Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
Más filtros

Medicinas Complementárias
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell Death Dis ; 15(2): 114, 2024 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-38321001

RESUMEN

As an alternative pathway for liver regeneration, liver progenitor cells and their derived ductular reaction cells increase during the progression of many chronic liver diseases. However, the mechanism underlying their hepatocyte repopulation after liver injury remains unknown. Here, we conducted progenitor cell lineage tracing in mice and found that fewer than 2% of hepatocytes were derived from liver progenitor cells after 9 weeks of injury with a choline-deficient diet supplemented with ethionine (CDE), and this percentage increased approximately three-fold after 3 weeks of recovery. We also found that the proportion of liver progenitor cells double positive for the ligand of glucocorticoid-induced tumour necrosis factor receptor (GITRL, also called Tnfsf18) and SRY-related HMG box transcription 9 (Sox9) among nonparenchymal cells increased time-dependently upon CDE injury and reduced after recovery. When GITRL was conditionally knocked out from hepatic progenitor cells, its expression in nonparenchymal cells was downregulated by approximately fifty percent, and hepatocyte repopulation increased by approximately three folds. Simultaneously, conditional knockout of GITRL reduced the proportion of liver-infiltrating CD8+ T lymphocytes and glucocorticoid-induced tumour necrosis factor receptor (GITR)-positive CD8+ T lymphocytes. Mechanistically, GITRL stimulated cell proliferation but suppressed the differentiation of liver progenitor organoids into hepatocytes, and CD8+ T cells further reduced their hepatocyte differentiation by downregulating the Wnt/ß-catenin pathway. Therefore, GITRL expressed by liver progenitor cells impairs hepatocyte differentiation, thus hindering progenitor cell-mediated liver regeneration.


Asunto(s)
Linfocitos T CD8-positivos , Glucocorticoides , Animales , Ratones , Linfocitos T CD8-positivos/patología , Fibrosis , Glucocorticoides/metabolismo , Hepatocitos/metabolismo , Inflamación/patología , Hígado/patología , Receptores del Factor de Necrosis Tumoral/metabolismo , Células Madre/metabolismo , Factores de Necrosis Tumoral/metabolismo
2.
J Biochem Mol Toxicol ; 36(10): e23154, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35734936

RESUMEN

Statins are widely used lipid-lowering drugs that cause many side effects. Withaferin-A (WA), popularly known as Ashwagandha, an ancient Indian medicinal herb, is extracted from Withania somnifera. Anti-atherosclerotic effect of WA has been reported. However, the mechanism remains unknown. Hence, we planned this study to investigate the WA mechanism in anti-atherosclerosis in a rat model. High cholesterol diet (HCD) was fed to induce atherosclerosis in Sprague-Dawley male rats. Five groups (N = 6 rats/group) were fed with normal diet, HCD, WA (10 mg/kg bw)+HCD, lovastatin (LS: 10 mg/kg bw)+HCD, WA (10 mg/kg bw) respectively for 90 days. Statistical analysis was done by GraphPad Prism (version 8.0.1) using one-way analysis of variance (ANOVA) followed by post hoc Duncan's test with a significance level (p < 0.05). The groups were compared for lipid profiles, oxidative stress, lipid peroxidation, inflammatory mediators, apoptotic markers, and histopathological changes in the liver and aorta. Treatment with HCD increased lipid profiles, inflammatory mediators, cytokines, and lipid peroxidation. WA as well as LS treatments significantly decreased these parameters restored the antioxidant status, and reduced lipid peroxidation (p < 0.05). Histopathological studies revealed that WA and LS reduced the hepatic fat and aortic plaque. WA reduced apoptosis via augmentation of phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) pathway; increased B-cell lymphoma 2 and inhibited Bcl-2 associated X-protein proapoptotic proteins; TNF receptor superfamily member 6, Bim, caspase-3, and -9; demonstrated significant hypolipidemic and anti-inflammatory properties against HCD induced atherosclerosis in rats through regulation of inflammatory mediators and apoptosis via the PI3K/AKT signaling pathway.


Asunto(s)
Aterosclerosis , Inhibidores de Hidroximetilglutaril-CoA Reductasas , Hipercolesterolemia , Animales , Antiinflamatorios/farmacología , Antioxidantes/metabolismo , Apoptosis , Aterosclerosis/tratamiento farmacológico , Caspasa 3/metabolismo , Colesterol , Citocinas/metabolismo , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Hipercolesterolemia/tratamiento farmacológico , Mediadores de Inflamación/metabolismo , Lovastatina , Masculino , Estrés Oxidativo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal
3.
Biomolecules ; 11(2)2021 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-33671607

RESUMEN

Tumor necrosis factor-α (TNF-α) is a drug target in rheumatoid arthritis and several other auto-immune disorders. TNF-α binds with TNF receptors (TNFR), located on the surface of several immunological cells to exert its effect. Hence, the use of inhibitors that can hinder the complex formation of TNF-α/TNFR can be of medicinal significance. In this study, multiple chem-informatics approaches, including descriptor-based screening, 2D-similarity searching, and pharmacophore modelling were applied to screen new TNF-α inhibitors. Subsequently, multiple-docking protocols were used, and four-fold post-docking results were analyzed by consensus approach. After structure-based virtual screening, seventeen compounds were mutually ranked in top-ranked position by all the docking programs. Those identified hits target TNF-α dimer and effectively block TNF-α/TNFR interface. The predicted pharmacokinetics and physiological properties of the selected hits revealed that, out of seventeen, seven compounds (4, 5, 10, 11, 13-15) possessed excellent ADMET profile. These seven compounds plus three more molecules (7, 8 and 9) were chosen for molecular dynamics simulation studies to probe into ligand-induced structural and dynamic behavior of TNF-α, followed by ligand-TNF-α binding free energy calculation using MM-PBSA. The MM-PBSA calculations revealed that compounds 4, 5, 7 and 9 possess highest affinity for TNF-α; 8, 11, 13-15 exhibited moderate affinities, while compound 10 showed weaker binding affinity with TNF-α. This study provides valuable insights to design more potent and selective inhibitors of TNF-α, that will help to treat inflammatory disorders.


Asunto(s)
Quimioinformática/métodos , Receptores del Factor de Necrosis Tumoral/metabolismo , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Simulación por Computador , Dimerización , Diseño de Fármacos , Descubrimiento de Drogas/métodos , Evaluación Preclínica de Medicamentos/métodos , Humanos , Inflamación , Ligandos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Unión Proteica , Conformación Proteica , Mapeo de Interacción de Proteínas , Bibliotecas de Moléculas Pequeñas/química
4.
J Exp Med ; 217(10)2020 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-32662821

RESUMEN

TNF plays a key role in immune-mediated inflammatory diseases including rheumatoid arthritis (RA) and spondyloarthritis (SpA). It remains incompletely understood how TNF can lead to different disease phenotypes such as destructive peripheral polysynovitis in RA versus axial and peripheral osteoproliferative inflammation in SpA. We observed a marked increase of transmembrane (tm) versus soluble (s) TNF in SpA versus RA together with a decrease in the enzymatic activity of ADAM17. In contrast with the destructive polysynovitis observed in classical TNF overexpression models, mice overexpressing tmTNF developed axial and peripheral joint disease with synovitis, enthesitis, and osteitis. Histological and radiological assessment evidenced marked endochondral new bone formation leading to joint ankylosis over time. SpA-like inflammation, but not osteoproliferation, was dependent on TNF-receptor I and mediated by stromal tmTNF overexpression. Collectively, these data indicate that TNF can drive distinct inflammatory pathologies. We propose that tmTNF is responsible for the key pathological features of SpA.


Asunto(s)
Artritis/metabolismo , Osteogénesis , Espondiloartritis/metabolismo , Factor de Necrosis Tumoral alfa/fisiología , Proteína ADAM17/metabolismo , Adulto , Animales , Artritis/etiología , Modelos Animales de Enfermedad , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Articulaciones/metabolismo , Masculino , Ratones , Receptores del Factor de Necrosis Tumoral/metabolismo , Espondiloartritis/etiología , Sinovitis/etiología , Sinovitis/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
5.
Front Immunol ; 10: 2040, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31555271

RESUMEN

An intricate network of molecular and cellular actors orchestrates the delicate balance between effector immune responses and immune tolerance. The pleiotropic cytokine tumor necrosis factor-alpha (TNF) proves as a pivotal protagonist promoting but also suppressing immune responses. These opposite actions are accomplished through specialist cell types responding to TNF via TNF receptors TNFR1 and TNFR2. Recent findings highlight the importance of TNFR2 as a key regulator of activated natural FoxP3+ regulatory T cells (Tregs) in inflammatory conditions, such as acute graft-vs.-host disease (GvHD) and the tumor microenvironment. Here we review recent advances in our understanding of TNFR2 signaling in T cells and discuss how these can reconcile seemingly conflicting observations when manipulating TNF and TNFRs. As TNFR2 emerges as a new and attractive target we furthermore pinpoint strategies and potential pitfalls for therapeutic targeting of TNFR2 for cancer treatment and immune tolerance after allogeneic hematopoietic cell transplantation.


Asunto(s)
Neoplasias/inmunología , Neoplasias/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Factores de Necrosis Tumoral/metabolismo , Estudios Clínicos como Asunto , Evaluación Preclínica de Medicamentos , Expresión Génica , Enfermedad Injerto contra Huésped/etiología , Trasplante de Células Madre Hematopoyéticas , Humanos , Terapia Molecular Dirigida , Neoplasias/genética , Neoplasias/terapia , Receptores del Factor de Necrosis Tumoral/genética , Transducción de Señal , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Trasplante Homólogo , Factores de Necrosis Tumoral/genética
6.
Biomed Pharmacother ; 92: 491-500, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28575806

RESUMEN

A number of drugs as well as lead molecules are isolated from natural sources. Phytol is one of such lead molecule belongs to terpenes group distributed widely in medicinal plants. In the present work, we investigated the cytotoxic behavior of phytol on human lung carcinoma cells (A549). Phytol was found to cause characteristic apoptotic morphological changes and generation of ROS in A549 cells. The mechanism of phytol involved the activation of TRAIL, FAS and TNF-α receptors along with caspase 9 and 3. In silico molecular docking studies revealed that phytol has a good binding affinity with glucose-6-phosphate dehydrogenase (G6PD), which is known to promote tumor proliferation. The ability of phytol to become potential drug candidate has been revealed from the pharmacokinetic study performed in the present study.


Asunto(s)
Caspasa 3/biosíntesis , Caspasa 9/biosíntesis , Glucosafosfato Deshidrogenasa/metabolismo , Neoplasias Pulmonares/metabolismo , Fitol/farmacología , Especies Reactivas de Oxígeno/metabolismo , Células A549 , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Línea Celular Tumoral , Inducción Enzimática/efectos de los fármacos , Inducción Enzimática/fisiología , Humanos , Fitol/química , Fitol/uso terapéutico , Estructura Secundaria de Proteína , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Receptor fas/metabolismo
7.
Sci Rep ; 7(1): 3424, 2017 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-28611375

RESUMEN

TNFα is a homotrimeric pro-inflammatory cytokine, whose direct targeting by protein biotherapies has been an undeniable success for the treatment of chronic inflammatory diseases. Despite many efforts, no orally active drug targeting TNFα has been identified so far. In the present work, we identified through combined in silico/in vitro/in vivo approaches a TNFα direct inhibitor, compound 1, displaying nanomolar and micromolar range bindings to TNFα. Compound 1 inhibits the binding of TNFα with both its receptors TNFRI and TNFRII. Compound 1 inhibits the TNFα induced apoptosis on L929 cells and the TNFα induced NF-κB activation in HEK cells. In vivo, oral administration of compound 1 displays a significant protection in a murine TNFα-dependent hepatic shock model. This work illustrates the ability of low-cost combined in silico/in vitro/in vivo screening approaches to identify orally available small-molecules targeting challenging protein-protein interactions such as homotrimeric TNFα.


Asunto(s)
Antiinflamatorios/farmacología , Simulación del Acoplamiento Molecular , Bibliotecas de Moléculas Pequeñas/farmacología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Administración Oral , Regulación Alostérica/efectos de los fármacos , Animales , Antiinflamatorios/administración & dosificación , Antiinflamatorios/química , Línea Celular Tumoral , Evaluación Preclínica de Medicamentos , Femenino , Células HEK293 , Ensayos Analíticos de Alto Rendimiento , Humanos , Ratones , Ratones Endogámicos BALB C , Unión Proteica/efectos de los fármacos , Receptores del Factor de Necrosis Tumoral/química , Receptores del Factor de Necrosis Tumoral/metabolismo , Bibliotecas de Moléculas Pequeñas/química , Factor de Necrosis Tumoral alfa/química , Factor de Necrosis Tumoral alfa/metabolismo
8.
SLAS Discov ; 22(8): 950-961, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28530838

RESUMEN

Tumor necrosis factor receptor 1 (TNFR1) is a transmembrane receptor that binds tumor necrosis factor or lymphotoxin-alpha and plays a critical role in regulating the inflammatory response. Upregulation of these ligands is associated with inflammatory and autoimmune diseases. Current treatments reduce symptoms by sequestering free ligands, but this can cause adverse side effects by unintentionally inhibiting ligand binding to off-target receptors. Hence, there is a need for new small molecules that specifically target the receptors, rather than the ligands. Here, we developed a TNFR1 FRET biosensor expressed in living cells to screen compounds from the NIH Clinical Collection. We used an innovative high-throughput fluorescence lifetime screening platform that has exquisite spatial and temporal resolution to identify two small-molecule compounds, zafirlukast and triclabendazole, that inhibit the TNFR1-induced IκBα degradation and NF-κB activation. Biochemical and computational docking methods were used to show that zafirlukast disrupts the interactions between TNFR1 pre-ligand assembly domain (PLAD), whereas triclabendazole acts allosterically. Importantly, neither compound inhibits ligand binding, proving for the first time that it is possible to inhibit receptor activation by targeting TNF receptor-receptor interactions. This strategy should be generally applicable to other members of the TNFR superfamily, as well as to oligomeric receptors in general.


Asunto(s)
Ensayos Analíticos de Alto Rendimiento/métodos , Receptores del Factor de Necrosis Tumoral/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , Técnicas Biosensibles , Dimerización , Evaluación Preclínica de Medicamentos , Transferencia Resonante de Energía de Fluorescencia , Células HEK293 , Humanos , Indoles , Ligandos , Simulación del Acoplamiento Molecular , Proteínas Mutantes/metabolismo , Inhibidor NF-kappaB alfa/metabolismo , FN-kappa B/metabolismo , Fenilcarbamatos , Dominios Proteicos , Proteolisis/efectos de los fármacos , Receptores del Factor de Necrosis Tumoral/química , Receptores del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal , Sulfonamidas , Compuestos de Tosilo/farmacología , Triclabendazol/farmacología
9.
Am J Chin Med ; 44(8): 1719-1735, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27848251

RESUMEN

Nuclear factor-[Formula: see text]B (NF-[Formula: see text]B)/Rel transcription factors are best known for their central roles in promoting cell survival in cancer. NF-[Formula: see text]B antagonizes tumor necrosis factor (TNF)-[Formula: see text]-induced apoptosis through a process involving attenuation of the c-Jun-N-terminal kinase (JNK). However, the role of JNK activation in apoptosis induced by negative regulation of NF-[Formula: see text]B is not completely understood. We found that allergen-removed Rhus verniciflua Stokes (aRVS) extract-mediated NF-[Formula: see text]B inhibition induces apoptosis in SKOV-3 ovarian cancer cells via the serial activation of caspases and SKOV-3 cells are most specifically suppressed by aRVS. Here, we show that in addition to activating caspases, aRVS extract negatively modulates the TNF-[Formula: see text]-mediated I[Formula: see text]B/NF-[Formula: see text]B pathway to promote JNK activation, which results in apoptosis. When the cytokine TNF-[Formula: see text] binds to the TNF receptor, I[Formula: see text]B dissociates from NF-[Formula: see text]B. As a result, the active NF-[Formula: see text]B translocates to the nucleus. aRVS extract (0.5[Formula: see text]mg/ml) clearly prevented NF-[Formula: see text]B from mobilizing to the nucleus, resulting in the upregulation of JNK phosphorylation. This subsequently increased Bax activation, leading to marked aRVS-induced apoptosis, whereas the JNK inhibitor SP600125 in aRVS extract treated SKOV-3 cells strongly inhibited Bax. Bax subfamily proteins induced apoptosis through caspase-3. Thus, these results indicate that aRVS extract contains components that inhibit NF-[Formula: see text]B signaling to upregulate JNK activation in ovarian cancer cells and support the potential of aRVS as a therapeutic agent for ovarian cancer.


Asunto(s)
Alérgenos/aislamiento & purificación , Apoptosis/efectos de los fármacos , Apoptosis/genética , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Extractos Vegetales/farmacología , Rhus/química , Caspasas/metabolismo , Femenino , Humanos , Proteínas I-kappa B/metabolismo , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , FN-kappa B/fisiología , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/metabolismo , Fosforilación/efectos de los fármacos , Fitoterapia , Extractos Vegetales/aislamiento & purificación , Receptores del Factor de Necrosis Tumoral/metabolismo , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/fisiología , Regulación hacia Arriba/efectos de los fármacos , Proteína X Asociada a bcl-2/antagonistas & inhibidores
10.
J Neuroinflammation ; 13(1): 265, 2016 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-27724971

RESUMEN

BACKGROUND: Nogo-A and its putative receptor NgR are considered to be among the inhibitors of axonal regeneration in the CNS. However, few studies so far have addressed the issue of local NgR complex multilateral localization within inflammation in an MS mouse model of autoimmune demyelination. METHODS: Chronic experimental autoimmune encephalomyelitis (EAE) was induced in C57BL/6 mice. Analyses were performed on acute (days 18-22) and chronic (day 50) time points and compared to controls. The temporal and spatial expression of the Nogo receptor complex (NgR and coreceptors) was studied at the spinal cord using epifluorescent and confocal microscopy or real-time PCR. Data are expressed as cells/mm2, as mean % ± SEM, or as arbitrary units of integrated density. RESULTS: Animals developed a moderate to severe EAE without mortality, followed by a progressive, chronic clinical course. NgR complex spatial expression varied during the main time points of EAE. NgR with coreceptors LINGO-1 and TROY was increased in the spinal cord in the acute phase whereas LINGO-1 and p75 signal seemed to be dominant in the chronic phase, respectively. NgR was detected on gray matter NeuN+ neurons of the spinal cord, within the white matter inflammatory foci (14.2 ± 4.3 % NgR+ inflammatory cells), and found to be colocalized with GAP-43+ axonal growth cones while no ß-TubIII+, SMI-32+, or APP+ axons were found as NgR+. Among the NgR+ inflammatory cells, 75.6 ± 9.0 % were microglial/macrophages (lectin+), 49.6 ± 14.2 % expressed CD68 (phagocytic ED1+ cells), and no cells were Mac-3+. Of these macrophages/monocytes, only Arginase-1+/NgR+ but not iNOS+/NgR+ were present in lesions both in acute and chronic phases. CONCLUSIONS: Our data describe in detail the expression of the Nogo receptor complex within the autoimmune inflammatory foci and suggest a possible immune action for NgR apart from the established inhibitory one on axonal growth. Its expression by inflammatory macrophages/monocytes could signify a possible role of these cells on axonal guidance and clearance of the lesioned area during inflammatory demyelination.


Asunto(s)
Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/patología , Encefalomielitis Autoinmune Experimental/patología , Regulación de la Expresión Génica/inmunología , Receptores Nogo/metabolismo , Transducción de Señal/fisiología , Animales , Antígenos de Diferenciación/metabolismo , Arginasa/metabolismo , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/complicaciones , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Adyuvante de Freund/inmunología , Adyuvante de Freund/toxicidad , Regulación de la Expresión Génica/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Glicoproteína Mielina-Oligodendrócito/inmunología , Glicoproteína Mielina-Oligodendrócito/toxicidad , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nogo/genética , Proteínas Nogo/metabolismo , Receptores Nogo/genética , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/toxicidad , Receptores de Factor de Crecimiento Nervioso/genética , Receptores de Factor de Crecimiento Nervioso/metabolismo , Receptores del Factor de Necrosis Tumoral/genética , Receptores del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Estadísticas no Paramétricas
11.
Ann N Y Acad Sci ; 1383(1): 5-20, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27505256

RESUMEN

Progranulin (PGRN) is a growth factor with a unique beads-on-a-string structure that is involved in multiple pathophysiological processes, including anti-inflammation, tissue repair, wound healing, neurodegenerative diseases, and tumorigenesis. This review presents up-to-date information concerning recent studies on the role of PGRN in inflammatory arthritis and osteoarthritis, with a special focus on the involvement of the interactions and interplay between PGRN and tumor necrosis factor receptor (TNFR) family members in regulating such musculoskeletal diseases. In addition, this paper highlights the applications of atsttrin, an engineered protein comprising three TNFR-binding fragments of PGRN, as a promising intervention in treating arthritis.


Asunto(s)
Artritis/tratamiento farmacológico , Artritis/metabolismo , Péptidos y Proteínas de Señalización Intercelular/fisiología , Receptores del Factor de Necrosis Tumoral/metabolismo , Animales , Humanos , Péptidos y Proteínas de Señalización Intercelular/uso terapéutico , Progranulinas , Unión Proteica/fisiología , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/uso terapéutico , Transducción de Señal/fisiología , Factor de Necrosis Tumoral alfa/metabolismo
12.
Zhongguo Zhong Xi Yi Jie He Za Zhi ; 36(3): 339-44, 2016 Mar.
Artículo en Chino | MEDLINE | ID: mdl-27236893

RESUMEN

OBJECTIVE: To study the inhibitory effect of paeoniflorin (PAE) on TNF-α-induced TNF receptor type I (TNFR1)-mediated signaling pathway in mouse renal arterial endothelial cells (AECs) and to explore its underlying molecular mechanisms. METHODS: Mouse AECs were cultured in vitro and then they were treated by different concentrations PAE or TNF-α for various time periods. Expression levels of intercellular cell adhesion molecule-1 (ICAM-1) were detected in the normal group (cultured by serum-free culture media), the TNF-α group (cultured by 2-h serum-free culture media plus 6-h TNF-α 30 ng/mL), the low dose PAE group (cultured by 2-h PAE 0.8 µmo/L plus 6-h TNF-α 30 ng/mL), the middle dose PAE group (cultured by 2-h PAE 8 µmol/L plus 6-h TNF-α 30 ng/mL), the high dose PAE group (cultured by 2-h PAE 80 µmol/L plus 6-h TNF-α 30 ng/mL) with Western blot analysis. Nuclear translocation of transcription factor NF-κB (NE-κB) was detected in the normal group (cultured by serum-free culture media), the TNF-α group (cultured by 2-h serum-free culture media plus 45-mm TNF-α 30 ng/mL), and the high dose PAE group (cultured by 2-h PAE 80 µmol/L plus 45-min TNF-α 30 ng/mL) by immunofluorescent staining. Expression levels of the phosphorylation of extracellular signal-regulated (protein) kinase (ph-ERK) and p38 (ph- p38) were detected in the normal group (cultured by serum-free culture media) and the high dose PAE group (2-h PAE 80 µmol/L culture) by Western blot. NF-κB inhibitor-α (IκBα) protein expressions were detected in the normal group (cultured by serum-free culture media), the TNF-α group (cultured by 2-h serum-free culture media plus 30-min TNF-α 30 ng/mL), the high dose PAE group (cultured by 2-h PAE 80 µmol/L plus 30-min TNF-α 30 ng/mL), the p38 inhibitor group (SB group, pretreatment with SB238025 25 µmol/L for 30 min, then treated by PAE 80 µmol/L for 2 h, and finally treated by TNF-α 30 ng/mL for 30 min), the ERK inhibitor group (PD group, treated by PD98059 50 µmol/L for 30 min, then treated by PAE 80 µmol/L for 2 h, and finally treated by TNF-α 30 ng/mL for 30 min) by Western blot. RESULTS: Compared with the normal group, ICAM-1 protein expression levels obviously increased (P < 0.01). Compared with the TNFα group, ICAM-1 protein expression levels were obviously inhibited in the high dose PAE group (P < 0.05). Protein expression levels of ph-p38 and ph-ERK were obviously higher in the hIgh dose PAE group (P < 0.05). Compared with the normal group, IκBα protein expression levels obviously decreased in the TNF-α group (P < 0.01). Compared with the TNFα group, TNF-α-induced IκBα degradation could be significantly inhibited in the high dose PAE group (P < 0.01); the inhibition of PAE on IκBα degradation could be significantly inhibited in the SB group (P < 0.05). NF-κB/p65 signal was mainly located in cytoplasm in the normal group. NF-κB/p65 was translocated from cytoplasm to nucleus after stimulated by 45 min TNF-α in the TNF-α group, while it could be significantly inhibited in the high dose PAE group. CONCLUSIONS: PAE inhibited TNF-α-induced expression of lCAM-1. Its action might be associated with inhibiting TNFR1/NF-κB signaling pathway. p38 participated and mediated these actions.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Glucósidos/farmacología , Monoterpenos/farmacología , FN-kappa B/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Células Cultivadas , Células Endoteliales/citología , Molécula 1 de Adhesión Intercelular/metabolismo , Ratones , Factor de Necrosis Tumoral alfa/farmacología
14.
Int Immunopharmacol ; 28(2): 1044-9, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26344431

RESUMEN

Evodiamine has been reported to exhibit anti-inflammatory and anti-nociceptive effects, but the underlying mechanisms remain to be defined. P2X4 receptor (P2X4R) is a subtype of ATP receptors and plays important roles in pain, inflammatory and immune responses. We aimed to investigate whether evodiamine has beneficial effects on endothelial inflammatory injury mediated by chronic high glucose condition. We found that culturing human umbilical vein endothelial cells (HUVECs) with high glucose significantly increased the expression of P2X4 receptor in HUVECs, cytosolic Ca(2+) concentrations and intracellular reactive oxygen species (ROS) while decreasing nitric oxide (NO); these effects could be reversed by evodiamine. High glucose also significantly increased the expression of the pro-inflammatory activators (NF-κB) and TNFR-ɑ, which was accompanied by the elevation of P2X4R levels. Evodiamine was able to down-regulate the elevated NF-κB, TNFR-ɑ, P2X4R and ROS, and up-regulate the decreased NO. Thus the evodiamine may exert the anti-inflammation activity on high-glucose challenge HUVEC via suppressing the P2X4R signaling pathway, exhibiting beneficial ability to protect HUVECs from glucotoxicity.


Asunto(s)
Endotelio Vascular/efectos de los fármacos , Evodia , Inflamación/tratamiento farmacológico , Quinazolinas/farmacología , Receptores Purinérgicos P2X4/metabolismo , Medicamentos Herbarios Chinos , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Glucosa/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , FN-kappa B/metabolismo , Óxido Nítrico/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptores Purinérgicos P2X4/genética , Receptores del Factor de Necrosis Tumoral/metabolismo , Venas Umbilicales/patología , Regulación hacia Arriba/efectos de los fármacos
15.
Nat Prod Commun ; 10(2): 253-6, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25920254

RESUMEN

The effect of puerarin (Pur) on expressions of Fas/FasL mRNAs in pulmonary ischemia and reperfusion injury (PIRI) in rabbit was investigated. The sole side lung ischemia and reperfusion model was used. Rabbits were randomly divided into three groups, a sham operated group (sham, n = 10), PIR group (IR, n = 30) and PIR + Pur group (Pur, n = 30). Changes of several parameters including apoptotic index (AI), wet to dry ratio of lung tissue weight (W/D) and index of quantitative assessment of histologic lung injury (IQA) were measured after 60, 180 and 300 minutes of reperfusion. Meanwhile, the location and expression of Fas/FasL mRNA were investigated. Lung tissue was prepared for light microscopic and electron microscopic observation after 60, 180 and 300 minutes of reperfusion. Compared with group IR, Fas/FasL mRNAs were slightly expressed in intima and extima of small pulmonary artery, alveoli, and bronchiole epithelia in group Pur. The values of AI, W/D and IQA were significantly lower than those in group IR after 60, 180, and 300 minutes of reperfusion in lung tissue (P <0.01 or P <0.05). Meanwhile, the abnormal changes in lung tissue morphology were markedly less in group Pur. Puerarin notably protects lung from PIRI by inhibiting Fas/FasL mRNA expression and decreasing lung cell apoptosis in rabbits.


Asunto(s)
Proteína Ligando Fas/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Isoflavonas/uso terapéutico , ARN Mensajero/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Daño por Reperfusión/complicaciones , Animales , Proteína Ligando Fas/genética , Femenino , Lesión Pulmonar/tratamiento farmacológico , Lesión Pulmonar/metabolismo , Masculino , ARN Mensajero/genética , Conejos , Receptores del Factor de Necrosis Tumoral/genética , Vasodilatadores/uso terapéutico
16.
Oncotarget ; 6(9): 7280-92, 2015 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-25730901

RESUMEN

We studied whether bee venom (BV) inhibits cervical tumor growth through enhancement of death receptor (DR) expressions and inactivation of nuclear factor kappa B (NF-κB) in mice. In vivo study showed that BV (1 mg/kg) inhibited tumor growth. Similar inhibitory effects of BV on cancer growth in primary human cervical cancer cells were also found. BV (1-5 µg/ml) also inhibited the growth of cancer cells, Ca Ski and C33Aby the induction of apoptotic cell death in a dose dependent manner. Agreed with cancer cell growth inhibition, expression of death receptors; FAS, DR3 and DR6, and DR downstream pro-apoptotic proteins including caspase-3 and Bax was concomitantly increased, but the NF-κB activity and the expression of Bcl-2 were inhibited by treatment with BV in tumor mice, human cancer cell and human tumor samples as well as cultured cancer cells. In addition, deletion of FAS, DR3 and DR6 by small interfering RNA significantly reversed BV-induced cell growth inhibitory effects as well as NF-κB inactivation. These results suggest that BV inhibits cervical tumor growth through enhancement of FAS, DR3 and DR6 expression via inhibition of NF-κB pathway.


Asunto(s)
Venenos de Abeja/uso terapéutico , Neoplasias del Cuello Uterino/tratamiento farmacológico , Animales , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , FN-kappa B/metabolismo , ARN Interferente Pequeño/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Miembro 25 de Receptores de Factores de Necrosis Tumoral/metabolismo , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto , Receptor fas/metabolismo
17.
Mol Endocrinol ; 29(4): 571-82, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25719402

RESUMEN

The nuclear receptor farnesoid X receptor (FXR) (nuclear receptor subfamily 1, group H, member 4, or NR1H4) is highly expressed in the liver and intestine. Previous reports have suggested beneficial functions of FXR in the homeostasis of bile acids, lipids, and glucose, as well as in promoting liver regeneration and inhibiting carcinogenesis. To investigate the effect of chronic FXR activation in vivo, we generated transgenic mice that conditionally and tissue specifically express the activated form of FXR in the liver and intestine. Unexpectedly, the transgenic mice showed several intriguing phenotypes, including partial neonatal lethality, growth retardation, and spontaneous liver toxicity. The transgenic mice also displayed heightened sensitivity to a high-cholesterol diet-induced hepatotoxicity but resistance to the gallstone formation. The phenotypes were transgene specific, because they were abolished upon treatment with doxycycline to silence the transgene expression. The perinatal toxicity, which can be rescued by a maternal vitamin supplement, may have resulted from vitamin deficiency due to low biliary bile acid output as a consequence of inhibition of bile acid formation. Our results also suggested that the fibroblast growth factor-inducible immediate-early response protein 14 (Fn14), a member of the proinflammatory TNF family, is a FXR-responsive gene. However, the contribution of Fn14 induction in the perinatal toxic phenotype of the transgenic mice remains to be defined. Because FXR is being explored as a therapeutic target, our results suggested that a chronic activation of this nuclear receptor may have an unintended side effect especially during the perinatal stage.


Asunto(s)
Colesterol/toxicidad , Mucosa Intestinal/metabolismo , Hígado/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Ácidos y Sales Biliares/metabolismo , Dieta Alta en Grasa , Intestinos/efectos de los fármacos , Hígado/efectos de los fármacos , Hepatopatías/metabolismo , Ratones , Ratones Transgénicos , Receptores Citoplasmáticos y Nucleares/genética , Receptores del Factor de Necrosis Tumoral/genética , Receptores del Factor de Necrosis Tumoral/metabolismo , Receptor de TWEAK , Vitamina A/sangre , Vitamina E/sangre
18.
J Cancer Res Clin Oncol ; 139(2): 315-25, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23073510

RESUMEN

BACKGROUND: The receptor for the cytokine TWEAK (TweakR) is a cell surface member of the tumor necrosis factor receptor superfamily with diverse biological roles. TNFRSF family members are appealing therapeutic targets in oncology due to their aberrant expression and function in tumor cells. The goal of the current study was to examine the potential of TweakR as a therapeutic target in breast cancer. METHODS: Expression of TweakR in primary breast cancer tissues and metastases was characterized using immunohistochemistry. To determine the functional relevance of TweakR, breast cancer cell lines were treated in vitro and in vivo with enavatuzumab, a humanized mAb against TweakR. RESULTS: Overexpression of TweakR was observed in infiltrating tumors compared to normal adjacent breast tissues, and strong staining of TweakR was observed in all subtypes of invasive ductal breast cancer. In addition, a positive correlation of TweakR and HER2 expression and co-localization were observed, irrespective of ER status. TweakR expression was also observed in bone metastasis samples from primary breast cancer but rarely in benign tumors. Enavatuzumab inhibited the in vitro growth of TweakR-expressing breast cancer cell lines, and this activity was augmented by cross-linking the mAb. In addition, enavatuzumab significantly inhibited the in vivo growth of multiple breast cancer xenograft models including a model of metastasis. CONCLUSIONS: TweakR is highly expressed in all subtypes of invasive ductal breast cancer, and enavatuzumab administration exhibited a dose-dependent inhibition of primary tumor growth and lung metastasis and enhanced the antitumor activity of several chemotherapy agents currently used to treat breast cancer. These data provide the rationale to evaluate enavatuzumab as a potential therapy for the treatment of breast cancer.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Antineoplásicos/uso terapéutico , Carcinoma Ductal de Mama/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Animales , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales Humanizados/farmacología , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Carcinoma Ductal de Mama/tratamiento farmacológico , Carcinoma Ductal de Mama/genética , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Sinergismo Farmacológico , Femenino , Expresión Génica , Humanos , Ratones , Invasividad Neoplásica/genética , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Receptores del Factor de Necrosis Tumoral/genética , Receptor de TWEAK , Trastuzumab , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Int J Cardiol ; 167(5): 1719-23, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23245690

RESUMEN

Tumor necrosis factor-alpha (TNF-α) blockers are widely used in the treatment of chronic inflammatory diseases, especially chronic arthritis. Current guidelines advise against the use of such agents in patients who have a concomitant heart failure. Consequently, a group of patients with a devastating inflammatory disease cannot benefit from an excellent treatment option. After a critical review of the current literature, we conclude that there is not sufficient evidence to warn against such a regimen if recommended standard doses are used. A negative effect on the heart function seems to occur if unconventional high doses of TNF-α blockers are given. The theoretical background for this is discussed.


Asunto(s)
Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/epidemiología , Inmunosupresores/administración & dosificación , Receptores del Factor de Necrosis Tumoral/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Animales , Artritis/tratamiento farmacológico , Artritis/epidemiología , Artritis/metabolismo , Relación Dosis-Respuesta a Droga , Etanercept , Insuficiencia Cardíaca/metabolismo , Humanos , Inmunoglobulina G/administración & dosificación , Ensayos Clínicos Controlados Aleatorios como Asunto/métodos , Receptores del Factor de Necrosis Tumoral/administración & dosificación , Receptores del Factor de Necrosis Tumoral/metabolismo , Síndrome de Respuesta Inflamatoria Sistémica/tratamiento farmacológico , Síndrome de Respuesta Inflamatoria Sistémica/epidemiología , Síndrome de Respuesta Inflamatoria Sistémica/metabolismo , Factor de Necrosis Tumoral alfa/fisiología
20.
Zhongguo Zhen Jiu ; 32(11): 1012-8, 2012 Nov.
Artículo en Chino | MEDLINE | ID: mdl-23213990

RESUMEN

OBJECTIVE: To explore the intervention timing of acupuncture in treatment of cerebral infarction and the relationship of cerebral ischemia reperfusion injury with inflammatory cytokine receptor. METHODS: One hundred and ten male healthy Wistar rats were randomly divided into a normal group (n=10), a sham operation group (n=10), a model group (n=10), an acupuncture at non-acupoint group (non-acupoint group, n=40), an acupuncture with regaining consciousness method group (regaining consciousness group, n=40). Four subgroups were set up 1 h ischemia reperfusion in 1 h group, 3 h group, 6 h group, 12 h group in the two acupuncture groups, 10 rats in each subgroup. Two acupuncture groups were treated with acupuncture at four time points (1 h, 3 h, 6 h and 12 h after ischemia reperfusion), and "Shuigou" (GV 26) and "Neiguan" (PC 6) were selected in regaining consciousness group, and the non-acupoints below the bilateral costal region were selected in non-acupoint group. At the corresponding time point, the tissues of the brain were removed and interleukin1 receptor (IL-1RI) and tumor necrosis factor receptor (TNFR-I) mRNA and protein changes were detected by using real-time quantitative polymerase chain reaction and immunoblot assay. RESULTS: The expression of IL-1RI and TNFR-I mRNA and protein in the model group were significantly higher than that in normal group, sham operation group, regaining consciousness group and non-acupoint group (P<0.01, P<0.05). The expression of IL-1RI and TNFR-I mRNA and protein in regaining consciousness group was weakest at 3 h after reperfusion followed successively by 6 h, 1 h, 12 h, and there was no significantly change of IL-1RI and TNFR-I mRNA and protein expression in non-acupoint group among different timing points, but which was decreased as compared with those in the model group at the same time point (all P<0.05). CONCLUSION: Acupuncture can reduce the expression of IL-1RI and TNFR-I mRNA and protein in rats with cerebral ischemia reperfusion, inhibit the excessive expression of proinflammatory cytokine receptor, block apoptosis signal transduction and extend time window for treatment of cerebral ischemia, so as to play the protective effect for brain. Within 3 h of ischemia is the best time for intervention of acupuncture treatment.


Asunto(s)
Terapia por Acupuntura , Isquemia Encefálica/cirugía , Receptores de Interleucina-1/genética , Receptores del Factor de Necrosis Tumoral/genética , Daño por Reperfusión/genética , Daño por Reperfusión/terapia , Animales , Isquemia Encefálica/genética , Isquemia Encefálica/metabolismo , Humanos , Masculino , Ratas , Ratas Wistar , Receptores de Interleucina-1/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Reperfusión , Daño por Reperfusión/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA