Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Chin J Nat Med ; 19(6): 432-441, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-34092294

RESUMEN

Esculetin, a natural derivative from the traditional and widely-used Chinese medicinal herb Cortex Fraxini, has a variety of pharmacological effects, especially in anti-inflammation. However, it is not clear whether esculetin has a therapeutic effect on sepsis. This study aimed to investigate the anti-inflammatory and protective effects of esculetin on early sepsis. The results showed that the lung injury was significantly relieved with the treatment of esculetin, accompanied with the restrained production of inflammatory factors including IL-1ß, IL-6, TNF-α, CCL2 and iNOS during the early phase of E.coli-induced sepsis. Of note, activation of NF-κB and STAT1/STAT3 signals, the main upstream signals of many inflammatory factors, were attenuated by esculetin in both lung tissues from septic mice and LPS-stimulated macrophage. These findings suggested that the protection of esculetin against early sepsis should be related to its anti-inflammatory effect, which was at least partly due to its inhibition on NF-κB and STAT1/STAT3 signaling pathway in macrophage. Thus, esculetin could serve as a potential therapeutic agent by rebalancing innate immune response in macrophage for the treatment of early sepsis.


Asunto(s)
FN-kappa B , Sepsis , Transducción de Señal/efectos de los fármacos , Umbeliferonas/farmacología , Animales , Inflamación/tratamiento farmacológico , Lipopolisacáridos , Ratones , FN-kappa B/antagonistas & inhibidores , Factor de Transcripción STAT1/antagonistas & inhibidores , Factor de Transcripción STAT3/antagonistas & inhibidores , Sepsis/tratamiento farmacológico
2.
Int J Mol Sci ; 21(14)2020 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-32708322

RESUMEN

Some coronavirus disease 2019 (COVID-19) patients develop acute pneumonia which can result in a cytokine storm syndrome in response to Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2) infection. The most effective anti-inflammatory drugs employed so far in severe COVID-19 belong to the cytokine-directed biological agents, widely used in the management of many autoimmune diseases. In this paper we analyze the efficacy of epigallocatechin 3-gallate (EGCG), the most abundant ingredient in green tea leaves and a well-known antioxidant, in counteracting autoimmune diseases, which are dominated by a massive cytokines production. Indeed, many studies registered that EGCG inhibits signal transducer and activator of transcription (STAT)1/3 and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) transcription factors, whose activities are crucial in a multiplicity of downstream pro-inflammatory signaling pathways. Importantly, the safety of EGCG/green tea extract supplementation is well documented in many clinical trials, as discussed in this review. Since EGCG can restore the natural immunological homeostasis in many different autoimmune diseases, we propose here a supplementation therapy with EGCG in COVID-19 patients. Besides some antiviral and anti-sepsis actions, the major EGCG benefits lie in its anti-fibrotic effect and in the ability to simultaneously downregulate expression and signaling of many inflammatory mediators. In conclusion, EGCG can be considered a potential safe natural supplement to counteract hyper-inflammation growing in COVID-19.


Asunto(s)
Antiinflamatorios/uso terapéutico , Catequina/análogos & derivados , Infecciones por Coronavirus/tratamiento farmacológico , Síndrome de Liberación de Citoquinas/tratamiento farmacológico , Neumonía Viral/tratamiento farmacológico , Antioxidantes/uso terapéutico , Enfermedades Autoinmunes/tratamiento farmacológico , Betacoronavirus/efectos de los fármacos , Betacoronavirus/inmunología , COVID-19 , Catequina/uso terapéutico , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/patología , Síndrome de Liberación de Citoquinas/patología , Humanos , FN-kappa B/antagonistas & inhibidores , Pandemias , Extractos Vegetales/uso terapéutico , Neumonía Viral/inmunología , Neumonía Viral/patología , Fibrosis Pulmonar/tratamiento farmacológico , Fibrosis Pulmonar/prevención & control , SARS-CoV-2 , Factor de Transcripción STAT1/antagonistas & inhibidores , Factor de Transcripción STAT3/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos
3.
J Ethnopharmacol ; 243: 112121, 2019 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-31356966

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Psoriasis is an immune system meditated disease, especially T cells. It disturbed many people around the world and hard to therapy. Paeonia lactiflora Pall has been used as a medicine in china for thousands of years. Recent studies found that the main component of Paeonia lactiflora Pall can alleviates the immune response in many diseases. In this study, we researched the effects and possible mechanisms of total glucosides of paeony (TGP) on animal psoriasis. AIM OF THE STUDY: To study the therapeutic effects and mechanisms of TGP in 5% propranolol cream-induced psoriasis in guinea pigs and Imiquimod (IMQ) cream-induced psoriasis in mice. MATERIALS AND METHODS: The effect of TGP was evaluated using a psoriasis-like model of guinea pigs and mice. Ear thickness was accessed, and pathology injury was observed by H&E staining. The levels of serum IL-1ß, IL-6, IL-12, IL-17, IL-23, TNF-α, and IFN-γ, skin IL-17A, IL-22 and orphan nuclear receptor (RORγt) mRNA expression, proliferating cell nuclear antigen (PCNA), total or phosphorylated signal transducers and activators of transcription (STAT1, STAT3) were determined by enzyme linked immunosorbent assays (ELISAs), real time PCR, immunohistochemical staining, and western blotting, respectively. RESULTS: Compared with model group, TGP treatment decreased the ear thickness, improved pathology of psoriasis, alleviated IMQ-induced keratinocyte proliferation, reduced the inflammatory cytokine, and downregulated IL-17A, IL-22, and RORγt mRNA in mice. Further study indicated that TGP inhibited STAT1 and STAT3 phosphorylation in lesion skins of psoriasis-like mice. CONCLUSIONS: TGP alleviates the symptoms of psoriasis-like guinea pigs and mice, and the possible mechanism may relate to inhibit T helper 17 (TH17) cell differentiation and keratinocytes proliferation by inhibiting STAT1 and STAT3 phosphorylation.


Asunto(s)
Glucósidos/uso terapéutico , Paeonia , Psoriasis/tratamiento farmacológico , Factor de Transcripción STAT1/antagonistas & inhibidores , Factor de Transcripción STAT3/antagonistas & inhibidores , Animales , Citocinas/sangre , Citocinas/genética , Modelos Animales de Enfermedad , Femenino , Glucósidos/farmacología , Cobayas , Imiquimod , Masculino , Ratones Endogámicos BALB C , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Fosforilación/efectos de los fármacos , Raíces de Plantas , Psoriasis/sangre , Psoriasis/inducido químicamente , Psoriasis/metabolismo , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT3/metabolismo
4.
Inflammopharmacology ; 27(6): 1255-1263, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30783895

RESUMEN

Influenza viruses can bring about acute respiratory diseases and are a potential hazard to human health. Antiviral drugs are the main ways to control the influenza virus infection except the vaccine. In this study, the immune regulation activity of pterodontic acid isolated from Laggera pterodonta induced by influenza A virus in vitro was evaluated. In studies on anti-influenza activity, our results showed that it maybe target the influenza protein of polymerase basic 1 (PB1), polymerase basic 2 (PB2), polymerase acid (PA), nuclear protein (NP), non-structural protein (NS), and matrix protein (M) but not hemagglutinin (HA) and neuraminidase (NA). In studies on immune regulation, our results demonstrated that pterodontic acid can inhibit the Retinoic acid inducible gene-I (RIG-I) expression in mRNA and protein level at 100 µg/ml, then further to clarify its action on the signalling pathway, The results indicated that pterodontic acid can inhibit the Tumor Necrosis Factor-related Apoptosis-inducing Ligand/Fas Ligand (TRAIL/Fasl) expression in mRNA level at 100 µg/ml; the cleaved caspase 3/7, p-NF-KB, and p-ERK were all suppressed in protein level by pterodontic acid at 100 µg/ml. This confirmed its mechanism that restrained the nuclear export of viral RNPs. The interferon system was also affected, the STAT1, IFN-α, IFN-ß expression were also inhibited by pterodontic acid at 25-100 µg/ml and also, the important programmed death-ligand of PD-L1 and PD-L2 was inhibited at 50-100 µg/ml. The mechanisms of pterodontic acid against influenza virus infection may be a cascade inhibition and it has the anti-inflammatory activity, which has no side effect, and can be as a supplement drug in clinical influenza virus infection.


Asunto(s)
Antivirales/farmacología , Asteraceae/química , Antígeno B7-H1/fisiología , Proteína 58 DEAD Box/antagonistas & inhibidores , Virus de la Influenza A/efectos de los fármacos , Interferón Tipo I/antagonistas & inhibidores , FN-kappa B/antagonistas & inhibidores , Proteína 2 Ligando de Muerte Celular Programada 1/antagonistas & inhibidores , Sesquiterpenos/farmacología , Células A549 , Antígeno B7-H1/antagonistas & inhibidores , Humanos , Virus de la Influenza A/fisiología , Proteína 2 Ligando de Muerte Celular Programada 1/fisiología , Receptores Inmunológicos , Ribonucleoproteínas/metabolismo , Factor de Transcripción STAT1/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Ligando Inductor de Apoptosis Relacionado con TNF/antagonistas & inhibidores
5.
J Pharm Pharmacol ; 71(1): 93-103, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28990659

RESUMEN

OBJECTIVES: St John's wort extract (SJW) and its component hyperforin (HPF) were shown to potently inhibit cytokine-induced STAT-1 and NF-κB activation in pancreatic ß cells and protect them against injury. This study aimed at exploring the time course of STAT-1 inhibition afforded by these natural compounds in the ß-cell line INS-1E. METHODS: INS-1E cells were pre-incubated with SJW extract (2-5 µg/ml) or HPF (0.5-2 µm) and then exposed to a cytokine mixture. In some experiments, these compounds were added after or removed before cytokine exposure. STAT-1 activation was assessed by electrophoretic mobility shift assay, apoptosis by caspase-3 activity assay, mRNA gene expression by RT-qPCR. KEY FINDINGS: Pre-incubation with SJW/HPF for 1-2 h exerted a remarkable STAT-1 downregulation, which was maintained upon removal of the compounds before early or delayed cytokine addition. When the protective compounds were added after cell exposure to cytokines, between 15 and 90 min, STAT-1 inhibition also occurred at a progressively decreasing extent. Upon 24-h incubation, SJW and HPF counteracted cytokine-induced ß-cell dysfunction, apoptosis and target gene expression. CONCLUSIONS: SJW and HPF confer to ß cells a state of 'cytokine resistance', which can be elicited both before and after cytokine exposure and safeguards these cells from deleterious cytokine effects.


Asunto(s)
Hypericum/química , Células Secretoras de Insulina/efectos de los fármacos , Floroglucinol/análogos & derivados , Extractos Vegetales/farmacología , Terpenos/farmacología , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Citocinas/metabolismo , Relación Dosis-Respuesta a Droga , Ensayo de Cambio de Movilidad Electroforética , Regulación de la Expresión Génica/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Floroglucinol/administración & dosificación , Floroglucinol/aislamiento & purificación , Floroglucinol/farmacología , Extractos Vegetales/administración & dosificación , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT1/antagonistas & inhibidores , Factor de Transcripción STAT1/metabolismo , Terpenos/administración & dosificación , Terpenos/aislamiento & purificación , Factores de Tiempo
6.
Int J Mol Sci ; 19(5)2018 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-29786649

RESUMEN

Kudzu (Pueraria montana var. lobata (Willd.) Sanjappa & Pradeep) is a perennial leguminous vine, and its root and flower have been used for herbal medicine in Asia for a long time. Most dietary flavonoids are reported to be concentrated in its root, not in its aerial parts including leaves. In this study, we investigated whether kudzu leaf and its major constituent, robinin (kaempferol-3-O-robinoside-7-O-rhanmoside) possessed anti-inflammatory activity. To test this hypothesis, we used peritoneal macrophages isolated from BALB/c mice and stimulated the cells with lipopolysaccharide (LPS) or LPS plus interferon (IFN)-γ. Compared with kudzu root extract, its leaf extract was more potent in inhibiting the production of inducible nitric oxide synthase (iNOS), cyclooxygenase-2, tumor necrosis factor-α, and interleukin-6. Kudzu leaf extract decreased LPS-induced activation of c-Jun N-terminal kinase (JNK) and TANK-binding kinase 1(TBK1) with no effects on nuclear factor-κB and activator protein 1 transcriptional activity. Also, kudzu leaf extract inhibited LPS/IFN-γ-induced signal transducer and activator of transcription 1 (STAT1) activation partly via an altered level of STAT1 expression. Robinin, being present in 0.46% of dry weight of leaf extract, but almost undetected in the root, decreased iNOS protein involving modulation of JNK and STAT1 activation. However, robinin showed no impact on other inflammatory markers. Our data provide evidence that kudzu leaf is an excellent food source of as yet unknown anti-inflammatory constituents.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Macrófagos/efectos de los fármacos , Extractos Vegetales/farmacología , Pueraria/química , Animales , Células Cultivadas , Ciclooxigenasa 2/metabolismo , Interleucina-6/metabolismo , MAP Quinasa Quinasa 4/antagonistas & inhibidores , MAP Quinasa Quinasa 4/metabolismo , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Óxido Nítrico Sintasa de Tipo II/metabolismo , Hojas de la Planta/química , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Factor de Transcripción STAT1/antagonistas & inhibidores , Factor de Transcripción STAT1/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
7.
Mol Med Rep ; 17(2): 2515-2522, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29207093

RESUMEN

Inflammatory skin disease are caused by multiple factors, including susceptibility genes, and immunologic and environmental factors, and are characterized by an increase in epidermal thickness and the infiltration of macrophages, keratinocytes, mast cells, eosinophils and other inflammatory cells. Keratinocytes may serve an important role in the pathogenesis of inflammatory skin diseases. The traditional herbal decoction Hyangso­san (HSS) has been used to treat symptoms of the common cold, including headache, pantalgia, fever and chills. However, to the best of our knowledge, there is no evidence regarding whether HSS has an effect on inflammatory skin diseases. The present study investigated the anti­skin inflammation activity of HSS using the HaCaT human keratinocyte cell line. The mRNA expression and production of inflammatory chemokines, including C­C motif chemokine ligand 22 (CCL22), CCL5, CCL17, and interleukin (IL)­8, was measured using reverse transcription polymerase chain reaction and ELISA analyses. Moreover, we evaluated the effect of HSS on signal transducer and activator of transcription 1 (STAT1) pathway in HaCaT cells. The cells were stimulated with tumor necrosis factor­α (TNF­α) and interferon­Î³ (IFN­Î³) to induce an inflammatory reaction. In the TNF­α­ and IFN­Î³­stimulated cells, the production and expression of inflammatory chemokines were observed, including CCL22, CCL5, CCL17 and IL­8. In addition, stimulation with TNF­α and IFN­Î³ increased the phosphorylation and nuclear translocation of STAT1 in HaCaT cells. By contrast, HSS extract treatment inhibited TNF­α­ and IFN­Î³­induced STAT1 activation. Results from the present study indicated that HSS exhibited inhibitory effects on TNF­α­ and IFN­Î³­mediated chemokine production and expression by targeting STAT1 in keratinocytes. Overall, the results indicated that HSS may be a potential candidate therapeutic drug for inflammatory skin diseases such as atopic dermatitis.


Asunto(s)
Antiinflamatorios/farmacología , Quimiocinas/biosíntesis , Dermatitis/metabolismo , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Extractos Vegetales/farmacología , Factor de Transcripción STAT1/antagonistas & inhibidores , Línea Celular , Supervivencia Celular/efectos de los fármacos , Quimiocinas/genética , Cromatografía Líquida de Alta Presión , Citocinas/biosíntesis , Citocinas/genética , Dermatitis/tratamiento farmacológico , Dermatitis/etiología , Dermatitis/patología , Relación Dosis-Respuesta a Droga , Expresión Génica , Humanos , Extractos Vegetales/química
8.
Cell Physiol Biochem ; 44(2): 607-617, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29161701

RESUMEN

BACKGROUND/AIMS: Type I interferon (IFN-1) production and IFN-1 signaling play critical roles in the host antiviral innate immune responses. Although transcription factor Yin Yang 1 (YY1) has been reported to have a dual activator/repressor role during the regulation of interferon beta (IFN-ß) promoter activity, the roles of YY1 in the regulation of upstream signaling pathways leading to IFN-1 induction and IFN-1 signaling during viral infection remain to be elucidated. METHODS: The roles of YY1 in IFN-1 production and IFN-1 signaling were investigated using immunoblotting, real-time PCR, small interfering RNA (siRNA)-mediated YY1 knockdown, YY1 overexpression by transient transfection, and co-immunoprecipitation, using mouse cells. RESULTS: YY1 was shown to interact with STAT1 in the absence of viral infection. Following viral infection, YY1 protein expression levels were decreased. YY1 knockdown led to a considerable downregulation of phosphorylated (p) TBK1 and pIRF3 expressions, while YY1 overexpression significantly upregulated pTBK1 and pIRF3 expression levels and promoted virus-induced IFN-ß production. Additionally, YY1 knockdown led to a significant upregulation of pSTAT1, pSTAT2 and antiviral interferon-stimulated genes, and inhibited viral replication. CONCLUSION: We demonstrated here that YY1 interacts with STAT1 and dynamically regulates the induction of IFN-1 production and activation of IFN-1 signaling in different stages during viral infection.


Asunto(s)
Inmunidad Innata , Factor de Transcripción YY1/metabolismo , Animales , Línea Celular , Quimiocina CXCL10/genética , Quimiocina CXCL10/metabolismo , Regulación hacia Abajo , Ensayo de Inmunoadsorción Enzimática , Inmunoprecipitación , Factor 3 Regulador del Interferón/metabolismo , Interferón beta/análisis , Interferón beta/metabolismo , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas de Resistencia a Mixovirus/genética , Proteínas de Resistencia a Mixovirus/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor de Transcripción STAT1/antagonistas & inhibidores , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo , Transducción de Señal , Simplexvirus/fisiología , Transfección , Regulación hacia Arriba , Vesiculovirus/fisiología , Factor de Transcripción YY1/antagonistas & inhibidores , Factor de Transcripción YY1/genética
9.
Skin Pharmacol Physiol ; 30(5): 268-276, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28873377

RESUMEN

BACKGROUND AND OBJECTIVES: The objective of this study was to evaluate the topical effects of sea buckthorn (SBT) oil on atopic dermatitis (AD)-like lesions in a mouse model generated by repeated topical administration of DNCB in BALB/c mice. METHODS: DNCB was applied repeatedly on the dorsal skin of mice to induce AD-like lesions. Following AD induction, SBT oil was applied daily on the dorsal skin for 4 weeks. The severity of skin lesions was examined macroscopically and histologically. We further measured the production of MDC/CCL22 and TARC/CCL17 in IFN-γ/TNF-α activated HaCaT cells. RESULTS: Topically applied SBT oil in DNCB-treated mice ameliorated the severity score of dermatitis, decreased epidermal thickness, reduced spleen and lymph node weights, and prevented mast cell infiltration. In addition, SBT oil suppressed the Th2 chemokines TARC and MDC via dose-dependent inhibition of NF-κB, JAK2/STAT1, and p38-MAPK signaling pathways in IFN-γ/TNF-α-activated HaCaT cells. CONCLUSION: These results suggest that SBT oil had a beneficial effect on AD-like skin lesions, partially via inhibition of the Th2 chemokines TARC and MDC in inflamed skin.


Asunto(s)
Dermatitis Atópica/tratamiento farmacológico , Hippophae , Aceites de Plantas/uso terapéutico , Animales , Línea Celular , Quimiocina CCL17/metabolismo , Quimiocina CCL22/metabolismo , Dermatitis Atópica/metabolismo , Dermatitis Atópica/patología , Dinitroclorobenceno , Femenino , Humanos , Irritantes , Ganglios Linfáticos/efectos de los fármacos , Ratones Endogámicos BALB C , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Aceites de Plantas/farmacología , Factor de Transcripción STAT1/antagonistas & inhibidores , Factor de Transcripción STAT1/metabolismo , Piel/efectos de los fármacos , Piel/metabolismo , Piel/patología , Bazo/efectos de los fármacos
10.
Daru ; 25(1): 18, 2017 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-28778215

RESUMEN

BACKGROUND: Regulation of a persistently-activated inflammatory response in macrophages is an important target for treatment of various chronic diseases. Pine needle extracts are well known to have potent immunomodulatory effects. The current study was designed to evaluate the effects of Pinus densiflora needle supercritical fluid extract (PDN-SCFE) on bacterial lipopolysaccharide (LPS)-induced inflammatory response in RAW 264.7 murine macrophages. METHODS: Cytotoxic effect of PDN-SCFE was determined by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. The levels of nitric oxide (NO) and the corresponding enzyme, inducible nitric oxide synthase (iNOS), were quantified by Griess and immunoblotting methods, respectively. The levels of cytokines were quantified using commercial ELISA kits. Quantitative real-time PCR (qRT-PCR) analysis was performed to assess the mRNA expression of iNOS and cytokines. To elucidate the mechanism of action, the involvement of nuclear transcription factor-kappa B (NFκB), mitogen activated protein kinases (MAPKs) and Janus kinase-signal transducers and activators of transcription (JAK-STAT) pathways were examined by an immunoblotting method. In addition, the cellular localization of NFκB was analyzed by immunofluorescence staining. RESULTS: MTT assay results indicated that PDN-SCFE is non-toxic to RAW 264.7 cells up to a maximum assayed concentration of 40 µg/mL. The PDN-SCFE exhibited a concentration-dependent inhibitory effect on LPS-induced NO production by down regulating the expression of iNOS. In addition, the extract suppressed the LPS-induced expression of interleukin-6 (IL-6) and interleukin-1ß (IL-1ß) but not tumour necrosis factor-α (TNFα). Mechanistic studies revealed that PDN-SCFE does not influence the NFκB and MAPK pathways. However, it showed a significant inhibitory effect on LPS-induced activation of STAT1 and STAT3 proteins in macrophages. CONCLUSION: The present findings revealed that the anti-inflammatory activity of PDN-SCFE in LPS-challenged RAW 264.7 macrophages is probably caused by the suppression of the JAK-STAT signaling pathway.


Asunto(s)
Interleucina-18/antagonistas & inhibidores , Interleucina-6/antagonistas & inhibidores , Macrófagos/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Pinus/química , Extractos Vegetales/farmacología , Hojas de la Planta/química , Factor de Transcripción STAT1/antagonistas & inhibidores , Factor de Transcripción STAT3/antagonistas & inhibidores , Animales , Western Blotting , Inflamación/tratamiento farmacológico , Lipopolisacáridos/farmacología , Ratones , Microscopía Fluorescente , Células RAW 264.7 , Reacción en Cadena en Tiempo Real de la Polimerasa
11.
J Agric Food Chem ; 64(47): 9012-9021, 2016 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-27933873

RESUMEN

Isogarcinol is a new natural immunosuppressant that was extracted from Garcinia mangostana L. in our laboratory. Knowledge of its effects on treatable diseases and its mechanism of action is still very limited. In this study, we explored the therapeutic effect of isogarcinol in experimental autoimmune encephalomyelitis (EAE), a murine model of multiple sclerosis (MS). Treatment with oral 100 mg/kg isogarcinol markedly ameliorated clinical scores, alleviated inflammation and demyelination of the spinal cord, and reduced intracranial lesions in EAE mice. The percentages of Th cells and macrophages were also strongly reduced. Isogarcinol appeared to act by inhibiting T helper (Th) 1 and Th17 cell differentiation via the janus kinase/signal transducers and activators of transcription pathway and by impairing macrophage function. Our data suggest that isogarcinol has the potential to be an effective therapeutic agent of low toxicity for treating MS and other autoimmune diseases.


Asunto(s)
Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Garcinia mangostana/química , Inmunosupresores/farmacología , Extractos Vegetales/farmacología , Terpenos/farmacología , Animales , Antiinflamatorios/farmacología , Diferenciación Celular/efectos de los fármacos , Femenino , Inflamación/tratamiento farmacológico , Quinasas Janus/metabolismo , Macrófagos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Factor de Transcripción STAT1/antagonistas & inhibidores , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT3/antagonistas & inhibidores , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo , Células Th17
12.
Chem Biol Interact ; 256: 102-10, 2016 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-27378624

RESUMEN

Urgent needs still exist for selective control of excessive inflammation. Despite the therapeutic potential of natural compounds against inflammation-associated chronic conditions, lack of specific molecular targets renders these bioactive compounds difficult for further development. Here we examined the bioactivity of coniferyl aldehyde (CA), a natural phenolic compound found in several dietary substances and medicinal plants, elucidating its efficacy both in vivo and in vitro with underlying molecular mechanisms. IFN-γ/TNF-α-stimulated human keratinocytes and lipopolysaccharide (LPS)-stimulated murine macrophages were used to examine the effect of CA in vitro and to elucidate the underlying mechanisms. In vivo models of phorbol 12-myristate 13-acetate (TPA)-induced ear edema and carrageenan (CRG)-induced paw edema were employed to investigate the topical and systemic anti-inflammatory effects of CA, respectively. CA significantly reduced nitric oxide (NO) production and inducible nitric oxide synthase (iNOS) expression in LPS-stimulated macrophages. While nuclear factor-κB (NF-κB) and mitogen-activated protein kinase (MAPKs) pathways, the representative cellular pathways for iNOS induction, were not affected by CA, phosphorylation of Janus kinase 2 (JAK2) and signal Transducers and Activators of Transcription 1 (STAT1) and subsequent nuclear translocation of p-STAT1 were significantly decreased by CA. The effect of CA on JAK2-STAT1-iNOS axis was also observed in human keratinocytes stimulated with IFN-γ/TNF-α. Topical application of CA to mice produced significant protection against TPA-induced ear edema along with suppressed epidermal hyperproliferation and leucocyte infiltration. Systemic administration of CA significantly reduced CRG-induced paw edema in rats, where CRG-induced iNOS expression and STAT1 phosphorylation were decreased by CA. In summary, CA has significant anti-inflammatory properties both in vitro and in vivo, mediated by significant selective inhibition of JAK2-STAT1-iNOS signaling. CA is an attractive novel candidate for treating inflammatory diseases associated with excessive production of NO.


Asunto(s)
Acroleína/análogos & derivados , Antiinflamatorios/uso terapéutico , Regulación hacia Abajo/efectos de los fármacos , Edema/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Janus Quinasa 2/inmunología , Óxido Nítrico Sintasa de Tipo II/genética , Factor de Transcripción STAT1/inmunología , Acroleína/farmacología , Acroleína/uso terapéutico , Animales , Antiinflamatorios/farmacología , Carragenina , Línea Celular , Oído/patología , Edema/inducido químicamente , Edema/genética , Edema/inmunología , Humanos , Inflamación/inducido químicamente , Inflamación/genética , Inflamación/inmunología , Janus Quinasa 2/antagonistas & inhibidores , Lipopolisacáridos/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos ICR , Óxido Nítrico/inmunología , Óxido Nítrico Sintasa de Tipo II/inmunología , Células RAW 264.7 , Ratas Sprague-Dawley , Factor de Transcripción STAT1/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Acetato de Tetradecanoilforbol/análogos & derivados
13.
PLoS One ; 9(12): e115146, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25517939

RESUMEN

The flower buds of Daphne genkwa Sieb. et Zucc. have been used as a traditional Chinese medicine although their functional mechanisms have not been discovered yet. We have studied the potential effects of the plant extracts on natural killer (NK) cell activation, and isolated an active fraction. Genkwadaphnin (GD-1) displayed a potent efficacy to induce IFN-γ transcription in NK cells with concentration- and time-dependent manners. GD-1 treatment triggered the phosphorylation of PKD1, a member of PKC family, MEK and ERK, resulting in IKK activation to induce IκB degradation, and the nuclear localization of p65, an NF-κB subunit, which regulates IFN-γ transcription. GD-1 effect on IFN-γ production was blocked by the addition of Rottlerin, a PKC inhibitor, CID 755673, a PKD inhibitor, or Bay11-7082, an IKKα inhibitor. The nuclear localization of p65 was also inhibited by the kinase inhibitors. Secreted IFN-γ activates STAT1 phosphorylation as autocrine-loops to sustain its secretion. GD-1 induced the phosphorylation of STAT1 probably through the increase of IFN-γ. STAT1 inhibitor also abrogated the sustained IFN-γ secretion. These results suggest that GD-1 is involved in the activation of PKD1 and/or ERK pathway, which activate NK-κB triggering IFN-γ production. As positive feedback loops, secreted IFN-γ activates STAT1 and elongates its production in NK-92 cells.


Asunto(s)
Diterpenos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Interferón gamma/genética , Células Asesinas Naturales/efectos de los fármacos , Linfoma/tratamiento farmacológico , FN-kappa B/metabolismo , Factor de Transcripción STAT1/metabolismo , Canales Catiónicos TRPP/metabolismo , Apoptosis/efectos de los fármacos , Western Blotting , Proliferación Celular/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Humanos , Interferón gamma/metabolismo , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/patología , Linfoma/metabolismo , Linfoma/patología , FN-kappa B/antagonistas & inhibidores , FN-kappa B/genética , Fosforilación/efectos de los fármacos , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT1/antagonistas & inhibidores , Factor de Transcripción STAT1/genética , Canales Catiónicos TRPP/antagonistas & inhibidores , Canales Catiónicos TRPP/genética , Células Tumorales Cultivadas
14.
Arterioscler Thromb Vasc Biol ; 34(9): 1953-60, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25012131

RESUMEN

OBJECTIVE: Activation of Janus kinase/signal transducers and activators of transcription (STAT) pathway by hyperglycemia and dislypidemia contributes to the progression of diabetic complications, including atherosclerosis. Suppressor of cytokine signaling (SOCS) proteins negatively regulate Janus kinase/STAT and have emerged as promising target for anti-inflammatory therapies. We investigated whether a cell-permeable lipopeptide corresponding to the kinase inhibitory region of SOCS1 could reduce atherosclerosis in diabetic mice and identified the mechanisms involved. APPROACH AND RESULTS: Streptozotocin-induced diabetic apolipoprotein E-deficient mice (aged 8 and 22 weeks) were given intraperitoneal injections of vehicle, SOCS1-derived peptide, or control mutant peptide for 6 to 10 weeks. SOCS1 therapy suppressed STAT1/STAT3 activation in atherosclerotic plaques of diabetic mice and significantly reduced lesion size at both early and advanced stages of lesion development compared with vehicle group. Plaque characterization demonstrated that SOCS1 peptide decreased the accumulation of lipids, macrophages, and T lymphocytes, whereas increasing collagen and smooth muscle cell content. This atheroprotective effect was accompanied by systemic (reduced proinflammatory Ly6C(high) monocytes and splenic cytokine expression) and local (reduced aortic expression of chemokines and cytokines) mechanisms, without impact on metabolic parameters. In vitro, SOCS1 peptide dose dependently inhibited STAT1/STAT3 activation and target gene expression in vascular smooth muscle cells and macrophages and also suppressed cytokine-induced cell migration and adhesion processes. CONCLUSIONS: SOCS1-based targeting Janus kinase/STAT restrains key mechanisms of atherogenesis in diabetic mice, thereby preventing plaque formation and increasing plaque stability. Approaches to mimic native SOCS1 functions may have a therapeutic potential to retard the progression of diabetic complications.


Asunto(s)
Diabetes Mellitus Experimental/complicaciones , Inflamación/tratamiento farmacológico , Quinasas Janus/antagonistas & inhibidores , Placa Aterosclerótica/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/uso terapéutico , Factor de Transcripción STAT1/antagonistas & inhibidores , Factor de Transcripción STAT3/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Proteínas Supresoras de la Señalización de Citocinas/uso terapéutico , Secuencia de Aminoácidos , Animales , Línea Celular , Dicroismo Circular , Diabetes Mellitus Experimental/metabolismo , Progresión de la Enfermedad , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Activación Enzimática/efectos de los fármacos , Inflamación/enzimología , Inflamación/etiología , Interferón gamma/farmacología , Interleucina-6/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Datos de Secuencia Molecular , Terapia Molecular Dirigida , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Fragmentos de Péptidos/química , Fragmentos de Péptidos/farmacología , Fragmentos de Péptidos/uso terapéutico , Placa Aterosclerótica/enzimología , Placa Aterosclerótica/etiología , Conformación Proteica , Inhibidores de Proteínas Quinasas/farmacocinética , Inhibidores de Proteínas Quinasas/farmacología , Factor de Transcripción STAT1/fisiología , Factor de Transcripción STAT3/fisiología , Proteína 1 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/química , Proteínas Supresoras de la Señalización de Citocinas/farmacocinética , Proteínas Supresoras de la Señalización de Citocinas/farmacología
15.
Cell Signal ; 26(3): 619-28, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24333668

RESUMEN

New negative regulators of interferon (IFN) signaling, preferably with tissue specificity, are needed to develop therapeutic means to enhance the efficacy of type I IFNs (IFN-α/ß) and reduce their side effects. We conducted cell-based screening for IFN signaling enhancer and discovered that luteolin, a natural flavonoid, sensitized the antiproliferative effect of IFN-α in hepatoma HepG2 cells and cervical carcinoma HeLa cells. Luteolin promoted IFN-ß-induced Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway activation by enhancing the phosphorylation of Jak1, Tyk2, and STAT1/2, thereby promoting STAT1 accumulation in the nucleus and endogenous IFN-α-regulated gene expression. Of interest, inhibition of phosphodiesterase (PDE) abolished the effect of IFN-ß and luteolin on STAT1 phosphorylation. Luteolin also increased the cAMP-degrading activity of PDE bound with type I interferon receptor 2 (IFNAR2) and decreased the intracellular cAMP level, indicating that luteolin may act on the JAK/STAT pathway via PDE. Protein kinase A (PKA) was found to negatively regulate IFN-ß-induced JAK/STAT signaling, and its inhibitory effect was counteracted by luteolin. Pull-down and immunoprecipitation assays revealed that type II PKA interacted with IFNAR2 via the receptor for activated C-kinase 1 (RACK-1), and such interaction was inhibited by luteolin. Src homology domain 2 containing tyrosine phosphatase-2 (SHP-2) was further found to mediate the inhibitory effect of PKA on the JAK/STAT pathway. These data suggest that PKA/PDE-mediated cAMP signaling, integrated by RACK-1 to IFNAR2, may negatively regulate IFN signaling through SHP-2. Inhibition of this signaling may provide a new way to sensitize the efficacy of IFN-α/ß.


Asunto(s)
Interferón-alfa/farmacología , Interferón beta/farmacología , Janus Quinasa 1/metabolismo , Luteolina/farmacología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/antagonistas & inhibidores , Adyuvantes Inmunológicos/farmacología , Anticuerpos/inmunología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular , Colforsina/farmacología , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/química , Proteínas de Unión al GTP , Células HEK293 , Células HeLa , Células Hep G2 , Humanos , Factores Inmunológicos/farmacología , Janus Quinasa 1/antagonistas & inhibidores , Janus Quinasa 1/inmunología , Proteínas de Neoplasias , Inhibidores de Fosfodiesterasa/farmacología , Hidrolasas Diéster Fosfóricas/metabolismo , Fosforilación/efectos de los fármacos , Receptor de Interferón alfa y beta/química , Receptor de Interferón alfa y beta/metabolismo , Receptores de Cinasa C Activada , Receptores de Superficie Celular , Factor de Transcripción STAT1/antagonistas & inhibidores , Factor de Transcripción STAT1/inmunología , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT2/antagonistas & inhibidores , Factor de Transcripción STAT2/inmunología , Factor de Transcripción STAT2/metabolismo , Transducción de Señal/efectos de los fármacos , TYK2 Quinasa/inmunología , TYK2 Quinasa/metabolismo
16.
PLoS One ; 8(7): e68391, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23950841

RESUMEN

Signal transducer and activator of transcription 3 (STAT3) is an oncogenic protein that is constitutively activated in numerous cancer cell lines and human cancers. Another STAT family member, STAT1, possesses cancer-inhibitory properties and can promote apoptosis in tumor cells upon activation. To better characterize these important cancer related genes, we tagged STAT3 and STAT1 loci with fluorescent protein (FP) sequences (RFP and GFP respectively) by targeted integration via zinc finger nuclease (ZFN)--mediated homologous recombination in A549 cells that express aberrantly activated STAT3. We inserted the FP transgenes at the N-terminus of the STAT3 locus and at the C-terminus of the STAT1 locus. The integration resulted in endogenous expression of fluorescent STAT3 and STAT1 chimeric fusion proteins. When stimulated with IL-6 or IFN-γ, the cells showed robust nuclear translocation of RFP-STAT3 or STAT1-GFP, respectively. Pre-incubation of cells with a known specific STAT3 inhibitor showed that IFN-γ-induced translocation of STAT1-GFP was not impaired. STAT3 activates multiple downstream targets such as genes involved in cell cycle progression - e.g. cyclin D1. To detect changes in expression of endogenous cyclin D1, we used ZFN technology to insert a secreted luciferase reporter behind the cyclin D1 promoter and separated the luciferase and cyclin D1 coding regions by a 2A sequence to induce a translational skip. The luciferase insertion was made in the RFP-STAT3/STAT1-GFP cell line to have all three reporters in a single cell line. Addition of a STAT3 inhibitor led to suppression of cyclin D1 promoter activity and cell growth arrest. The triple-modified cell line provides a simple and convenient method for high-content screening and pre-clinical testing of potential STAT3 inhibitors in live cells while ensuring that the STAT1 pathway is not affected. This approach of reporting endogenous gene activities using ZFN technology could be applied to other cancer targets.


Asunto(s)
Ciclina D1/genética , Genes Reporteros , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT3/genética , Secuencia de Bases , Línea Celular Tumoral , Ciclina D1/metabolismo , Evaluación Preclínica de Medicamentos/métodos , Proteínas Fluorescentes Verdes/análisis , Proteínas Fluorescentes Verdes/genética , Humanos , Luciferasas/análisis , Luciferasas/genética , Proteínas Luminiscentes/análisis , Proteínas Luminiscentes/genética , Datos de Secuencia Molecular , Mutagénesis Insercional , Ingeniería de Proteínas , Proteínas Recombinantes de Fusión/análisis , Recombinación Genética , Factor de Transcripción STAT1/antagonistas & inhibidores , Factor de Transcripción STAT3/antagonistas & inhibidores , Proteína Fluorescente Roja
17.
J Biol Chem ; 288(20): 14417-14427, 2013 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-23580655

RESUMEN

Signal transducers and activators of transcription 1 (STAT1) transduces signals from cytokines and growth factors, particularly IFN-γ, and regulates expression of genes involved in cell survival/death, proliferation, and migration. STAT1 is activated through phosphorylation on its tyrosine 701 by JAKs and is inactivated through dephosphorylation by tyrosine phosphatases. We discovered a natural compound, wedelolactone, that increased IFN-γ signaling by inhibiting STAT1 dephosphorylation and prolonging STAT1 activation through specific inhibition of T-cell protein tyrosine phosphatase (TCPTP), an important tyrosine phosphatase for STAT1 dephosphorylation. More interestingly, wedelolactone inhibited TCPTP through interaction with the C-terminal autoinhibition domain of TCPTP. We also found that wedelolactone synergized with IFN-γ to induce apoptosis of tumor cells. Our data suggest a new target for anticancer or antiproliferation drugs, a new mechanism to regulate PTPs specifically, and a new drug candidate for treating cancer or other proliferation disorders.


Asunto(s)
Antineoplásicos/farmacología , Cumarinas/farmacología , Regulación Neoplásica de la Expresión Génica , Interferón gamma/metabolismo , Factor de Transcripción STAT1/antagonistas & inhibidores , Línea Celular Tumoral , Supervivencia Celular , Ensayos de Selección de Medicamentos Antitumorales , Genes Reporteros , Células Hep G2 , Humanos , Monoéster Fosfórico Hidrolasas/metabolismo , Fosforilación , Extractos Vegetales/farmacología , Regiones Promotoras Genéticas , Interferencia de ARN
18.
Biol Pharm Bull ; 36(3): 399-406, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23318249

RESUMEN

Mollugin, a kind of naphthohydroquinone, is a major constituent isolated from Rubia cordifolia L. and demonstrated to possess anti-inflammatory activity in recent reports. However, the effects and mechanism of action of mollugin in inflammation have not been fully defined. The present study was therefore designed to investigate whether mollugin suppresses the inflammatory response in lipopolysaccharide (LPS)-stimulated RAW264.7 macrophages. Mollugin attenuated the LPS-induced expression of nitric oxide (NO), inducible nitric oxide synthase (iNOS), interleukin (IL)-1ß and IL-6 but augmented the expression of tumor necrosis factor (TNF)-α. Mollugin did not inhibit the degradation of inhibitory kappa B (IκB)-α or the nuclear translocation of p65 nuclear factor-kappa B (NF-κB) but rather enhanced the phosphorylation of p65 subunits evoked by LPS. Mollugin did not inhibit the phosphorylation of extracellular-signal-related kinase (ERK) 1/2, p38, and c-Jun N-terminal kinase (JNK) 1/2 either. Mollugin significantly reduced the LPS-mediated phosphorylation of Janus kinase (JAK) 2, signal transducers and activators of transcription (STAT) 1 and STAT3. Molecular docking analysis showed that mollugin binds to JAK2 in a manner similar to that of AG490, a specific JAK2 inhibitor. We conclude that mollugin may be a JAK2 inhibitor and inhibits LPS-induced inflammatory responses by blocking the activation of the JAK-STAT pathway.


Asunto(s)
Antiinflamatorios/farmacología , Medicamentos Herbarios Chinos/farmacología , Janus Quinasa 2/antagonistas & inhibidores , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Piranos/farmacología , Factor de Transcripción STAT1/antagonistas & inhibidores , Factor de Transcripción STAT3/antagonistas & inhibidores , Animales , Células Cultivadas , Interleucina-1beta/biosíntesis , Interleucina-6/biosíntesis , Janus Quinasa 2/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT3/metabolismo
19.
Psychopharmacology (Berl) ; 227(1): 149-63, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23254377

RESUMEN

RATIONALE: Hypericum perforatum, popularly called St. John's wort (SJW), is a medicinal plant mainly used as antidepressant with a favorable safety profile than standard antidepressants. Some studies have also documented other SJW bioactivities, including pain modulation. OBJECTIVES: The aim of this study was to demonstrate the capability of SJW to relieve nitric oxide (NO)-induced nociceptive hypersensitivity and identify the effective component. METHODS: Nociceptive hypersensitivity induced by administration of the NO donors nitroglycerin (GTN) and sodium nitroprusside (SNP) was assessed by cold and hot plate tests. The cellular pathways and molecular targets involved were investigated by Western blotting. RESULTS: GTN and SNP produced a prolonged allodynia and hyperalgesia in mice. A single oral administration of low doses of an SJW dried extract or purified hypericin reversed the NO donor-induced nociceptive behavior whereas hyperforin and flavoinoids were ineffective. Investigating into the cellular pathways involved, an increased CREB and STAT1 phosphorylation, and activation of NF-κB were detected within PAG and thalamus following NO donors' administration. These cellular events were prevented by SJW or hypericin. Since hypericin showed PKC blocking properties, a role of PKC as an upstream modulator of these transcription factors was hypothesized. NO donors increased expression and phosphorylation of protein kinase C (PKC) γ and ε isoforms, molecular events prevented by SJW or hypericin. CONCLUSIONS: SJW reversed NO-induced nociceptive hypersensitivity through the blockade of a supraspinal signaling pathway involving a PKC-dependent CREB, STAT1 and NF-κB activation due to presence of hypericin. These data indicate SJW/hypericin as a therapeutic perspective for pain treatment.


Asunto(s)
Proteína de Unión a CREB/metabolismo , Hiperalgesia/tratamiento farmacológico , Hypericum , FN-kappa B/metabolismo , Donantes de Óxido Nítrico/toxicidad , Perileno/análogos & derivados , Factor de Transcripción STAT1/metabolismo , Animales , Antracenos , Proteína de Unión a CREB/antagonistas & inhibidores , Frío/efectos adversos , Calor/efectos adversos , Hiperalgesia/inducido químicamente , Hiperalgesia/metabolismo , Masculino , Ratones , FN-kappa B/antagonistas & inhibidores , Óxido Nítrico/metabolismo , Dimensión del Dolor/efectos de los fármacos , Dimensión del Dolor/métodos , Perileno/farmacología , Perileno/uso terapéutico , Extractos Vegetales/aislamiento & purificación , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/metabolismo , Distribución Aleatoria , Factor de Transcripción STAT1/antagonistas & inhibidores
20.
Ann Rheum Dis ; 71(3): 440-7, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22121136

RESUMEN

OBJECTIVES: The objective of this study was to investigate the effect of the novel Janus kinase inhibitor CP-690,550 in fibroblast-like synoviocytes (FLSs) from patients with rheumatoid arthritis (RA). METHODS: RA FLSs were isolated from tissue obtained by arthroplasty, cultured and serum-starved 48 h prior to stimulation. Messenger RNA and protein levels were determined by quantitative PCR and ELISA or multiplex bead assay, respectively. Phosphorylation of STAT (signal transducers and activators of transcription) proteins was determined by western blot. RESULTS: Interleukin-6-induced phosphorylation of STAT1 and STAT3 was inhibited by CP-690,550 with IC(50) values of 23 and 77 nM, respectively. Unexpectedly, although tumour necrosis factor (TNF) did not induce immediate phosphorylation of either STAT, CP-690,550 inhibited TNF-induced expression of several chemokines (IP-10, RANTES and MCP1) at the messenger RNA and protein levels. Chemokine expression was inhibited by cycloheximide, implying a need for de novo protein synthesis, and cycloheximide abolished the effect of CP-690,550 (tofacitinib). TNF induced early interferon (IFN) ß expression and STAT1 phosphorylation beginning at 3 h, which was blocked by CP-690,550. The dependence of TNF-induced chemokine expression on type I IFN was confirmed in FLSs from mice lacking type I IFN receptors (IFNARs) and in RA FLSs using an IFNAR blocking antibody. CONCLUSIONS: The Janus kinase/STAT pathway in FLS is indirectly activated by TNF through autocrine expression of type I IFN, resulting in IFNAR engagement and production of T cell chemokines. These findings illuminate a novel role of CP-690,550 in the treatment of RA: the reduction of chemokine synthesis by FLS, thereby limiting recruitment of T cells and other infiltrating leucocytes.


Asunto(s)
Antirreumáticos/farmacología , Artritis Reumatoide/patología , Fibroblastos/efectos de los fármacos , Janus Quinasa 3/antagonistas & inhibidores , Pirimidinas/farmacología , Pirroles/farmacología , Membrana Sinovial/efectos de los fármacos , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Animales , Artritis Reumatoide/metabolismo , Comunicación Autocrina/efectos de los fármacos , Células Cultivadas , Quimiocinas/biosíntesis , Evaluación Preclínica de Medicamentos/métodos , Humanos , Interferón Tipo I/fisiología , Interleucina-6/antagonistas & inhibidores , Interleucina-6/farmacología , Ratones , Fosforilación/efectos de los fármacos , Piperidinas , Factor de Transcripción STAT1/antagonistas & inhibidores , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT3/antagonistas & inhibidores , Factor de Transcripción STAT3/metabolismo , Membrana Sinovial/patología , Factor de Necrosis Tumoral alfa/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA