Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 443
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
2.
Aging (Albany NY) ; 13(21): 23913-23935, 2021 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-34731090

RESUMEN

LianHuaQingWen (LHQW) improves clinical symptoms and alleviates the severity of COVID-19, but the mechanism is unclear. This study aimed to investigate the potential molecular targets and mechanisms of LHQW in treating COVID-19 using a network pharmacology-based approach and molecular docking analysis. The main active ingredients, therapeutic targets of LHQW, and the pathogenic targets of COVID-19 were screened using the TCMSP, UniProt, STRING, and GeneCards databases. According to the "Drug-Ingredients-Targets-Disease" network, Interleukin 6 (IL6) was identified as the core target, and quercetin, luteolin, and wogonin as the active ingredients of LHQW associated with IL6. The response to lipopolysaccharide was the most significant biological process identified by gene ontology enrichment analysis, and AGE-RAGE signaling pathway activation was prominent based on the interaction between LHQW and COVID-19. Protein-protein docking analysis showed that IL6 receptor (IL6R)/IL6/IL6 receptor subunit beta (IL6ST) and Spike protein were mainly bound via conventional hydrogen bonds. Furthermore, protein-small molecule docking showed that all three active ingredients could bind stably in the binding model of IL6R/IL6 and IL6ST. Our findings suggest that LHQW may inhibit the lipopolysaccharide-mediated inflammatory response and regulate the AGE-RAGE signaling pathway through IL6. In addition, the N-terminal domain of the S protein of COVID-19 has a good binding activity to IL6ST, and quercetin and wogonin in LHQW may affect IL6ST-mediated IL6 signal transduction and a large number of signaling pathways downstream to other cytokines by directly affecting protein-protein interaction. These findings suggest the potential molecular mechanism by which LHQW inhibits COVID-19 through the regulation of IL6R/IL6/IL6ST.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , COVID-19 , Medicamentos Herbarios Chinos/farmacología , Productos Finales de Glicación Avanzada/metabolismo , Interleucina-6/metabolismo , Receptor para Productos Finales de Glicación Avanzada/metabolismo , SARS-CoV-2 , Antivirales/farmacología , COVID-19/inmunología , Receptor gp130 de Citocinas/metabolismo , Flavanonas/farmacología , Humanos , Luteolina/farmacología , Simulación del Acoplamiento Molecular , Quercetina/farmacología , Receptores de Interleucina-6/metabolismo , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Glicoproteína de la Espiga del Coronavirus/metabolismo
3.
J Immunol Res ; 2021: 1125199, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34595242

RESUMEN

BACKGROUND: Acute lung injury (ALI) always leads to severe inflammation. As inflammation and oxidative stress are the common pathological basis of endotoxin-induced inflammatory injury and ischemic reperfusion injury (IRI), we speculate that remote ischemic preconditioning (RIPC) can be protective for ALI when used as remote inflammatory preconditioning (RInPC). METHOD: A total of 21 Sprague-Dawley rats were used for the animal experiments. Eighteen rats were equally and randomly divided into the control (NS injection), LPS (LPS injection), and RInPC groups. The RInPC was performed prior to the LPS injection via tourniquet blockage of blood flow to the right hind limb and adopted three cycles of 5 min tying followed by 5 min untying. Animals were sacrificed 24 hours later. There were 2 rats in the LPS group and 1 in the RInPC group who died before the end of the experiment. Supplementary experiments in the LPS and RInPC groups were conducted to ensure that 6 animals in each group reached the end of the experiment. RESULTS: In the present study, we demonstrated that the RInPC significantly attenuated the LPS-induced ALI in rats. Apoptotic cells were reduced significantly by the RInPC, with the simultaneous improvement of apoptosis-related proteins. Reduction of MPO and MDA and increasing of SOD activity were found significantly improved by the RInPC. Increasing of TNF-α, IL-1ß, and IL-6 induced by the LPS was inhibited, while IL-10 was significantly increased by RInPC, compared to the LPS group. CONCLUSION: RInPC could inhibit inflammation and attenuate oxidative stress, thereby reducing intrinsic apoptosis and providing lung protection in the LPS-induced ALI in rats.


Asunto(s)
Lesión Pulmonar Aguda/inmunología , Apoptosis/inmunología , Precondicionamiento Isquémico/métodos , Pulmón/inmunología , Transducción de Señal/inmunología , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/metabolismo , Animales , Caspasas/inmunología , Caspasas/metabolismo , Citocinas/inmunología , Citocinas/metabolismo , Lipopolisacáridos , Pulmón/metabolismo , Pulmón/patología , Malondialdehído/inmunología , Malondialdehído/metabolismo , Peroxidasa/inmunología , Peroxidasa/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/inmunología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas Sprague-Dawley , Superóxido Dismutasa/inmunología , Superóxido Dismutasa/metabolismo , Proteína X Asociada a bcl-2/inmunología , Proteína X Asociada a bcl-2/metabolismo
4.
Signal Transduct Target Ther ; 6(1): 331, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34471099

RESUMEN

The recently emerged severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which is the causative agent of ongoing global pandemic of COVID-19, may trigger immunosuppression in the early stage and overactive immune response in the late stage of infection; However, the underlying mechanisms are not well understood. Here we demonstrated that the SARS-CoV-2 nucleocapsid (N) protein dually regulated innate immune responses, i.e., the low-dose N protein suppressed type I interferon (IFN-I) signaling and inflammatory cytokines, whereas high-dose N protein promoted IFN-I signaling and inflammatory cytokines. Mechanistically, the SARS-CoV-2 N protein dually regulated the phosphorylation and nuclear translocation of IRF3, STAT1, and STAT2. Additionally, low-dose N protein combined with TRIM25 could suppress the ubiquitination and activation of retinoic acid-inducible gene I (RIG-I). Our findings revealed a regulatory mechanism of innate immune responses by the SARS-CoV-2 N protein, which would contribute to understanding the pathogenesis of SARS-CoV-2 and other SARS-like coronaviruses, and development of more effective strategies for controlling COVID-19.


Asunto(s)
COVID-19/inmunología , Proteínas de la Nucleocápside de Coronavirus/inmunología , Inmunidad Innata , SARS-CoV-2/inmunología , Transducción de Señal/inmunología , Células A549 , COVID-19/patología , Células CACO-2 , Células HEK293 , Células Hep G2 , Humanos , Interferón Tipo I/inmunología , Fosfoproteínas/inmunología
5.
Int Immunopharmacol ; 98: 107913, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34218218

RESUMEN

The objective of this work was to study the effects and mechanisms of S-allylmercapto-N-acetylcysteine (ASSNAC) in the treatment of pulmonary emphysema based on network pharmacology analysis and other techniques. Firstly, the potential targets associated with ASSNAC and COPD were integrated using public databases. Then, a protein-protein interaction network was constructed using String database and Cytoscape software. The Gene Ontology analysis and Kyoto Encyclopedia of Genes and Genomes pathway analysis were performed on DAVID platform. The molecular docking of ASSNAC with some key disease targets was implemented on the SwissDock platform. To verify the results of the network pharmacology, a pulmonary emphysema mice model was established and treated with ASSNAC. Besides, the expressions of the predicted targets were detected by immunohistochemistry, Western blot analysis or enzyme-linked immunosorbent assay. Results showed that 33 overlapping targets are achieved, including CXCL8, ICAM1, MAP2K1, PTGS2, ACE and so on. The critical pathways of ASSNAC against COPD involved arachidonic acid metabolism, chemokine pathway, MAPK pathway, renin-angiotensin system, and others. Pharmacodynamic experiments demonstrated that ASSNAC decreased the pulmonary emphysema and inflammation in the pulmonary emphysema mice. Therefore, these results confirm the perspective of network pharmacology in the target verification, and indicate the treatment potential of ASSNAC against COPD.


Asunto(s)
Acetilcisteína/análogos & derivados , Compuestos Alílicos/farmacología , Antiinflamatorios/farmacología , Enfisema Pulmonar/tratamiento farmacológico , Acetilcisteína/farmacología , Acetilcisteína/uso terapéutico , Compuestos Alílicos/uso terapéutico , Animales , Antiinflamatorios/uso terapéutico , Líquido del Lavado Bronquioalveolar/citología , Líquido del Lavado Bronquioalveolar/inmunología , Quimiocinas/metabolismo , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Humanos , Masculino , Ratones , Simulación del Acoplamiento Molecular , Farmacología en Red , Mapeo de Interacción de Proteínas , Mapas de Interacción de Proteínas/efectos de los fármacos , Mapas de Interacción de Proteínas/inmunología , Enfisema Pulmonar/diagnóstico , Enfisema Pulmonar/inmunología , Enfisema Pulmonar/patología , Sistema Renina-Angiotensina/efectos de los fármacos , Sistema Renina-Angiotensina/inmunología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología
6.
JCI Insight ; 6(13)2021 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-34236052

RESUMEN

Iron is an essential nutrient for mammals as well as for pathogens. Inflammation-driven changes in systemic and cellular iron homeostasis are central for host-mediated antimicrobial strategies. Here, we studied the role of the iron storage protein ferritin H (FTH) for the control of infections with the intracellular pathogen Salmonella enterica serovar Typhimurium by macrophages. Mice lacking FTH in the myeloid lineage (LysM-Cre+/+Fthfl/fl mice) displayed impaired iron storage capacities in the tissue leukocyte compartment, increased levels of labile iron in macrophages, and an accelerated macrophage-mediated iron turnover. While under steady-state conditions, LysM-Cre+/+Fth+/+ and LysM-Cre+/+Fthfl/fl animals showed comparable susceptibility to Salmonella infection, i.v. iron supplementation drastically shortened survival of LysM-Cre+/+Fthfl/fl mice. Mechanistically, these animals displayed increased bacterial burden, which contributed to uncontrolled triggering of NF-κB and inflammasome signaling and development of cytokine storm and death. Importantly, pharmacologic inhibition of the inflammasome and IL-1ß pathways reduced cytokine levels and mortality and partly restored infection control in iron-treated ferritin-deficient mice. These findings uncover incompletely characterized roles of ferritin and cellular iron turnover in myeloid cells in controlling bacterial spread and for modulating NF-κB and inflammasome-mediated cytokine activation, which may be of vital importance in iron-overloaded individuals suffering from severe infections and sepsis.


Asunto(s)
Apoferritinas , Susceptibilidad a Enfermedades/metabolismo , Inflamación , Hierro , Macrófagos , Infecciones por Salmonella , Salmonella typhimurium/inmunología , Animales , Apoferritinas/deficiencia , Apoferritinas/metabolismo , Inmunidad Innata , Inflamasomas/metabolismo , Inflamación/metabolismo , Inflamación/microbiología , Interleucina-1beta/inmunología , Hierro/inmunología , Hierro/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Infecciones por Salmonella/inmunología , Infecciones por Salmonella/metabolismo , Transducción de Señal/inmunología
7.
J Immunol Res ; 2021: 8029963, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34258300

RESUMEN

BACKGROUND: Asthma is a complex inflammatory disorder that plagues a large number of people. Schisandrin B is an active ingredient of the traditional Chinese herbal medicine Schisandra with various proven physiological activities such as anti-inflammatory and antioxidant activities. In this study, we explored the anti-inflammatory and antioxidant effects and provided the mechanistic insights into the activity of schisandrin B in a mouse model of ovalbumin- (OVA-) induced allergic asthma. METHODS: Male BALB/c mice were sensitized and challenged with OVA to induce asthma and treated with various doses (15 mg/kg, 30 mg/kg, and 60 mg/kg) of SCH to alleviate the features of allergic asthma, airway hyperresponsiveness, inflammatory response, OVA-specific immunoglobulin (Ig)E level, and pathological injury. RESULTS: Schisandrin B significantly attenuated the airway hyperresponsiveness induced by OVA. Moreover, schisandrin B administration suppressed inflammatory responses, reduced the level of IgE, and attenuated pathological injury. Mechanistically, schisandrin B treatment promoted the activation of nuclear erythroid 2-related factor 2 (Nrf2), but suppressed the stimulation of the NF-κB pathway caused by OVA. CONCLUSION: Taken together, our study suggests that schisandrin B attenuates the features of asthmatic lungs by inhibiting the NF-κB pathway and activating the Nrf2 signaling pathway.


Asunto(s)
Asma/tratamiento farmacológico , Lignanos/farmacología , Factor 2 Relacionado con NF-E2/agonistas , FN-kappa B/antagonistas & inhibidores , Compuestos Policíclicos/farmacología , Animales , Asma/diagnóstico , Asma/inmunología , Asma/patología , Líquido del Lavado Bronquioalveolar/citología , Líquido del Lavado Bronquioalveolar/inmunología , Ciclooctanos/farmacología , Ciclooctanos/uso terapéutico , Modelos Animales de Enfermedad , Humanos , Lignanos/uso terapéutico , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/patología , Masculino , Ratones , Factor 2 Relacionado con NF-E2/metabolismo , FN-kappa B/metabolismo , Ovalbúmina/administración & dosificación , Ovalbúmina/inmunología , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/inmunología , Compuestos Policíclicos/uso terapéutico , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Organismos Libres de Patógenos Específicos
8.
Front Immunol ; 12: 697663, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34249001

RESUMEN

The prevalence of chronic inflammatory diseases including inflammatory bowel disease (IBD), autoimmunity and cancer have increased in recent years. Herbal-based compounds such as flavonoids have been demonstrated to contribute to the modulation of these diseases although understanding their mechanism of action remains limited. Flavonoids are able to interact with cellular immune components in a distinct way and influence immune responses at a molecular level. In this mini review, we highlight recent progress in our understanding of the modulation of immune responses by the aryl hydrocarbon receptor (AhR), a ligand-dependent transcription factor whose activity can be regulated by diverse molecules including flavonoids. We focus on the role of AhR in integrating signals from flavonoids to modulate inflammatory responses using in vitro and experimental animal models. We also summarize the limitations of these studies. Medicinal herbs have been widely used to treat inflammatory disorders and may offer a valuable therapeutic strategy to treat aberrant inflammatory responses by modulation of the AhR pathway.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/inmunología , Inmunomodulación , Fitoterapia , Plantas Medicinales/química , Receptores de Hidrocarburo de Aril/inmunología , Animales , Enfermedades Autoinmunes/tratamiento farmacológico , Enfermedades Autoinmunes/inmunología , Flavonoides/química , Flavonoides/inmunología , Flavonoides/uso terapéutico , Humanos , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/inmunología , Ligandos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología
9.
Hepatology ; 74(6): 3110-3126, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34322898

RESUMEN

BACKGROUND AND AIMS: NASH is an advanced stage of liver disease accompanied by lipid accumulation, inflammation, and liver fibrosis. Guanine nucleotide-binding protein G(i) subunit alpha-2 (GNAI2) is a member of the "inhibitory" class of α-subunits, and recent studies showed that Gnai2 deficiency is known to cause reduced weight in mice. However, the role of GNAI2 in hepatocytes, particularly in the context of liver inflammation and lipid metabolism, remains to be elucidated. Herein, we aim to ascertain the function of GNAI2 in hepatocytes and its impact on the development of NASH. APPROACH AND RESULTS: Human liver tissues were obtained from NASH patients and healthy persons to evaluate the expression and clinical relevance of GNAI2. In addition, hepatocyte-specific Gnai2-deficient mice (Gnai2hep-/- ) were fed either a Western diet supplemented with fructose in drinking water (WDF) for 16 weeks or a methionine/choline-deficient diet (MCD) for 6 weeks to investigate the regulatory role and underlying mechanism of Gnai2 in NASH. GNAI2 was significantly up-regulated in liver tissues of patients with NASH. Following feeding with WDF or MCD diets, livers from Gnai2hep-/- mice had reduced steatohepatitis with suppression of markers of inflammation and an increase in lipophagy compared to Gnai2flox/flox mice. Toll-like receptor 4 signals through nuclear factor kappa B to trigger p65-dependent transcription of Gnai2. Intriguingly, immunoprecipitation, immunofluorescence, and mass spectrometry identified peroxiredoxin 1 (PRDX1) as a binding partner of GNAI2. Moreover, the function of PRDX1 in the suppression of TNF receptor-associated factor 6 ubiquitin-ligase activity and glycerophosphodiester phosphodiesterase domain-containing 5-related phosphatidylcholine metabolism was inhibited by GNAI2. Suppression of GNAI2 combined with overexpression of PRDX1 reversed the development of steatosis and fibrosis in vivo. CONCLUSIONS: GNAI2 is a major regulator that leads to the development of NASH. Thus, inhibition of GNAI2 could be an effective therapeutic target for the treatment of NASH.


Asunto(s)
Subunidad alfa de la Proteína de Unión al GTP Gi2/metabolismo , Enfermedad del Hígado Graso no Alcohólico/inmunología , Peroxirredoxinas/metabolismo , Adulto , Animales , Autofagia/inmunología , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Femenino , Subunidad alfa de la Proteína de Unión al GTP Gi2/genética , Hepatocitos , Humanos , Hígado/inmunología , Hígado/patología , Masculino , Ratones , Ratones Noqueados , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Unión Proteica/inmunología , Transducción de Señal/inmunología , Adulto Joven
10.
Int J Med Sci ; 18(12): 2480-2492, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34104079

RESUMEN

Background: Trans-cinnamaldehyde (tCA), a bioactive component found in Cinnamomum cassia, has been reported to exhibit anti-inflammatory and antioxidant effects, but its efficacy in muscle cells has yet to be found. In this study, we investigated the inhibitory effect of tCA on inflammatory and oxidative stress induced by lipopolysaccharide (LPS) in C2C12 mouse skeletal myoblasts. Methods: To investigate the anti-inflammatory and antioxidant effects of tCA in LPS-treated C2C12 cells, we measured the levels of pro-inflammatory mediator, cytokines, and reactive oxygen species (ROS). To elucidate the mechanism underlying the effect of tCA, the expression of genes involved in the expression of inflammatory and oxidative regulators was also investigated. We further evaluated the anti-inflammatory and antioxidant efficacy of tCA against LPS in the zebrafish model. Results: tCA significantly inhibited the LPS-induced release of pro-inflammatory mediators and cytokines, which was associated with decreased expression of their regulatory genes. tCA also suppressed the expression of Toll-like receptor 4 (TLR4) and myeloid differentiation factor, and attenuated the nuclear translocation of nuclear factor-kappa B (NF-κB) and the binding of LPS to TLR4 on the cell surface in LPS-treated C2C12 cells. Furthermore, tCA abolished LPS-induced generation of ROS and expression levels of ROS producing enzymes, NADPH oxidase 1 (NOX1) and NOX2. However, tCA enhanced the activation of nuclear translocation of nuclear factor-E2-related factor 2 (Nrf2) and the expression of heme oxygenase-1 (HO-1) in LPS-stimulated C2C12 myoblasts. In addition, tCA showed strong protective effects against NO and ROS production in LPS-injected zebrafish larvae. Conclusions: Our findings suggest that tCA exerts its inhibitory ability against LPS-induced inflammatory and antioxidant stress in C2C12 myoblasts by targeting the TLR4/NF-κB, which might be mediated by the NOXs and Nrf2/HO-1 pathways.


Asunto(s)
Acroleína/análogos & derivados , Antiinflamatorios/farmacología , Antioxidantes/farmacología , Inflamación/tratamiento farmacológico , Estrés Oxidativo/efectos de los fármacos , Acroleína/farmacología , Acroleína/uso terapéutico , Animales , Antiinflamatorios/uso terapéutico , Antioxidantes/uso terapéutico , Línea Celular , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Inflamación/inmunología , Lipopolisacáridos/inmunología , Ratones , Mioblastos , FN-kappa B/metabolismo , Estrés Oxidativo/inmunología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Receptor Toll-Like 4/metabolismo , Pez Cebra
11.
J Immunol ; 206(12): 2791-2802, 2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-34127520

RESUMEN

Murine models to elucidate the pathogenesis of pollen food allergy syndrome (PFAS), characterized by oral hypersensitivity symptoms induced by specific foods in patients previously sensitized with a pollen, are lacking. The study aimed to examine PFAS pathogenesis in a novel murine model. Birch pollen-immunized mice were orally administered apple extract, and oral symptoms were evaluated based on oral rubbing frequency following the challenge. The birch pollen-immunized mice orally challenged with apple extract exhibited PFAS-like symptoms, including oral rubbing and positive reaction of swelling by the prick test. The apple extract administered with a protease inhibitor reduced the oral rubbing frequency, which was also significantly reduced in the immunized Fcer1a -/- and mast cell-deficient mice compared with the immunized control mice. The oral rubbing frequency, serum IgE levels, and Th2-cytokine production by the cervical lymph node cells were significantly reduced in the immunized Il-33 -/- and thymic stromal lymphopoietin receptor-deficient (Crlf2 -/-) mice as compared with the immunized wild-type mice. IL-33 and thymic stromal lymphopoietin involve the pathogenesis of PFAS. The apple-extract stimulation did not lead to increased Th2-cytokine production in the oral mucosa or number of group 2 innate lymphoid cells or eosinophils. PFAS involves an early-phase response by mast cell degranulation via IgE signaling after the cross-reactivity of Bet v 1-specific IgE and the food allergen, and exacerbation of allergic symptom via proteases in food; PFAS does not involve a late phase with local Th2/eosinophilic inflammation in the oral mucosa. This novel murine model might be used for elucidating the pathogenesis and assessing new therapeutic strategies for PFAS.


Asunto(s)
Citocinas/inmunología , Modelos Animales de Enfermedad , Células Epiteliales/inmunología , Hipersensibilidad a los Alimentos/inmunología , Inmunoglobulina E/inmunología , Polen/inmunología , Animales , Mastocitos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Transducción de Señal/inmunología
12.
Int Immunopharmacol ; 98: 107882, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34182245

RESUMEN

Daphnetin (7, 8-dihydroxycoumarin, DAPH), a coumarin derivative isolated from Daphne odora var., recently draws much more attention as a promising drug candidate to treat neuroinflammatory diseases due to its protective effects against neuroinflammation. However, itscontribution to chronic inflammatory pain is largely unknown. In the current work, we investigated the effects of DAPH in a murine model of inflammatory pain induced by complete Freund's adjuvant (CFA) and its possible underlying mechanisms. Our results showed that DAPH treatment significantly attenuated mechanical allodynia provoked by CFA. A profound inhibition of spinal glial activation, followed by attenuated expression levels of spinal pro-inflammatory cytokines, was observed in DAPH-treated inflammatory pain mice. Further study demonstrated that DAPH mediated negative regulation of spinal NF-κB pathway, as well as its preferential activation of Nrf2/HO-1 signaling pathway in inflammatory pain mice. This study, for the first time, indicated that DAPH might preventthe development of mechanical allodynia in mice with inflammatory pain. And more importantly, these data provide evidence for the potential application of DAPH in the treatment of chronic inflammatory pain.


Asunto(s)
Dolor Crónico/tratamiento farmacológico , Hiperalgesia/tratamiento farmacológico , Dolor/tratamiento farmacológico , Umbeliferonas/farmacología , Animales , Dolor Crónico/inmunología , Dolor Crónico/patología , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Adyuvante de Freund/administración & dosificación , Adyuvante de Freund/inmunología , Hemo-Oxigenasa 1/metabolismo , Humanos , Hiperalgesia/inmunología , Hiperalgesia/patología , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Factor 2 Relacionado con NF-E2/metabolismo , FN-kappa B/metabolismo , Neuroglía/efectos de los fármacos , Neuroglía/inmunología , Neuroglía/patología , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/inmunología , Dolor/inmunología , Dolor/patología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Médula Espinal/efectos de los fármacos , Médula Espinal/inmunología , Médula Espinal/patología , Umbeliferonas/uso terapéutico
13.
Neurosci Lett ; 758: 136002, 2021 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-34090938

RESUMEN

BACKGROUND: Epilepsy is a common neurological disease that cannot be well controlled by existing antiepileptic drugs. Studies have implicated oxidative stress and inflammation in the pathophysiology of epilepsy. Rhein has a comprehensive pharmacological function in reducing inflammation and can play a neuroprotective role in many neurological diseases, however little is known about its effects on epilepsy. METHODS: A model of acute epilepsy in mice was established using the Pentylenetetrazol (PTZ) ignition method to evaluate the effects of Rhein on the duration and latency of convulsions, and the number and severity of seizures. Modified Neurological Severity Score (mNSS), Rotarod and open-field behavioral task tests were performed to evaluate the neuroprotective effect of Rhein. TUNEL staining was used to assess neuronal damage, and western blot, qPCR and ELISA kits were utilized to determine the expression of inflammatory signaling protein molecules and levels of inflammatory cytokines. RESULTS: In this study, we demonstrate that Rhein delayed the onset of seizures, decreased their severity, and reduced the duration and frequency of seizures in PTZ-induced epileptic mice. Furthermore, we found that Rhein blocked neurological deficits induced by PTZ. In addition, our results show that Rhein inhibited the activation of the TLR4-NFκB signaling pathway and decreased the secretion of the inflammatory cytokines TNF-α, IL-6, IL-1ß, and IL-18. CONCLUSION: Our results suggest that the anticonvulsant and neuroprotective effects of Rhein are achieved by disrupting the processes involved in PTZ acquisition of epilepsy.


Asunto(s)
Antraquinonas/farmacología , Epilepsia/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Animales , Antraquinonas/uso terapéutico , Citocinas/metabolismo , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Epilepsia/inducido químicamente , Epilepsia/diagnóstico , Epilepsia/inmunología , Humanos , Masculino , Ratones , FN-kappa B/metabolismo , Fármacos Neuroprotectores/uso terapéutico , Pentilenotetrazol/administración & dosificación , Pentilenotetrazol/toxicidad , Índice de Severidad de la Enfermedad , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Receptor Toll-Like 4
14.
Int J Med Sci ; 18(11): 2285-2293, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33967604

RESUMEN

This study examined the effect of the Flos Lonicerae Japonicae water extract (FLJWE), chlorogenic acid, and luteolin on pseudorabies virus (PRV)-induced inflammation in RAW264.7 cells and elucidated related molecular mechanisms. The results revealed that FLJWE and luteolin, but not chlorogenic acid, inhibited the production of inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), and inflammatory cytokines in PRV-infected RAW 264.7 cells. We found that the FLJWE and luteolin suppressed nuclear factor (NF)-κB activation by inhibiting the phosphorylation of signal transducer and activator of transcription 1 and 3 (STAT1 and STAT3, respectively). Moreover, the FLJWE significantly upregulated the expression of pNrf2 and its downstream target gene heme oxygenase-1 (HO-1). Our data indicated that FLJWE and luteolin reduced the expression of proinflammatory mediators and inflammatory cytokines, such as COX-2 and iNOS, through the suppression of the JAK/STAT1/3-dependent NF-κB pathway and the induction of HO-1 expression in PRV-infected RAW264.7 cells. The findings indicate that the FLJWE can be used as a potential antiviral agent.


Asunto(s)
Antiinflamatorios/farmacología , Antivirales/farmacología , Lonicera/química , Extractos Vegetales/farmacología , Virosis/tratamiento farmacológico , Animales , Antiinflamatorios/aislamiento & purificación , Antiinflamatorios/uso terapéutico , Antivirales/aislamiento & purificación , Antivirales/uso terapéutico , Modelos Animales de Enfermedad , Flores/química , Hemo-Oxigenasa 1/metabolismo , Herpesvirus Suido 1/inmunología , Humanos , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Inflamación/virología , Proteínas de la Membrana/metabolismo , Ratones , FN-kappa B/metabolismo , Extractos Vegetales/aislamiento & purificación , Extractos Vegetales/uso terapéutico , Células RAW 264.7 , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Virosis/inmunología , Virosis/virología , Agua/química
15.
Mol Cells ; 44(5): 335-341, 2021 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-33986184

RESUMEN

Zinc is an essential micronutrient with crucial roles in multiple facets of biological processes. Dysregulated zinc homeostasis impairs overall immune function and resultantly increases susceptibility to infection. Clinically, zinc supplementation is practiced for treatment of several infectious diseases, such as diarrhea and malaria. Recent focus on zinc as a beneficial element for immune system support has resulted in investigation of the immunomodulatory roles of zinc in a variety of immune cells. Besides its classical role as a cofactor that regulates the structural function of thousands of proteins, accumulating evidence suggests that zinc also acts, in a manner similar to calcium, as an ionic regulator of immune responses via participation as an intracellular messenger in signaling pathways. In this review, we focus on the role of zinc as a signaling molecule in major pathways such as those downstream of Toll-like receptors-, T cell receptor-, and cytokine-mediated signal transduction that regulate the activity and function of monocytes/macrophages and T cells, principal players in the innate and adaptive immune systems.


Asunto(s)
Transducción de Señal/inmunología , Zinc/uso terapéutico , Humanos , Zinc/farmacología
16.
Eur J Pharmacol ; 904: 174193, 2021 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-34015316

RESUMEN

Coronavirus (SARS-CoV-2) is spreading rapidly in the world and is still taking a heavy toll. Studies show that cytokine storms and imbalances in T-helper (Th)1/Th2 play a significant role in most acute cases of the disease. A number of medications have been suggested to treat or control the disease but have been discontinued due to their side effects. Melatonin, as an intrinsic molecule, possesses pharmacological anti-inflammatory and antioxidant properties that decreases in concentration with age; as a result, older people are more prone to various diseases. In this study, patients who were hospitalized with a diagnosis of coronavirus disease 2019 (COVID-19) were given a melatonin adjuvant (9 mg daily, orally) for fourteen days. In order to measure markers of Th1 and Th2 inflammatory cytokines (such as interleukin (IL)-2, IL-4, and interferon (IFN)-γ) as well as the expression of Th1 and Th2 regulatory genes (signal transducer and activator of transcription (STAT)4, STAT6, GATA binding protein 3 (GATA3), and T-box expressed in T cell (T-bet)), blood samples were taken from patients at the beginning and end of the treatment. Adjuvant therapy with melatonin controlled and reduced inflammatory cytokines in patients with COVID-19. Melatonin also controlled and modulated the dysregulated genes that regulate the humoral and cellular immune systems mediated by Th1 and Th2. In this study, it was shown for the first time that melatonin can be used as a medicinal adjuvant with anti-inflammatory mechanism to reduce and control inflammatory cytokines by regulating the expression of Th1 and Th2 regulatory genes in patients with COVID-19.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , COVID-19 , Citocinas/sangre , Melatonina , Transducción de Señal , Células TH1 , Células Th2 , Antiinflamatorios/administración & dosificación , Antiinflamatorios/inmunología , COVID-19/diagnóstico , COVID-19/epidemiología , COVID-19/inmunología , Femenino , Humanos , Inmunidad Celular/efectos de los fármacos , Inmunidad Humoral/efectos de los fármacos , Factores Inmunológicos/administración & dosificación , Factores Inmunológicos/inmunología , Irán/epidemiología , Masculino , Melatonina/administración & dosificación , Melatonina/inmunología , Persona de Mediana Edad , SARS-CoV-2 , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Células TH1/efectos de los fármacos , Células TH1/inmunología , Células Th2/efectos de los fármacos , Células Th2/inmunología , Resultado del Tratamiento
17.
Int Immunopharmacol ; 97: 107657, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33878544

RESUMEN

Osteoarthritis (OA) is a common joint disease that takes joint degeneration or aging as its pathological basis, and joint swelling, pain or dysfunction as its main clinical manifestations. Decursin (DE), the major active component isolated from Angelica gigas Nakai, has been demonstrated to possess anti-inflammatory effect in many diseases. But, the specific physiological mechanism of DE on OA is not clear yet. Therefore, the object of this study was to assess the therapeutic effect of DE on OA, and to explore its potential anti-inflammatory mechanisms. In vitro cell experiments, the inflammatory response in chondrocytes is mediated via interleukin-1ß (IL-1ß), which led to abnormal secretion of pro-inflammatory factors, such as prostaglandin E2 (PGE2), interleukin-6 (IL-6), tumor necrosis factor alpha (TNF-α), cyclooxygenase-2 (COX-2), nitric oxide (NO) and inducible nitric oxide synthase (iNOS). These cytokines were all decreased by the preconditioning of DE in a dose-dependent form of 1, 5, and 10 µM. Moreover, DE could restrain IL-1ß-mediated inflammatory reaction and the collapse of extracellular matrix (ECM) via reducing the secretion of ADAMTS (a disintegrin and metalloproteinase with thrombospondin motifs) and MMPs (matrix metalloproteinases). In short, DE restrained IL-1ß-mediated abnormal excitation of PI3K/AKT/NF-κB axis. Furthermore, molecular docking analysis showed that DE has a strong binding affinity with the inhibitory targets of PI3K. In vivo animal studies, DE treatment could helped to improve destruction of articular cartilage and decreased the serum inflammatory factor levels in an operationally induced mouse OA model. To sum up, these data obtained from the experiment indicate that DE has good prospects for the treatment of osteoarthritis.


Asunto(s)
Antiinflamatorios/farmacología , Benzopiranos/farmacología , Butiratos/farmacología , Osteoartritis/tratamiento farmacológico , Animales , Antiinflamatorios/uso terapéutico , Benzopiranos/uso terapéutico , Butiratos/uso terapéutico , Cartílago Articular/efectos de los fármacos , Cartílago Articular/inmunología , Cartílago Articular/patología , Progresión de la Enfermedad , Evaluación Preclínica de Medicamentos , Humanos , Interleucina-1beta/metabolismo , Masculino , Ratones , Simulación del Acoplamiento Molecular , FN-kappa B/metabolismo , Osteoartritis/inmunología , Osteoartritis/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología
18.
Immunity ; 54(6): 1219-1230.e7, 2021 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-33915109

RESUMEN

The sympathetic nervous system (SNS) controls various physiological functions via the neurotransmitter noradrenaline. Activation of the SNS in response to psychological or physical stress is frequently associated with weakened immunity. Here, we investigated how adrenoceptor signaling influences leukocyte behavior. Intravital two-photon imaging after injection of noradrenaline revealed transient inhibition of CD8+ and CD4+ T cell locomotion in tissues. Expression of ß-adrenergic receptor in hematopoietic cells was not required for NA-mediated inhibition of motility. Rather, chemogenetic activation of the SNS or treatment with adrenergic receptor agonists induced vasoconstriction and decreased local blood flow, resulting in abrupt hypoxia that triggered rapid calcium signaling in leukocytes and halted cell motility. Oxygen supplementation reversed these effects. Treatment with adrenergic receptor agonists impaired T cell responses induced in response to viral and parasitic infections, as well as anti-tumor responses. Thus, stimulation of the SNS impairs leukocyte mobility, providing a mechanistic understanding of the link between adrenergic receptors and compromised immunity.


Asunto(s)
Adrenérgicos/inmunología , Movimiento Celular/inmunología , Inmunidad/inmunología , Leucocitos/inmunología , Sistema Nervioso Simpático/inmunología , Animales , Señalización del Calcio/inmunología , Línea Celular Tumoral , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Receptores Adrenérgicos/inmunología , Transducción de Señal/inmunología , Linfocitos T/inmunología
19.
Molecules ; 26(7)2021 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-33806085

RESUMEN

Cicadae Periostracum (CP), derived from the slough of Cryptotympana pustulata, has been used as traditional medicine in Korea and China because of its diaphoretic, antipyretic, anti-inflammatory, antioxidant, and antianaphylactic activities. The major bioactive compounds include oleic acid (OA), palmitic acid, and linoleic acid. However, the precise therapeutic mechanisms underlying its action in asthma remain unclear. The objective of this study was to determine the antiasthmatic effects of CP in an ovalbumin (OVA)-induced asthmatic mouse model. CP and OA inhibited the inflammatory cell infiltration, airway hyperresponsiveness (AHR), and production of interleukin (IL)7 and Th2 cytokines (IL-5) in the bronchoalveolar lavage fluid and OVA-specific imunoglobin E (IgE) in the serum. The gene expression of IL-5, IL-13, CCR3, MUC5AC, and COX-2 was attenuated in lung tissues. CP and OA might inhibit the nuclear translocation of GATA-binding protein 3 (GATA-3) and retinoic acid receptor-related orphan receptor γt (RORγt) via the upregulation of forkhead box p3 (Foxp3), thereby preventing the activation of GATA-3 and RORγt. In the in vitro experiment, a similar result was observed for Th2 and GATA-3. These results suggest that CP has the potential for the treatment of asthma via the inhibition of the GATA-3/Th2 and IL-17/RORγt signaling pathways.


Asunto(s)
Asma , Mezclas Complejas , Factor de Transcripción GATA3/inmunología , Hemípteros/química , Interleucina-17/inmunología , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/inmunología , Ácido Oléico , Transducción de Señal , Células Th2/inmunología , Animales , Asma/inducido químicamente , Asma/tratamiento farmacológico , Asma/inmunología , Asma/patología , Mezclas Complejas/química , Mezclas Complejas/farmacología , Masculino , Ratones , Ratones Endogámicos BALB C , Ácido Oléico/química , Ácido Oléico/farmacología , Ovalbúmina/toxicidad , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Células Th2/patología
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA