Your browser doesn't support javascript.
loading
Mostrar: 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.
Life Sci ; 338: 122406, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38176583

RESUMEN

AIMS: Curcumin is a natural compound and has good antitumor properties, but its clinical use is limited by its low bioavailability. We constructed the derivative CP41 (3,5-bis(2-chlorobenzylidene)-1-piperidin-4-one) by enhancing the bioavailability of curcumin while retaining its antitumor properties. MAIN METHODS: CCK-8 (Cell Counting Kit-8) was used to detect the effect of CP41 on cell proliferation; Western blotting, immunofluorescence, immunoprecipitation, quantitative PCR and enzyme-linked immunosorbent assay were used to evaluate the expression of subcutaneous tumor-related molecules in cells and mice. KEY FINDINGS: Our results showed that CP41 inhibited the proliferation of endometrial cancer cells by suppressing the proliferation of AN3CA and HEC-1-B cells. We found that CP41 significantly increased H3F3A and inhibited proteasome activity, which activated MAPK signaling and led to apoptosis. Further experiments showed that H3F3A is a potential target of CP41. Correlation analysis showed that H3F3A was positively correlated with the sensitivity to chemotherapeutic agents in endometrial cancer. CP41 significantly induced reactive oxygen species (ROS) levels and activated endoplasmic reticulum stress, which led to apoptosis. The safety profile of CP41 was also evaluated, and CP41 did not cause significant drug toxicity in mice. SIGNIFICANCE: CP41 showed stronger antitumor potency than curcumin, and its antitumor activity may be achieved by inducing ROS and activating H3F3A-mediated apoptosis.


Asunto(s)
Curcumina , Neoplasias Endometriales , Animales , Femenino , Humanos , Ratones , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular , Curcumina/análogos & derivados , Curcumina/farmacología , Neoplasias Endometriales/tratamiento farmacológico , Estrés del Retículo Endoplásmico , Estrés Oxidativo/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Piperidinas/farmacología , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo
3.
Toxicol In Vitro ; 93: 105667, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37625625

RESUMEN

BACKGROUND: Breast cancer is the highest incidence of all types of cancer in women, and the cancer metastasis process accounts for a majority of cancer deaths. Two major cannabinoids, Δ-9-tetrahydrocannabinol (THC) and cannabidiol (CBD), from Cannabis sativa are expected to have anti-cancer activity. This study aimed to investigate the effects of THC, CBD, and standardized cannabis extracts (F1, F2, and F3) on migration, invasion, and apoptosis of human breast cancer (MCF-7) cells. METHODS: Cell viability, survival, and apoptosis were determined using the MTT, clonogenic, and nuclear staining assays, respectively, while cancer cell migration and invasion were evaluated by the wound healing, trans-well, and filopodia assays. Western blot analysis was used to find out the mechanisms of the cannabinoids against MCF-7 cells. RESULTS: CBD, THC, and F1 inhibited filopodia formation, migration, and invasion of MCF-7 cells through suppressing the expression of the FAK, Akt, ERK1/2, p38MAPKs, and NF-κB upstream pathways, as well as inhibiting the Rac1/Cdc42 downstream pathways. In addition, CBD significantly inhibited the mTOR pathway. Furthermore, CBD and F1 induced apoptosis in MCF-7 cells via the Bcl-2/caspase-3 pathways. CONCLUSION: These results indicate that THC, CBD, and F1 have great abilities for preventing breast cancer cell metastasis in in vitro experiments.


Asunto(s)
Neoplasias de la Mama , Cannabidiol , Cannabinoides , Cannabis , Femenino , Humanos , Apoptosis , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Cannabidiol/farmacología , Cannabinoides/farmacología , Cannabis/metabolismo , Células MCF-7 , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas c-akt , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo
4.
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
5.
J Ethnopharmacol ; 296: 115489, 2022 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-35728711

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Aesculetin (6,7-dihydroxy-2H-1-benzopyran-2-one) has been reported to exhibit potent anti-inflammatory property both in vitro and in vivo. AIMS OF THIS STUDY: In this study, we evaluated the anti-inflammatory effect and investigated underlying molecular mechanisms of aesculetin in LPS-induced RAW264.7 macrophages and DSS-induced colitis. MATERIALS AND METHODS: In this study, the production of NO, TNF-α, and IL-6 were measured to identify the aesculetin with potent anti-inflammatory effect. Then, the underlying anti-inflammatory mechanisms were explored by western blotting in LPS-induced cells. Next, we verify the anti-inflammatory potential of aesculetin in DSS-induced colitis in vivo. The clinical symptoms of colitis, including weight loss, DAI, colon length and MPO activity, and the secretion of TNF-α and IL-6 were evaluated. Finally, Western blot analysis was applied to further investigate underlying mechanism in DSS-induced colitis model. RESULTS: Our studies showed that aesculetin exhibited anti-inflammatory potential by inhibiting NO, TNF-α, and IL-6 production and reducing iNOS and NLRP3 expression in LPS-induced RAW264.7 cells. Mechanically, we found that aesculetin significantly inhibited LPS-induced activation of NF-κB and MAPKs signaling pathways. In DSS-induced mouse model, the colitis-related symptoms were relieved by treatment with aesculetin. Besides, aesculetin also inhibited the secretion of TNF-α and IL-6, and the activation of NF-κB and MAPKs signaling pathways in DSS-induced colitis. CONCLUSIONS: The anti-inflammatory effect of aesculetin was connected with its inhibition on the activation of NF-κB and MAPKs signaling pathways both in vitro and in vivo. Therefore, aesculetin was expected to be developed as an anti-inflammatory drug.


Asunto(s)
Colitis , FN-kappa B , Umbeliferonas , Animales , Antiinflamatorios/efectos adversos , Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Citocinas , Sulfato de Dextran , Interleucina-6 , Lipopolisacáridos , Ratones , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Umbeliferonas/farmacología , Umbeliferonas/uso terapéutico
6.
Biomed Pharmacother ; 147: 112701, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35131657

RESUMEN

Sustained usage of the chemotherapeutic drug cisplatin may lead to chronic kidney disease (CKD). Despite cisplatin being toxic to the kidneys, the efficiency of its therapeutic effects cannot be completely replaced with other drugs. Probiotics can produce various strain-specific health-promoting effects and suppress many specific diseases. In this study, we present the alleviation of cisplatin-induced CKD with a probiotic, Lactobacillus rhamnosus GKLC1. Intermittent low doses of cisplatin were given to male CB57BL/6 mice (n = 6), which induced CKD symptoms such as weight loss, lesions in kidney tissue, and increases in blood urea nitrogen (BUN) and creatinine (CRE) in serum. The rats received two weeks of L. rhamnosus GKLC1 orally at doses of 125, 250, and 500 mg/kg B.W./day. After the treatment, significant dose-dependent reductions were observed in the kidney index, histopathological scoring, serum BUN, and CRE. An LLC-PK1 kidney cell assay revealed that L. rhamnosus GKLC1 suppressed the nephrotoxicity of cisplatin by reducing the inflammation via the MAPKs/NF-ĸB/COX-2 pathway, inhibiting apoptosis via the p53/Bax/Caspase-3 pathway, and ameliorating fibrosis via the STAT3 pathway. We conclude that L. rhamnosus GKLC1 could be applied as an agent to ameliorate the development of CKD.


Asunto(s)
Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/patología , Antineoplásicos/efectos adversos , Cisplatino/efectos adversos , Lacticaseibacillus rhamnosus , Probióticos/farmacología , Animales , Apoptosis/efectos de los fármacos , Nitrógeno de la Urea Sanguínea , Caspasa 3/efectos de los fármacos , Creatinina/sangre , Relación Dosis-Respuesta a Droga , Inflamación/patología , Mediadores de Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Probióticos/administración & dosificación , Ratas , Transducción de Señal/efectos de los fármacos , Proteína X Asociada a bcl-2/efectos de los fármacos
7.
Biomed Pharmacother ; 147: 112651, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35063859

RESUMEN

Abnormal melanin synthesis upon UV exposure causes excessive oxidative stress, which leads to skin hyperpigmentation disorders such as freckles, melisma, and age spots. The present study investigated the anti-melanogenic effects of decursin and the underlying mechanism using multiple approaches. Decursin exhibited no cytotoxicity and significantly reduced intracellular tyrosinase activity and melanin content in B16F10 melanoma cells. Decursin also inhibited the expression of melanogenic enzymes such as tyrosinase and tyrosinase-related protein (TRP)- 1, but not TRP-2. Mechanistically, decursin suppressed melanin synthesis through cAMP-dependent protein kinase (PKA)/cAMP response element-binding protein (CREB)-dependent downregulation of microphthalmia-associated transcription factor (MITF), a master transcription factor in melanogenesis. Further, decursin exerted anti-melanogenic effects by downregulating the p38 signaling pathway and upregulating extracellular signal-regulated kinase (ERK) and phosphatidylinositol 3 kinase (PI3K)/protein kinase B (Akt)/glycogen synthesis kinase-3ß (GSK-3ß) cascades. in silico analysis showed that decursin formed specific interactions with residues of upstream regulators of MITF and exhibited optimal pharmacokinetic profiles, including permeability and skin sensitization. Finally, the anti-melanogenic effects of decursin were confirmed ex vivo in 3D human skin models, suggesting its applicability as a protective agent against hyperpigmentation.


Asunto(s)
Benzopiranos/farmacología , Butiratos/farmacología , Melaninas/metabolismo , Factor de Transcripción Asociado a Microftalmía/efectos de los fármacos , Proteína de Unión a CREB/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Proteínas Quinasas Dependientes de AMP Cíclico/efectos de los fármacos , Glucógeno Sintasa Quinasa 3 beta/efectos de los fármacos , Humanos , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Piel/efectos de los fármacos
8.
Biomed Pharmacother ; 147: 112640, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35033946

RESUMEN

Pyrrosia lingua (Thunb.) Farw is a common plant that has been widely used as a traditional herbal medicine in China and Korea to treat patients suffering from pain, vaginal bleeding and urolithiasis. However, the pharmacological effects of P. lingua on bone remain unknown. We investigated the anti-osteoporotic effects of an ethanolic extract of P. lingua (EEPL). We found that EEPL suppressed osteoclast differentiation by directly acting on osteoclast precursor cells. EEPL suppressed the expression of receptor activator of nuclear factor-κB ligand (RANKL)-induced nuclear factor of activated T cells 1, a major transcription factor for osteoclastogenesis, by inhibiting RANKL-induced expression of aryl hydrocarbon receptor/c-Fos, and activation of nuclear factor-κB and mitogen-activated protein kinases. Moreover, administration of EEPL inhibited trabecular bone loss and weight gain in ovariectomized mice. Furthermore, we identified phytochemicals in EEPL that are known to exert anti-osteoclastogenic or anti-osteoporotic effects using ultra-high-performance liquid chromatography-tandem mass-spectrometry analysis. Overall, the results of this study suggest that EEPL is effective therapeutic candidate that can be used to prevent or treat postmenopausal osteoporosis.


Asunto(s)
Osteoclastos/efectos de los fármacos , Extractos Vegetales/farmacología , Polypodiaceae , Ligando RANK/efectos de los fármacos , Animales , Hueso Esponjoso/efectos de los fármacos , Línea Celular , Modelos Animales de Enfermedad , Femenino , Ratones , Ratones Endogámicos C57BL , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , FN-kappa B/efectos de los fármacos , Osteoporosis/patología , Ovariectomía , Proteínas Proto-Oncogénicas c-fos/efectos de los fármacos , Receptores de Hidrocarburo de Aril/efectos de los fármacos , Aumento de Peso/efectos de los fármacos
9.
Bioengineered ; 13(2): 2851-2865, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35037840

RESUMEN

HBV (hepatitis B virus) infection still threatens human health. Therefore, it is essential to find new effective anti-HBV compounds. Here, we identified matrine as a novel inhibitor of PKC (protein kinase C) phosphorylated kinase by screening a natural compound library. After HepG2.215 cells were treated with matrine, we carried out a phosphorylated proteomics sequence study and analyzed the prediction of related kinase expression level. In the case of HBV infection, it was found that PKC kinase mediates the activation of mitogen-activated protein kinase (MAPK) signaling pathway known as son of sevenless (SOS) activation. It was also found that PKC kinase inhibits the expression of C-X-C Motif Chemokine Ligand 8 (CXCL8) by inhibiting the activity of activating transcription factor 2/ cAMP response element binding protein (ATF2/CREB), and this effect is independent of its activated MAPK signaling pathway. Finally, Western blot was used to detect the expression of MAPK, ATF2, CREB3 phosphorylation and nonphosphorylation in matrine-treated cells and PKC-treated cells. PKC phosphorylated kinase inhibitor-matrine suppresses the replication of HBV via modulating the MAPK/ATF2 signal. Matrine is a good clinical drug to enhance the autoimmunity in the adjuvant treatment of chronic HBV infection.


Asunto(s)
Alcaloides/farmacología , Virus de la Hepatitis B/efectos de los fármacos , Quinolizinas/farmacología , Replicación Viral/efectos de los fármacos , Alcaloides/uso terapéutico , Células Hep G2 , Hepatitis B/tratamiento farmacológico , Hepatitis B/metabolismo , Virus de la Hepatitis B/fisiología , Humanos , 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 , Fosfoproteínas/efectos de los fármacos , Fosfoproteínas/metabolismo , Fosforilación/efectos de los fármacos , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/metabolismo , Proteoma/efectos de los fármacos , Proteoma/metabolismo , Quinolizinas/uso terapéutico , Transducción de Señal/efectos de los fármacos , Matrinas
10.
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
11.
Biomed Pharmacother ; 146: 112547, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34929579

RESUMEN

Prolonged exposure to polycyclic aromatic hydrocarbons (PAHs) may result in autoimmune diseases, such as rheumatoid arthritis (RA) and osteoporosis (OP), which are based on an imbalance in bone homeostasis. These diseases are characterized by bone erosion and even a disruption in homeostasis, including in osteoblasts and osteoclasts. Current evidence indicates that multiple factors affect the progression of bone homeostasis, such as genetic susceptibility and epigenetic modifications. However, environmental factors, especially PAHs from various sources, have been shown to play an increasingly prominent role in the progression of bone homeostasis. Hence, it is essential to investigate the effects and pathogenesis of PAHs in bone homeostasis. In this review, recent progress is summarized concerning the effects and mechanisms of PAHs and their ligands and receptors in bone homeostasis. Moreover, strategies based on the effects and mechanisms of PAHs in the regulation of the bone balance and alleviation of bone destruction are also reviewed. We further discuss the future challenges and perspectives regarding the roles of PAHs in autoimmune diseases based on bone homeostasis.


Asunto(s)
Huesos/efectos de los fármacos , Exposición a Riesgos Ambientales/efectos adversos , Contaminantes Ambientales/toxicidad , Homeostasis/efectos de los fármacos , Hidrocarburos Policíclicos Aromáticos/toxicidad , Exposición a Riesgos Ambientales/análisis , Contaminantes Ambientales/análisis , Humanos , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , FN-kappa B/efectos de los fármacos , Proteína Oncogénica v-akt/efectos de los fármacos , Osteoblastos/efectos de los fármacos , Osteoclastos/efectos de los fármacos , Osteoporosis/patología , Fosfatidilinositol 3-Quinasas/efectos de los fármacos , Hidrocarburos Policíclicos Aromáticos/análisis , Transducción de Señal/efectos de los fármacos
12.
Drug Deliv ; 28(1): 2480-2494, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34766543

RESUMEN

Cisplatin (DDP) is a first-line chemotherapeutic drug applied for the treatment of oral squamous cell carcinoma (OSCC). The anticancer activity of DDP is tightly linked to its intracellular uptake. It is unwise to increase the DDP intake by increasing the dose or shortening the dosing interval because of the severe systemic toxicity (nephrotoxicity, ototoxicity and neurotoxicity) in DDP application. The main uptake pathways of DDP include passive diffusion and active transporter transport. Therefore, finding additional uptake pathways that can improve the effective intracellular concentration of DDP is critical. Macropinocytosis, an endocytic mechanism for extracellular material absorption, contributes to the intracellular uptake of anticancer drugs. No research has been conducted to determine whether macropinocytosis can augment the intracellular uptake of DDP in OSCC cells or not. Based on that, we proved for the first time that silmitasertib (previously CX-4945) could trigger macropinocytosis, which may increase the intracellular uptake of DDP and enhance apoptosis via in vivo and in vitro experiments. We hope that our findings will inspire a new approach for the application of DDP in cancer treatment.


Asunto(s)
Antineoplásicos/farmacocinética , Naftiridinas/farmacología , Fenazinas/farmacología , Pinocitosis/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Carcinoma de Células Escamosas/patología , Caspasas/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cisplatino/farmacocinética , Liberación de Fármacos , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Neoplasias de la Boca/patología , Ensayos Antitumor por Modelo de Xenoinjerto
13.
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
14.
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
15.
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
16.
J Antibiot (Tokyo) ; 74(11): 807-816, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34408288

RESUMEN

Berberine hydrochloride (BH), an active component of Coptis chinensis and other plant taxa, has broad antimicrobial activity and may be useful for the treatment of Candida infections. In this study, the mechanisms underlying the inhibitory effect of BH against Candida albicans were evaluated, with a focus on the high-osmolarity glycerol mitogen-activated protein kinase (HOG-MAPK) pathway, which regulates multiple physiological functions. BH (256 and 64 µg ml-1) significantly increased intracellular glycerol and ROS levels in C. albicans, inhibited germ tube and hyphal formation, and increased chitin and ß-1,3-glucan exposure on the cell wall. The inhibitory effect of BH was positively correlated with its concentration, and the inhibitory effect of 256 µg ml-1 BH was greater than that of 4 µg ml-1 fluconazole (FLC). Furthermore, RT-PCR analysis showed that 256 and 64 µg ml-1 BH altered the HOG-MAPK pathway in C. albicans. In particular, the upregulation of the core genes, SLN1, SSK2, HOG1, and PBS2 may affect the expression of key downstream factors related to glycerol synthesis and osmotic pressure (GPD1), ROS accumulation (ATP11 and SOD2), germ tube and hyphal formation (HWP1), and cell wall integrity (CHS3 and GSC1). BH affects multiple biological processes in C. albicans; thus, it can be an effective alternative to conventional azole antifungal agents.


Asunto(s)
Antifúngicos/farmacología , Berberina/farmacología , Candida albicans/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Adenosina Trifosfato/metabolismo , Candida albicans/genética , Fluconazol/farmacología , Genes Fúngicos/efectos de los fármacos , Glucano 1,3-beta-Glucosidasa/efectos de los fármacos , Glicerol/metabolismo , Hifa/efectos de los fármacos , Pruebas de Sensibilidad Microbiana , Especies Reactivas de Oxígeno/metabolismo
17.
Biomolecules ; 11(6)2021 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-34208655

RESUMEN

The RTK-RAS-MAPK axis is one of the most extensively studied signaling cascades and is related to the development of both cancers and RASopathies. In the last 30 years, many ideas and approaches have emerged for directly targeting constituent members of this cascade, predominantly in the context of cancer treatment. These approaches are still insufficient due to undesirable drug toxicity, resistance, and low efficacy. Significant advances have been made in understanding the spatiotemporal features of the constituent members of the RTK-RAS-MAPK axis, which are linked and modulated by many accessory proteins. Given that the majority of such modulators are now emerging as attractive therapeutic targets, a very small number of accessory inhibitors have yet to be discovered.


Asunto(s)
Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Proteínas Tirosina Quinasas Receptoras/efectos de los fármacos , Proteínas ras/efectos de los fármacos , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas Receptoras/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas ras/metabolismo
18.
Front Immunol ; 12: 683879, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34135911

RESUMEN

Diseases caused by pathogenic bacteria in animals (e.g., bacterial pneumonia, meningitis and sepsis) and plants (e.g., bacterial wilt, angular spot and canker) lead to high prevalence and mortality, and decomposition of plant leaves, respectively. Melatonin, an endogenous molecule, is highly pleiotropic, and accumulating evidence supports the notion that melatonin's actions in bacterial infection deserve particular attention. Here, we summarize the antibacterial effects of melatonin in vitro, in animals as well as plants, and discuss the potential mechanisms. Melatonin exerts antibacterial activities not only on classic gram-negative and -positive bacteria, but also on members of other bacterial groups, such as Mycobacterium tuberculosis. Protective actions against bacterial infections can occur at different levels. Direct actions of melatonin may occur only at very high concentrations, which is at the borderline of practical applicability. However, various indirect functions comprise activation of hosts' defense mechanisms or, in sepsis, attenuation of bacterially induced inflammation. In plants, its antibacterial functions involve the mitogen-activated protein kinase (MAPK) pathway; in animals, protection by melatonin against bacterially induced damage is associated with inhibition or activation of various signaling pathways, including key regulators such as NF-κB, STAT-1, Nrf2, NLRP3 inflammasome, MAPK and TLR-2/4. Moreover, melatonin can reduce formation of reactive oxygen and nitrogen species (ROS, RNS), promote detoxification and protect mitochondrial damage. Altogether, we propose that melatonin could be an effective approach against various pathogenic bacterial infections.


Asunto(s)
Antibacterianos/farmacología , Inflamasomas/metabolismo , Melatonina/farmacología , Sepsis/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Humanos , Inflamasomas/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/efectos de los fármacos , FN-kappa B/metabolismo , Hojas de la Planta , Especies Reactivas de Oxígeno , Sepsis/genética , Sepsis/inmunología
19.
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
20.
Drug Discov Ther ; 15(2): 66-72, 2021 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-33716240

RESUMEN

Curcumin, a major component of turmeric, is known to exhibit multiple biological functions including antitumor activity. We previously reported that the mitogen-activated protein kinase (MAPK) scaffold protein c-Jun NH2-terminal kinase (JNK)-associated leucine zipper protein (JLP) reduces curcumin-induced cell death by modulating p38 MAPK and autophagy through the regulation of lysosome positioning. In this study, we investigated the role of JNK/stress-activated protein kinase-associated protein 1 (JSAP1), a JLP family member, in curcumin-induced stress, and found that JSAP1 also attenuates curcumin-induced cell death. However, JSAP1 knockout showed no or little effect on the activation of JNK and p38 MAPKs in response to curcumin. In addition, small molecule inhibitors of JNK and p38 MAPKs did not increase curcumin-induced cell death. Furthermore, JSAP1 depletion did not impair lysosome positioning and autophagosome-lysosome fusion. Instead, we noticed substantial autolysosome accumulation accompanied by an inefficient autophagic flux in JSAP1 knockout cells. Taken together, these results indicate that JSAP1 is involved in curcumin-induced cell death differently from JLP, and may suggest that JSAP1 plays a role in autophagosome degradation and its dysfunction results in enhanced cell death. The findings of this study may contribute to the development of novel therapeutic approaches using curcumin for cancer.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/farmacología , Antineoplásicos/farmacología , Curcumina/farmacología , Proteína Quinasa 9 Activada por Mitógenos/metabolismo , Proteínas del Tejido Nervioso/farmacología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Antineoplásicos/efectos adversos , Autofagia/efectos de los fármacos , Autofagia/genética , Técnicas de Cultivo de Célula , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Curcumina/efectos adversos , Desarrollo de Medicamentos/métodos , Humanos , Leucina Zippers/genética , Lisosomas/efectos de los fármacos , Lisosomas/genética , 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 , Neoplasias/tratamiento farmacológico , Proteínas del Tejido Nervioso/metabolismo , Sustancias Protectoras , Especies Reactivas de Oxígeno/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA