Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 951
Filtrar
1.
Int J Mol Sci ; 25(4)2024 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-38396824

RESUMEN

Sasanquasaponin (SQS), a secondary metabolite that is derived from Camellia seeds, reportedly possesses notable biological properties. However, the anti-inflammatory effects of SQS and its underlying mechanisms remain poorly explored. Herein, we aimed to investigate the anti-inflammatory properties of SQS against lipopolysaccharide (LPS)-induced inflammatory responses in RAW264.7 cells, focusing on the nuclear factor-κB (NF-κB) and MAPK signaling pathways. SQS was isolated using a deep eutectic solvent and D101 macroporous adsorption resin and analyzed using high-performance liquid chromatography. The viability of LPS-stimulated RAW264.7 was assessed using the CCK-8 assay. The presence of reactive oxygen species (ROS) was evaluated using 2',7'-dichlorofluorescein-diacetate. The expression levels of inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß), and interleukin-6 (IL-6) were detected using reverse transcription-quantitative PCR and ELISA. Western blot was performed to analyze the protein expression of LPS-induced RAW264.7 cells. Herein, SQS exhibited anti-inflammatory activity: 30 µg/mL of SQS significantly reduced ROS generation, inhibited the LPS-induced expression of iNOS and COX-2, and attenuated the production of pro-inflammatory cytokines IL-1ß, IL-6, and TNF-α. The anti-inflammatory activity was potentially mediated by inhibiting the phosphorylation of IκBα and p65 in the NF-κB signaling pathway and the phosphorylation of ERK and JNK in the MAPK signaling pathway. Accordingly, SQS could inhibit inflammation in LPS-induced RAW264.7 cells by suppressing the NF-κB and MAPK signaling pathways. This study demonstrated the potential application of SQS as an anti-inflammatory agent.


Asunto(s)
FN-kappa B , Saponinas , Factor de Necrosis Tumoral alfa , Animales , Ratones , Antiinflamatorios/farmacología , Ciclooxigenasa 2/metabolismo , Interleucina-6/farmacología , Lipopolisacáridos/farmacología , FN-kappa B/metabolismo , Óxido Nítrico/metabolismo , Células RAW 264.7 , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo
2.
Int J Mol Sci ; 24(5)2023 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-36901935

RESUMEN

Oral squamous cell carcinoma (OSCC) is the sixth most common type of cancer worldwide. Despite advancement in treatment, advanced-stage OSCC is associated with poor prognosis and high mortality. The present study aimed to investigate the anticancer activities of semilicoisoflavone B (SFB), which is a natural phenolic compound isolated from Glycyrrhiza species. The results revealed that SFB reduces OSCC cell viability by targeting cell cycle and apoptosis. The compound caused cell cycle arrest at the G2/M phase and downregulated the expressions of cell cycle regulators including cyclin A and cyclin-dependent kinase (CDK) 2, 6, and 4. Moreover, SFB induced apoptosis by activating poly-ADP-ribose polymerase (PARP) and caspases 3, 8, and 9. It increased the expressions of pro-apoptotic proteins Bax and Bak, reduced the expressions of anti-apoptotic proteins Bcl-2 and Bcl-xL, and increased the expressions of the death receptor pathway protein Fas cell surface death receptor (FAS), Fas-associated death domain protein (FADD), and TNFR1-associated death domain protein (TRADD). SFB was found to mediate oral cancer cell apoptosis by increasing reactive oxygen species (ROS) production. The treatment of the cells with N-acetyl cysteine (NAC) caused a reduction in pro-apoptotic potential of SFB. Regarding upstream signaling, SFB reduced the phosphorylation of AKT, ERK1/2, p38, and JNK1/2 and suppressed the activation of Ras, Raf, and MEK. The human apoptosis array conducted in the study identified that SFB downregulated survivin expression to induce oral cancer cell apoptosis. Taken together, the study identifies SFB as a potent anticancer agent that might be used clinically to manage human OSCC.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias de la Boca , Humanos , Apoptosis/efectos de los fármacos , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/metabolismo , Línea Celular Tumoral , Proliferación Celular , Quinasas de Proteína Quinasa Activadas por Mitógenos , Neoplasias de la Boca/tratamiento farmacológico , Neoplasias de la Boca/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas ras/efectos de los fármacos , Proteínas ras/metabolismo , Proteínas Proto-Oncogénicas c-raf/efectos de los fármacos , Proteínas Proto-Oncogénicas c-raf/metabolismo
3.
Anticancer Drugs ; 33(1): e235-e246, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34419964

RESUMEN

One cost-effective way for identifying novel cancer therapeutics is in the repositioning of available drugs for which current therapies are inadequate. Levofloxacin prevents DNA duplication in bacteria by inhibiting the activity of DNA helicase. As eukaryotic cells have similar intracellular biologic characteristics as prokaryotic cells, we speculate that antibiotics inhibiting DNA duplication in bacteria may also affect the survival of cancer cells. Here we report that levofloxacin significantly inhibited the proliferation and clone formation of cancer cells and xenograft tumor growth through cell cycle arrest at G2/M and by enhancing apoptosis. Levofloxacin significantly altered gene expression in a direction favoring anticancer activity. THBS1 and LAPTM5 were dose-dependently upregulated whereas SRD5A3, MFAP5 and P4HA1 were downregulated. Pathway analysis revealed that levofloxacin significantly regulated canonical oncogenic pathways. Specific network enrichment included a MAPK/apoptosis/cytokine-cytokine receptor interaction pathway network that associates with cell growth, differentiation, cell death, angiogenesis and development and repair processes and a bladder cancer/P53 signaling pathway network mediating the inhibition of angiogenesis and metastasis. THBS1 overlapped in 16 of the 22 enriched apoptotic pathways and the 2 pathways in the bladder cancer/P53 signaling pathway network. P4HA1 enriched in 7 of the top 10 molecular functions regulated by differential downregulated genes. Our results indicate that levofloxacin has broad-spectrum anticancer activity with the potential to benefit cancer patients already treated or requiring prophylaxis for an infectious syndrome. The efficacy we find with levofloxacin may provide insight into the discovery and the design of novel less toxic anticancer drugs.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Levofloxacino/farmacología , Animales , Apoptosis/efectos de los fármacos , Moléculas de Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Citocinas/efectos de los fármacos , ADN Helicasas/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Masculino , Proteínas de la Membrana/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Exp Cell Res ; 403(1): 112550, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33675806

RESUMEN

Increased matrix metalloproteinase 9 (MMP9) expression is involved in delayed wound healing in diabetic foot ulcers. We created skin wounds in normal SD rats and STZ-induced diabetic SD rats, then we found protein levels of activator protein-1 (AP1), a crucial transcription factor to increase MMP9 transcription, as well as MMP9 was up-regulated in epithelium of diabetic skin tissues. Then, we evaluated the mRNA and protein stability of AP1 subunits C-FOS/C-Jun in HaCaT cells after high glucose treatment. Results showed that high glucose could increase protein stability of C-FOS and C-Jun. Additionally, high glucose also activated extracellular signaling-related kinase 1/2 (ERK1/2). ERK1/2 inhibitor could rescue phosphorylation of C-FOS and C-Jun, increased protein stability of C-Jun, and increased MMP9 expressions. Thus, our study demonstrated that high glucose could activate ERK1/2 to stabilize AP1 and increase MMP9 expression in diabetic skin and HaCaT cells.


Asunto(s)
Pie Diabético/tratamiento farmacológico , Glucosa/farmacología , Metaloproteinasa 9 de la Matriz/metabolismo , Factor de Transcripción AP-1/metabolismo , Animales , Diabetes Mellitus/tratamiento farmacológico , Pie Diabético/metabolismo , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Factor de Transcripción AP-1/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
5.
Am J Pathol ; 190(10): 2155-2164, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32679231

RESUMEN

Aberrant DNA methylation is an epigenetic hallmark of melanoma, but the expression of DNA methyltransferase (Dnmt)-1 in melanocytic tumors is unknown. Dnmt1 expression was analyzed in primary melanocytes, melanoma cell lines, and 83 melanocytic tumors, and its associations with proliferation, mutational status, and response to B-Raf and mitogen-activated protein kinase kinase (MEK) inhibition were explored. Dnmt1 expression was increased incrementally from nevi [mean fluorescence intensity (MFI), 48.1; interquartile range, 41.7 to 59.6] to primary melanomas (MFI, 68.8; interquartile range, 58.4 to 77.0) and metastatic melanomas (MFI, 87.5; interquartile range, 77.1 to 114.5) (P < 0.001). Dnmt1 expression was correlated with Ki-67 expression (Spearman correlation, 0.483; P < 0.001) and was independent of BRAF mutation status (P = 0.55). In BRAF-mutant melanoma, Dnmt1 was down-regulated during response to B-Raf and MEK inhibition and was again up-regulated on drug resistance in vitro and in vivo. Degradation of Dnmt1 by the histone deacetylase inhibitor suberoylanilide hydroxamic acid was associated with decreased cell viability in B-Raf inhibitor-sensitive and -resistant cell lines. This study demonstrates that Dnmt1 expression is correlated with proliferation in melanocytic tumors, is increased with melanoma progression, and is associated with response to B-Raf and MEK inhibition. Given its strong expression in metastatic melanoma, Dnmt1 may be a promising target for combined epigenetic and immunotherapy.


Asunto(s)
Proliferación Celular/efectos de los fármacos , ADN/metabolismo , Melanoma/metabolismo , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Proteínas Proto-Oncogénicas B-raf/metabolismo , Neoplasias Cutáneas/metabolismo , Línea Celular Tumoral , ADN/efectos de los fármacos , Inhibidores de Histona Desacetilasas/uso terapéutico , Humanos , Melanocitos/efectos de los fármacos , Melanocitos/metabolismo , Melanoma/genética , Melanoma/patología , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas B-raf/genética , Neoplasias Cutáneas/genética , Vorinostat/farmacología , Melanoma Cutáneo Maligno
6.
Invest New Drugs ; 39(1): 163-174, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32737717

RESUMEN

BACKGROUND: This Phase Ib study explored combination dosing of the allosteric MEK1/2 inhibitor cobimetinib and the ATP-competitive pan-AKT inhibitor ipatasertib. METHODS: Patients with advanced solid tumors were enrolled to two dose escalation arms, each using a 3 + 3 design in 28-day cycles. In Arm A, patients received concurrent cobimetinib and ipatasertib on days 1-21. In Arm B, cobimetinib was administered intermittently with ipatasertib for 21 days. Primary objectives evaluated dose-limiting toxicities (DLTs), maximum tolerated doses (MTD), and the recommended Phase II dose (RP2D). Secondary objectives included analysis of pharmacokinetic parameters, MAPK and PI3K pathway alterations, changes in tissue biomarkers, and preliminary anti-tumor efficacy. Expansion cohorts included patients with PTEN-deficient triple-negative breast cancer and endometrial cancer. RESULTS: Among 66 patients who received ≥1 dose of study drug, all experienced an adverse event (AE). Although no DLTs were reported, 6 patients experienced Cycle 1 DLT-equivalent AEs. The most common treatment-related AEs were diarrhea, nausea, vomiting, dermatitis acneiform, and fatigue. Thirty-five (53%) patients experienced drug-related AEs of ≥ grade 3 severity. Cobimetinb/ipatasertib MTDs were 60/200 mg on Arm A and 150/300 mg on Arm B; the latter was chosen as the RP2D. No pharmacokinetic interactions were identified. Biomarker analyses indicated pathway blockade and increases in IFNγ and PD-L1 gene expression following the combination. Three patients with endometrial or ovarian cancer achieved partial response, all with PTEN-low disease and two with tumor also harboring KRAS mutation. CONCLUSION: There was limited tolerability and efficacy for this MEK and AKT inhibitor combination. Nonetheless, pharmacodynamic analyses indicated target engagement and suggest rationale for further exploration of cobimetinib or ipatasertib in combination with other anticancer agents. ClinicalTrials.gov identifier: NCT01562275.


Asunto(s)
Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Azetidinas/farmacología , Azetidinas/uso terapéutico , Piperazinas/farmacología , Piperazinas/uso terapéutico , Piperidinas/farmacología , Piperidinas/uso terapéutico , Pirimidinas/farmacología , Pirimidinas/uso terapéutico , Anciano , Anciano de 80 o más Años , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Azetidinas/efectos adversos , Azetidinas/farmacocinética , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Fosfatidilinositol 3-Quinasas/efectos de los fármacos , Piperazinas/efectos adversos , Piperazinas/farmacocinética , Piperidinas/efectos adversos , Piperidinas/farmacocinética , Pirimidinas/efectos adversos , Pirimidinas/farmacocinética
7.
Anticancer Drugs ; 32(8): 842-854, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-33929988

RESUMEN

LFZ-4-46, that is [2-hydroxy-1-phenyl-1,5,6,10b-tetrahydropyrazolo(5,1-a) isoquinolin-3(2H)-yl](phenyl) methanone, a tetrahydroisoquinoline derivative with a pyrazolidine moiety, was synthetically prepared. The anti-cancer mechanism of the compound has not been clarified yet. In this study, the anticancer effects and potential mechanisms of LFZ-4-46 on human breast and prostate cancer cells were explored. (a) 3-(4,5-Dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazoliumbromide assay was first performed to detect the effects of LFZ-4-46 on the viability of human cancer cells. (b) Comet assay was utilized to evaluate DNA damage. (c) Cell cycle, apoptosis and mitochondrial membrane potential were detected by flow cytometry. (d) The expression of relative proteins was detected by western blotting assay. LFZ-4-46 significantly inhibited the viability of cancer cells in a time- and dose-dependent manner and had no obviously inhibitory effect on the viability of mammary epithelial MCF-10A cells. Mechanistic studies demonstrated that LFZ-4-46-induced cell apoptosis and cycle arrest were mediated by DNA damage. It caused DNA damage through activating γ-H2AX and breaking DNA strands. Further studies showed that mitogen-activated protein kinasess pathway was involved in these activated several key molecular events. Finally, LFZ-4-46 showed a potent antitumor effect in vivo. These results suggest that LFZ-4-46 may be a potential lead compound for the treatment of breast and prostate cancer.


Asunto(s)
Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Animales , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Masculino , Ratones , Ratones Endogámicos BALB C , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Tetrahidroisoquinolinas , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Anticancer Drugs ; 32(5): 517-525, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33290316

RESUMEN

Phosphatidylinositol-4,5-bisphosphate 3-kinases (PI3Ks) regulate several important cellular and subcellular processes including cell proliferation and differentiation. LY294002 was originally reported to be a selective inhibitor of PI3K-Akt. Later, it showed that this compound also inhibits several other molecules. In this study, we investigated the effect of LY294002 on the growth of suspension (MV4-11 and TF-1a) and tissue (Hep-G2) cells. In exponential phase, MV4-11 cells, but not TF-1a and Hep-G2 cells, expressed a low level of PI3Kp85 and addition of LY294002 inhibited the phosphorylation of PI3Kp85. LY294002 also significantly inhibited the proliferation of MV4-11, TF-1a and Hep-G2 cell and caused formation of cell clusters/aggregates measured by MTT and BrdU assays, and observed under an inverted microscope, respectively. Surprisingly, we found that LY294002 markedly repressed the activation of mitogen-activated protein kinase (MAPK) signal molecules, MEK and ERK, in all these cells. The inhibition of MEK and ERK was confirmed by using MEK stimulators, GM-CSF and phorbol 12-myristate 13-acetate, and MEK-specific inhibitor, PD98059. Although transforming growth factor beta (TGFß) also inhibited the growth of Hep-G2 cells, it had no effect on the activity of MEK and ERK. The clusters/aggregates found in LY294002-treated cells were not detectable in TGFß-treated cells. Our data suggest that LY294002 may directly inhibit the activation of MEK and ERK by its ability to bind to the ATP-binding site of the MAPK molecules.


Asunto(s)
Cromonas/farmacología , Inhibidores Enzimáticos/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Morfolinas/farmacología , Fosfatidilinositol 3-Quinasas/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular , Células Hep G2 , Humanos , Quinasas de Proteína Quinasa Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Transducción de Señal
9.
BMC Anesthesiol ; 21(1): 257, 2021 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-34702181

RESUMEN

BACKGROUND: Angiogenesis, the formation of blood vessel from pre-existing ones, plays an important role in many pathophysiological diseases, such as cancer. Opioids are often used in clinic for the management of chronic pain in cancer patients at terminal phases. Here, we investigated and compared the effects and mechanisms of four opioids on angiogenesis. METHODS: We performed angiogenesis assays on human umbilical vein endothelial cells (HUVEC) that represent an in vitro model to assess the toxicity of drugs to endothelium. RESULTS: Morphine and oxycodone at 0.1 µM to 100 µM dose-dependently increased endothelial cell tube formation and proliferation. We observed the same in endothelial cells exposed to fentanyl at 0.1 µM to 10 µM but there was a gradual loss of stimulation by fentanyl at 100 µM and 1000 µM. Morphine and fentanyl reduced endothelial cell apoptosis-induced by serum withdrawal whereas oxycodone did not display anti-apoptotic effect, via decreasing Bax level. Oxycodone at the same concentrations was less potent than morphine and fentanyl. Different from other three opioids, codeine at all tested concentrations did not affect endothelial cell tube formation, proliferation and survival. Mechanism studies demonstrated that opioids acted on endothelial cells via µ-opioid receptor-independent pathway. Although we observed the increased phosphorylation of mitogen-activated protein kinase (MAPK) in cells exposed to morphine, fentanyl and oxycodone, the rescue studies demonstrated that the stimulatory effects of morphine but not fentanyl nor oxycodone were reversed by a specific MAPK inhibitor. CONCLUSION: Our work demonstrates the differential effects and mechanisms of opioids on angiogenesis.


Asunto(s)
Analgésicos Opioides/farmacología , Células Endoteliales/efectos de los fármacos , Neovascularización Fisiológica/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Fentanilo/farmacología , Humanos , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Morfina/farmacología , Oxicodona/farmacología , Fosforilación , Venas Umbilicales/citología
10.
Int J Mol Sci ; 22(21)2021 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-34768840

RESUMEN

Obesity is a lipid metabolism disorder caused by genetic, medicinal, nutritional, and other environmental factors. It is characterized by a complex condition of excess lipid accumulation in adipocytes. Adipogenesis is a differentiation process that converts preadipocytes into mature adipocytes and contributes to excessive fat deposition. Saikosaponin A (SSA) and saikosaponin D (SSD) are triterpenoid saponins separated from the root of the Bupleurum chinensis, which has long been used to treat inflammation, fever, and liver diseases. However, the effects of these constituents on lipid accumulation and obesity are poorly understood. We investigated the anti-obesity effects of SSA and SSD in mouse 3T3-L1 adipocytes. The MTT assay was performed to measure cell viability, and Oil Red O staining was conducted to determine lipid accumulation. Various adipogenic transcription factors were evaluated at the protein and mRNA levels by Western blot assay and quantitative reverse transcription polymerase chain reaction (qRT-PCR). Here, we showed that SSA and SSD significantly inhibited lipid accumulation without affecting cell viability within the range of the tested concentrations (0.938-15 µM). SSA and SSD also dose-dependently suppressed the expression of peroxisome proliferator-activated receptor gamma (PPARγ), CCAAT/enhancer binding protein alpha (C/EBPα), sterol regulatory element binding protein-1c (SREBP-1c), and adiponectin. Furthermore, the decrease of these transcriptional factors resulted in the repressed expression of several lipogenic genes including fatty acid binding protein (FABP4), fatty acid synthase (FAS), and lipoprotein lipase (LPL). In addition, SSA and SSD enhanced the phosphorylation of adenosine monophosphate-activated protein kinase (AMPK) and its substrate, acetyl-CoA carboxylase (ACC), and inhibited the phosphorylation of extracellular-regulated kinase 1/2 (ERK1/2) and p38, but not c-Jun-N-terminal kinase (JNK). These results suggest that SSA and SSD inhibit adipogenesis through the AMPK or mitogen-activated protein kinase (MAPK) pathways in the early stages of adipocyte differentiation. This is the first study on the anti-adipogenic effects of SSA and SSD, and further research in animals and humans is necessary to confirm the potential of saikosaponins as therapeutic agents for obesity.


Asunto(s)
Adipocitos/metabolismo , Adipogénesis/fisiología , Ácido Oleanólico/análogos & derivados , Saponinas/farmacología , Células 3T3-L1 , Proteínas Quinasas Activadas por AMP/metabolismo , Adenilato Quinasa/efectos de los fármacos , Adenilato Quinasa/metabolismo , Adipogénesis/genética , Adiponectina/metabolismo , Animales , Antiinflamatorios no Esteroideos/farmacología , Fármacos Antiobesidad/farmacología , Bupleurum , Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Diferenciación Celular/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Lipogénesis/fisiología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Obesidad/tratamiento farmacológico , Ácido Oleanólico/farmacología , PPAR gamma/efectos de los fármacos , PPAR gamma/metabolismo , Fosforilación/efectos de los fármacos , Extractos Vegetales/farmacología , Transducción de Señal/fisiología , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo
11.
Molecules ; 26(20)2021 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-34684681

RESUMEN

Bisphenol A (BPA) is a typical environmental endocrine disruptor that exhibits estrogen-mimicking, hormone-like properties and can cause the collapse of bone homeostasis by an imbalance between osteoblasts and osteoclasts. Various BPA substitutes, structurally similar to BPA, have been used to manufacture 'BPA-free' products; however, the regulatory role of BPA alternatives in osteoclast differentiation still remains unelucidated. This study aimed to investigate the effects of these chemicals on osteoclast differentiation using the mouse osteoclast precursor cell line RAW 264.7. Results confirmed that both BPA and its alternatives, bisphenol F and tetramethyl bisphenol F (TMBPF), were nontoxic to RAW 264.7 cells. In particular, tartrate-resistant acid phosphatase (TRAP)-positive multinucleated cell staining and activity calculation assays revealed that TMBPF enhanced osteoclast differentiation upon stimulation of the receptor activator of nuclear factor-kappa B ligand (RANKL). Additionally, TMBPF activated the mRNA expression of osteoclast-related target genes, such as the nuclear factor of activated T-cells, cytoplasmic 1 (NFATc1), tartrate-resistant acid phosphatase (TRAP), and cathepsin K (CtsK). Western blotting analysis indicated activation of the mitogen-activated protein kinase signaling pathway, including phosphorylation of c-Jun N-terminal kinase and p38. Together, the results suggest that TMBPF enhances osteoclast differentiation, and it is critical for bone homeostasis and skeletal health.


Asunto(s)
Compuestos de Bencidrilo/farmacología , Estrógenos no Esteroides/farmacología , Osteoblastos/efectos de los fármacos , Fenoles/farmacología , Animales , Resorción Ósea , Diferenciación Celular/efectos de los fármacos , Estrógenos/análogos & derivados , Estrógenos/farmacología , Macrófagos/efectos de los fármacos , Ratones , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Células RAW 264.7 , Transducción de Señal/efectos de los fármacos
12.
Toxicol Appl Pharmacol ; 391: 114919, 2020 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-32045587

RESUMEN

Ulcerative Colitis is a universal autoimmune disease with high incidence rates worldwide. It is characterized by the existence of many other concurrent immune-associated ailments, including diabetes. The used strategies for the management of this highly costing and complicated disease face great challenges. Therefore, the urge for new medication with fewer side effects and high efficacy is growing. The peroxisome proliferator-activated receptor-gamma (PPARγ) and nuclear factor Kappa-B (NF-κB) can be considered as crucial targets for the treatment of ulcerative colitis. Several studies reported the antioxidants, anti-inflammatory, and antiapoptotic actions of gliclazide and evaluated its cardioprotective and renoprotective effects. However, its impact on ulcerative colitis has never been investigated. This study delineated the effect of gliclazide administration on ulcerative colitis induced by acetic acid in rats and the underlying molecular mechanisms. Gliclazide (10 mg/kg; p.o) prominently decreased colon tissue injury as assessed by the histopathological analysis as well as myeloperoxidase, and intercellular adhesion molecule-1 levels. Gliclazide significantly alleviated the proinflammatory mediator, IL-6, promoted the anti-inflammatory cytokine, IL-10 and, withheld oxidative stress in the injured colon tissues. The protective effect of gliclazide was mediated through the upregulation of PPARγ and downregulation of NF-κB expression. The diminution of ulcerative colitis was also accompanied by an inhibition of the elevated activity and expression of mitogen-activated protein kinases and caspase-3 as assessed by Western blot and immunohistochemistry, respectively. Our findings spotlight, for the first time, the potential of the antidiabetic agent, gliclazide, to attenuate the experimentally induced ulcerative colitis. Therefore, gliclazide might be a propitious agent for the management of ulcerative colitis in diabetic patients.


Asunto(s)
Colitis Ulcerosa/inducido químicamente , Colitis Ulcerosa/tratamiento farmacológico , Gliclazida/uso terapéutico , Transducción de Señal/efectos de los fármacos , Ácido Acético , Animales , Apoptosis/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Caspasa 3/metabolismo , Inhibidores de Caspasas/farmacología , Colitis Ulcerosa/patología , Colon/patología , Regulación hacia Abajo/efectos de los fármacos , Molécula 1 de Adhesión Intercelular/biosíntesis , Molécula 1 de Adhesión Intercelular/genética , Masculino , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , FN-kappa B/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , PPAR gamma/efectos de los fármacos , Peroxidasa/biosíntesis , Peroxidasa/genética , Ratas , Ratas Wistar
13.
Cell Mol Neurobiol ; 40(7): 1117-1131, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31989355

RESUMEN

Chronic morphine-induced antinociceptive tolerance is a major unresolved issue in clinical practices, which is associated with microglia activation in the spinal cord. E3 ubiquitin ligase Pellino1 (Peli1) is known to be an important microglia-specific regulator. However, it is unclear whether Peli1 is involved in morphine tolerance. Here, we found that Peli1 levels in the spinal cord were significantly elevated in morphine tolerance mouse model. Notably, Peli1 was expressed in a great majority of microglia in the spinal dorsal horn, while downregulation of spinal Peli1 attenuated the development of morphine tolerance and associated hyperalgesia. Our biochemical data revealed that morphine tolerance-induced increase in Peli1 was accompanied by spinal microglia activation, activation of mitogen-activated protein kinase (MAPK) signaling, and production of proinflammatory cytokines. Peli1 additionally was found to promote K63-linked ubiquitination of tumor necrosis factor receptor-associated factor 6 (TRAF6) in the spinal cord after repeated morphine treatment. Furthermore, knocking down Peli1 in cultured BV2 microglial cells significantly attenuated inflammatory reactions in response to morphine challenge. Therefore, we conclude that the upregulation of Peli1 in the spinal cord plays a curial role in the development of morphine tolerance via Peli1-dependent mobilization of spinal microglia, activation of MAPK signaling, and production of proinflammatory cytokines. Modulation of Peli1 may be a potential strategy for the prevention of morphine tolerance.


Asunto(s)
Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Microglía/metabolismo , Morfina/farmacología , Proteínas Nucleares/metabolismo , Médula Espinal/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Tolerancia a Medicamentos/fisiología , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Activación de Macrófagos/efectos de los fármacos , Masculino , Ratones , Microglía/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Médula Espinal/efectos de los fármacos
14.
Inflamm Res ; 69(2): 233-244, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31907559

RESUMEN

OBJECTIVE: Recently, Rodgersia podophylla has been reported to exhibit anti-inflammatory activity. However, little is known about the potential mechanisms about its anti-inflammatory activity. We elucidated the anti-inflammatory mechanisms of leaves extracts from Rodgersia podophylla (RP-L) in RAW264.7 cells. MATERIALS AND METHODS: LPS-induced NO was measured by Griess and mRNA of pro-inflammatory mediators was analyzed by RT-PCR. Cell viability was measured using MTT assay. The protein level was analyzed by Western blot. RESULTS: RP-L significantly inhibited the production of the pro-inflammatory mediators such as NO, iNOS, IL-1ß and IL-6 in LPS-stimulated RAW264.7 cells. RP-L increased HO-1 expression in RAW264.7 cells, and the inhibition of HO-1 by ZnPP reduced the inhibitory effect of RP-L against LPS-induced NO production in RAW264.7 cells. Inhibition of p38, ROS and GSK3ß attenuated RP-L-mediated HO-1 expression. Inhibition of ROS inhibited p38 phosphorylation and GSK3ß expression induced by RP-L. In addition, inhibition of GSK3ß blocked RP-L-mediated p38 phosphorylation. RP-L induced nuclear accumulation of Nrf2, and inhibition of p38, ROS and GSK3ß abolished RP-L-mediated nuclear accumulation of Nrf2. Furthermore, RP-L blocked LPS-induced degradation of IκB-α and nuclear accumulation of p65. RP-L also attenuated LPS-induced phosphorylation of ERK1/2 and p38. In GC/MS analysis of RP-L, pyrogallol was detected as bioactive compound for anti-inflammatory activity of RP-L. Pyrogallol was observed to activate HO-1 expression through ROS/GSK3ß/p38/Nrf2/HO-1 signaling. CONCLUSIONS: Our results suggest that RP-L exerts potential anti-inflammatory activity by activating ROS/GSK3ß/p38/Nrf2/HO-1 signaling and inhibiting NF-κB and MAPK signaling in RAW264.7 cells. These findings suggest that RP-L may have great potential for the development of anti-inflammatory drug.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Hemo-Oxigenasa 1/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Factor 2 Relacionado con NF-E2/metabolismo , FN-kappa B/efectos de los fármacos , Extractos Vegetales/farmacología , Saxifragaceae/química , Transducción de Señal/efectos de los fármacos , Animales , Ratones , Óxido Nítrico/biosíntesis , Hojas de la Planta/química , Células RAW 264.7
15.
Neurol Sci ; 41(4): 763-768, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31838631

RESUMEN

Vanadium, a transition metal, ubiquitous in nature is known to have therapeutic effect as well as toxic effect. It is known to possess antidiabetic, antitumor and antiparasitic activity. However, on long term exposure, it produces neurotoxicity which may result in memory impairment. The possible mechanism known to cause neurotoxicity suggested is oxidative stress and inflammation of neuronal cells. The present review has focused on discussing the role of protein P38 mitogen-activated protein kinase and oxidative stress as possible targets to treat vanadium-induced neurotoxicity.


Asunto(s)
Inflamación/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Síndromes de Neurotoxicidad/metabolismo , Estrés Oxidativo/fisiología , Inhibidores de Proteínas Quinasas/uso terapéutico , Oligoelementos/toxicidad , Vanadio/toxicidad , Animales , Humanos , Inflamación/tratamiento farmacológico , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Síndromes de Neurotoxicidad/tratamiento farmacológico , Estrés Oxidativo/efectos de los fármacos
16.
J Toxicol Environ Health A ; 83(9): 341-350, 2020 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-32340567

RESUMEN

Ambient particulate matter (PM) is associated with adverse health consequences. However, the influence of PM on the innate immune system is poorly understood. The aim of the present study was to examine the effect of diesel particulate matter 2.5 µm (PM2.5, SRM1650b) on dendritic cells. PM2.5 significantly reduced cytokine levels of interleukin (IL)-12 p40, IL-6 and TNF-α levels in CpG-DNA (TLR9 ligand)-stimulated dendritic cells. To determine the mechanisms underlying this observed inhibition induced by PM2.5, western blot analysis was conducted. PM2.5 was found to downregulate ERK1/2, JNK1/2, p38 MAPKs, and NF-κB pathways. PM2.5 exposure decreased TLR9-dependent NF-κB and activator protein (AP-1) reporter luciferase activities. Our findings demonstrate that PM2.5 reduced the production of cytokines which may be associated with inhibition of MAPK and NF-κB signaling pathway. Further, data suggest the immunosuppressive effect of PM2.5 on the innate immune cells may lead to serious damage to the host immune system.


Asunto(s)
Citocinas/efectos de los fármacos , Células Dendríticas/efectos de los fármacos , Gasolina/efectos adversos , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , FN-kappa B/efectos de los fármacos , Material Particulado/efectos adversos , Receptor Toll-Like 9/efectos de los fármacos , Animales , Citocinas/metabolismo , Femenino , Humanos , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Modelos Animales , FN-kappa B/metabolismo , Transducción de Señal/efectos de los fármacos
17.
World J Microbiol Biotechnol ; 36(5): 74, 2020 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-32388765

RESUMEN

Probiotics are known to modulate gut microbiota, intestinal barrier function and host immune response, but due to the species and strain specific response their mechanisms are not clearly understood. Thus, the present study was designed to isolate, assess the anti-inflammatory potential and underlying modulatory mechanisms of indigenous probiotics in murine macrophage cell line, RAW 264.7. Forty lactic acid bacteria (LAB) were isolated from different sources and monitored for their anti-inflammatory potential against lipopolysaccharide (LPS) induced inflammatory stress employing RAW 264.7 cells. Among these isolates, only four LAB isolates exhibited more than 90% nitric oxide inhibition and possessed the probiotic attributes. Further, these selected LAB isolates reduced the level of pro-inflammatory cytokines, TNF-α, IL-1ß and IL-6, inhibited the phosphorylation of Mitogen Activated Protein Kinases (MAPKs) i.e. p38 MAPK, ERK1/2 and SAPK/JNK and expression of cyclooxygenase-2 (COX-2) in LPS stimulated RAW 264.7 cells. The in vitro analysis suggested that the selected probiotic isolates attenuated the LPS-induced inflammation by downregulating MAPK pathway vis-a-vis inhibiting COX-2 and can be employed as anti-inflammatory agents in various inflammatory diseases.


Asunto(s)
Antiinflamatorios/aislamiento & purificación , Antiinflamatorios/farmacología , Lipopolisacáridos/efectos adversos , Macrófagos/efectos de los fármacos , Probióticos/aislamiento & purificación , Probióticos/farmacología , Animales , Supervivencia Celular/efectos de los fármacos , Ciclooxigenasa 2/efectos de los fármacos , Citocinas/metabolismo , Inflamación/tratamiento farmacológico , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Macrófagos/inmunología , Ratones , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Óxido Nítrico/metabolismo , Fosforilación , Células RAW 264.7/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo
18.
J Cell Physiol ; 234(5): 6463-6476, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30246289

RESUMEN

Acute lung injury (ALI) and its severe form acute respiratory distress syndrome (ARDS) remain a major cause of morbidity and mortality in critically ill patients, and no specific therapies are still available to control the mortality rate. Thus, we explored the preventive and therapeutic effects of tannic acid (TA), a natural polyphenol in the context of ALI. We used in vivo and in vitro models, respectively, using lipopolysaccharide (LPS) to induce ALI in mice and exposing J774 and BEAS-2B cells to LPS. In both preventive and therapeutic approaches, TA attenuated LPS-induced histopathological alterations, lipid peroxidation, lung permeability, infiltration of inflammatory cells, and the expression of proinflammatory mediators. In addition, in-vitro study showed that TA treatment could reduce the expression of proinflammatory mediators. Further studies revealed that TA-dampened inflammatory responses by downregulating the LPS-induced toll-like receptor 4 (TLR4) expression and inhibiting extracellular-signal-regulated kinase (ERK)1/2 and p38 mitogen-activated protein kinase (MAPK) activation. Furthermore, cells treated with the inhibitors of ERK1/2 (PD98059) and p38 (SB203580) mitigated the expression of cytokines induced by LPS, thus suggesting that ERK1/2 and p38 activity are required for the inflammatory response. In conclusion, TA could attenuate LPS-induced inflammation and may be a potential therapeutic agent for ALI-associated inflammation in clinical settings.


Asunto(s)
Lesión Pulmonar Aguda/patología , Proteínas Quinasas Activadas por Mitógenos/biosíntesis , Taninos/farmacología , Receptor Toll-Like 4/biosíntesis , Lesión Pulmonar Aguda/metabolismo , Animales , Modelos Animales de Enfermedad , Regulación hacia Abajo , Masculino , Ratones , Ratones Endogámicos BALB C , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Receptor Toll-Like 4/efectos de los fármacos
19.
Am J Physiol Gastrointest Liver Physiol ; 316(2): G229-G246, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30406698

RESUMEN

Whether zinc (Zn2+) regulates barrier functions by modulating tight-junction (TJ) proteins when pathogens such as Shigella alter epithelial permeability is still unresolved. We investigated the potential benefits of Zn2+ in restoring impaired barrier function in vivo in Shigella-infected mouse tissue and in vitro in T84 cell monolayers. Basolateral Shigella infection triggered a time-dependent decrease in transepithelial resistance followed by an increase in paracellular permeability of FITC-labeled dextran and altered ion selectivity. This led to ion and water loss into the intestinal lumen. Immunofluorescence studies revealed redistribution of claudin-2 and -4 to an intracellular location and accumulation of these proteins in the cytoplasm following infection. Zn2+ ameliorated this perturbed barrier by redistribution of claudin-2 and -4 back to the plasma membrane and by modulating the phosphorylation state of TJ proteins t hough extracellular signal-regulated kinase (ERK)1/2 dependency. Zn2+ prevents elevation of IL-6 and IL-8. Mice challenged with Shigella showed that oral Zn2+supplementation diminished diverse pathophysiological symptoms of shigellosis. Claudin-2 and -4 were susceptible to Shigella infection, resulting in altered barrier function and increased levels of IL-6 and IL-8. Zn2+ supplementation ameliorated this barrier dysfunction, and the inflammatory response involving ERK-mediated change of phosphorylation status for claudin-2 and -4. Thus, Zn2+ may have potential therapeutic value in inflammatory diarrhea and shigellosis. NEW & NOTEWORTHY Our study addresses whether Zn2+ could be an alternative strategy to reduce Shigella-induced inflammatory response and epithelial barrier dysfunction. We have defined a mechanism in terms of intracellular signaling pathways and tight-junction protein expression by Zn2+. Claudin-2 and -4 are susceptible to Shigella infection, whereas in the presence of Zn2+ they are resistant to infection-related barrier dysfunction involving ERK-mediated change of phosphorylation status of claudins.


Asunto(s)
Claudina-2/metabolismo , Claudina-4/metabolismo , Permeabilidad/efectos de los fármacos , Zinc/farmacología , Animales , Claudina-2/efectos de los fármacos , Claudina-4/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Enfermedades Intestinales/tratamiento farmacológico , Enfermedades Intestinales/metabolismo , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Proteína Quinasa 3 Activada por Mitógenos/efectos de los fármacos , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/metabolismo , Zinc/metabolismo
20.
J Neuroinflammation ; 16(1): 24, 2019 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-30709405

RESUMEN

BACKGROUND: Previous studies had showed that Apelin 13 could protect against apoptosis induced by ischemic/reperfusion (I/R). However, the mechanisms whereby Apelin 13 protected brain I/R remained to be elucidated. The present study was designed to determine whether Apelin 13 provided protection through AMPK/GSK-3ß/Nrf2 pathway. METHODS: In vivo, the I/R model was induced and Apelin 13 was given intracerebroventricularly 15 min before reperfusion. The neurobehavioral scores, infarction volumes, and some cytokines in the brain were measured. For in vitro study, PC12 cells were used. To clarify the mechanisms, proteases inhibitors or siRNA were used. Protein levels were investigated by western blotting. RESULTS: The results showed that Apelin 13 treatment significantly reduced infarct size, improved neurological outcomes, decreased brain edema, and inhibited cell apoptosis, oxidative stress, and neuroinflammation after I/R. Apelin 13 significantly increased the expression of Nrf2 and the phosphorylation levels of AMPK and GSK-3ß. Furthermore, in cultured PC12 cells, the same protective effects were also observed. Silencing Nrf2 gene with its siRNA abolished the Apelin 13's prevention of I/R-induced PC12 cell injury, oxidative stress, and inflammation. Inhibition of AMPK by its siRNA decreased the level of Apelin 13-induced Nrf2 expression and diminished the protective effects of Apelin 13. The interplay relationship between GSK-3ß and Nrf2 was also verified with relative overexpression. Using selective inhibitors, we further identified the upstream of AMPK/GSK-3ß/Nrf2 is AR/Gα/PLC/IP3/CaMKK. CONCLUSIONS: In conclusion, the previous results showed that Apelin 13 protected against I/R-induced ROS-mediated inflammation and oxidative stress through activating the AMPK/GSK-3ß pathway by AR/Gα/PLC/IP3/CaMKK signaling, and further upregulated the expression of Nrf2-regulated antioxidant enzymes.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Glucógeno Sintasa Quinasa 3 beta/efectos de los fármacos , Péptidos y Proteínas de Señalización Intercelular/farmacología , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Factor 2 Relacionado con NF-E2/agonistas , Fármacos Neuroprotectores/farmacología , Accidente Cerebrovascular/tratamiento farmacológico , Animales , Infarto de la Arteria Cerebral Media/patología , Infarto de la Arteria Cerebral Media/prevención & control , Inyecciones Intraventriculares , Péptidos y Proteínas de Señalización Intercelular/administración & dosificación , Masculino , Fármacos Neuroprotectores/administración & dosificación , Estrés Oxidativo/efectos de los fármacos , Células PC12 , ARN Interferente Pequeño/farmacología , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda