamación Neurogénica/genética , Óxido Nítrico/metabolismo , Hojas de la Planta , Factor de Necrosis Tumoral alfa/metabolismo
19.
Neurobiol Aging ; 36(11): 3008-3019, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26362942

RESUMEN

The modulation of endocannabinoid (EC) levels and the activation of cannabinoid receptors are seen as promising therapeutic strategies in a variety of diseases, including Alzheimer's disease (AD). We aimed to evaluate the effect of the pharmacologic and genetic inhibition of anandamide-degrading enzyme in a mouse model of AD (5xFAD). Pharmacologic inhibition of the fatty acid amide hydrolase (FAAH) had little impact on the expression of key enzymes and cytokines and also on the cognitive impairment, plaque deposition, and gliosis in 5xFAD mice. CB1 blockade exacerbated inflammation in this transgenic mouse model of AD. The genetic inactivation of FAAH led to increases in the expression of inflammatory cytokines. At the same time, FAAH-null 5xFAD mice exhibited a behavioral improvement in spatial memory that was independent of the level of anxiety and was not CB1 mediated. Finally, mice lacking FAAH showed diminished soluble amyloid levels, neuritic plaques, and gliosis. These data reinforce the notion of a role for the EC system in neuroinflammation and open new perspectives on the relevance of modulating EC levels in the inflamed brain.


Asunto(s)
Enfermedad de Alzheimer/genética , Endocannabinoides/metabolismo , Inflamación Neurogénica/etiología , Inflamación Neurogénica/genética , Receptores de Cannabinoides/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Amidohidrolasas/genética , Amidohidrolasas/metabolismo , Amidohidrolasas/fisiología , Amiloide/metabolismo , Animales , Modelos Animales de Enfermedad , Endocannabinoides/fisiología , Femenino , Gliosis , Mediadores de Inflamación/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Terapia Molecular Dirigida , Placa Amiloide , Receptores de Cannabinoides/fisiología , Memoria Espacial
20.
J Invest Dermatol ; 135(9): 2209-2218, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25905588

RESUMEN

Although pituitary adenylate cyclase-activating polypeptide (PACAP) was described as a key vasoregulator in human skin, little is known about its expression in mouse skin. As it is important to investigate PACAP signaling in translational mouse dermatitis models, we determined its presence, regulation, and role in neurogenic and non-neurogenic cutaneous inflammatory mechanisms. The mRNA of PACAP and its specific receptor PAC1 was detected with real-time PCR in several skin regions at comparable levels. PACAP-38-immunoreactivity measured with radioimmunoassay was similar in plantar and dorsal paw skin and the ear but significantly smaller in the back skin. PACAP and PAC1 mRNA, as well as PACAP-38 and PAC1 protein expression, significantly increased in the plantar skin after intraplantar administration of capsaicin (50 µl, 100 µg ml(-1)), an agonist of the transient receptor potential vanilloid 1 (TRPV1) receptor, evoking chiefly neurogenic inflammation without inflammatory cell accumulation. Intraplantar complete Freund's adjuvant (CFA; 50 µl, 1 mg ml(-1)) also increased PACAP/PAC1 mRNA but not the PACAP peptide. Capsaicin-induced neurogenic paw edema, but not CFA-evoked non-neurogenic swelling, was significantly smaller in PACAP-deficient mice throughout a 24-hour period. To our knowledge, we provide previously unreported evidence for PACAP and PAC1 expression upregulation during skin inflammation of different mechanisms and for its pro-inflammatory function in neurogenic edema formation.


Asunto(s)
Dermatitis/patología , Inflamación Neurogénica/genética , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/genética , Canales Catiónicos TRPV/farmacología , Análisis de Varianza , Animales , Capsaicina/farmacología , Dermatitis/genética , Dermatitis/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Inflamación Neurogénica/inducido químicamente , Inflamación Neurogénica/fisiopatología , ARN Mensajero/análisis , Radioinmunoensayo , Distribución Aleatoria , Estadísticas no Paramétricas , Activación Transcripcional , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...