Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
J Med Food ; 27(4): 301-311, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38377551

RESUMEN

Baicalin has been acknowledged for its anti-inflammatory properties. However, its potential impact on osteoarthritis (OA) has not yet been explored. Therefore, our study aimed to examine the effects of Baicalin on OA, both in laboratory and animal models. To evaluate its efficacy, human chondrocytes affected by OA were treated with interleukin-1ß and/or Baicalin. The effects were then assessed through viability tests using the cell counting kit-8 (CCK-8) method and flow cytometry. In addition, we analyzed the expressions of various factors such as FOXO1, autophagy, apoptosis, and cartilage synthesis and breakdown to corroborate the effects of Baicalin. We also assessed the severity of OA through analysis of tissue samples. Our findings demonstrate that Baicalin effectively suppresses inflammatory cytokines and MMP-13 levels caused by collagenase-induced osteoarthritis, while simultaneously preserving the levels of Aggrecan and Col2. Furthermore, Baicalin has been shown to enhance autophagy. Through the use of FOXO1 inhibitors, lentivirus-mediated knockdown, and chromatin immunoprecipitation, we verified that Baicalin exerts its protective effects by activating FOXO1, which binds to the Beclin-1 promoter, thereby promoting autophagy. In conclusion, our results show that Baicalin has potential as a therapeutic agent for treating OA (Clinical Trial Registration number: 2023-61).


Asunto(s)
Cartílago Articular , Flavonoides , Proteína Forkhead Box O1 , Osteoartritis , Animales , Humanos , Apoptosis , Cartílago Articular/efectos de los fármacos , Cartílago Articular/metabolismo , Condrocitos , Flavonoides/farmacología , Flavonoides/uso terapéutico , Proteína Forkhead Box O1/efectos de los fármacos , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/metabolismo , Homeostasis , Interleucina-1beta/metabolismo , Osteoartritis/tratamiento farmacológico , Osteoartritis/genética , Osteoartritis/metabolismo
2.
Cell Mol Biol (Noisy-le-grand) ; 69(12): 44-51, 2023 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-38063119

RESUMEN

Traditional Chinese medicine (TCM) encompasses treatment strategies for diabetes, which is referred to as "Consumptive Thirsty" syndrome. Recently, there has been discovery regarding the mapping between TCM and signaling molecules, which has revealed a remarkable consistency between TCM and modern medicine from a molecular perspective. In this manuscript, we have summarized the etiology and treatment strategies for diabetes in TCM and have examined these strategies in the context of molecular mechanisms. Our review demonstrates that the targeting molecule of TCM for the treatment of diabetes is FoxO1, a transcription factor that plays a pivotal role in regulating gluconeogenesis and glycogenolysis. TCM ranks the development of diabetes into three stages and utilizes different herbal formulas to control FoxO1 accordingly. At Stage 1, TCM inhibits FoxO1 by lowering its expression in the lung. At Stage 2, TCM increases the expression of FoxO1 by suppressing its activity in the stomach. At Stage 3, TCM utilizes the famous herbal formula Liuwei Dihuang Pill to amplify the expression of FoxO1, and to enhance the concentrations of potassium, phosphorus, and Wnt, but to reduce the concentration of calcium. These TCM treatment strategies are in accordance with corresponding mechanisms in modern medicine.


Asunto(s)
Diabetes Mellitus , Medicamentos Herbarios Chinos , Proteína Forkhead Box O1 , Humanos , Diabetes Mellitus/tratamiento farmacológico , Medicamentos Herbarios Chinos/farmacología , Medicamentos Herbarios Chinos/uso terapéutico , Medicina Tradicional China/métodos , Síndrome , Proteína Forkhead Box O1/efectos de los fármacos , Proteína Forkhead Box O1/metabolismo
3.
Chem Biol Drug Des ; 99(2): 344-361, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34862852

RESUMEN

Diabetes mellitus type 2 (T2D) is one of the metabolic disorders suffered by a global human being. Certain factors, such as lifestyle and heredity, can increase a person's tendency for T2D. Various genes and proteins play a role in the development of insulin resistance and ultimately diabetes in which one central protein that is discussed in this review is FoxO1. In this review, we regard FoxO1 activation as detrimental, promote high plasma glucose level, and induce insulin resistance. Indeed, many contrasting studies arise since FoxO1 is an important protein to alleviate oxidative stress and promote cell survival, for example, also by preventing hyperglycemic-induced cell death. Inter-relation to PPARG, another important protein in metabolism, is also discussed. Ultimately, we discussed contrasting approaches of targeting FoxO1 to combat diabetes mellitus by small molecules.


Asunto(s)
Diabetes Mellitus Tipo 2/fisiopatología , Proteína Forkhead Box O1/fisiología , Bibliotecas de Moléculas Pequeñas/farmacología , Glucemia/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Proteína Forkhead Box O1/efectos de los fármacos , Humanos , Resistencia a la Insulina , Estrés Oxidativo , PPAR gamma/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal
4.
Exp Biol Med (Maywood) ; 246(21): 2307-2316, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34233525

RESUMEN

Megestrol acetate is a common and efficient anticancer progesterone. To explore the activity and the therapeutic mechanisms of megestrol acetate in endometrial cancer, human endometrial cancer cell lines Ishikawa and HHUA overexpressing progesterone receptor A (PR-A) and progesterone receptor B (PR-B) were treated with megestrol acetate. Cell viability, apoptosis, cycle arrest, and senescence, as well as the expressions of p21 and p16, two hallmarks of cellular senescence, were evaluated. Compared with the control, >10 nmol/L megestrol acetate treatment could significantly reduce endometrial cancer cell growth, and induce the irreversible G1 arrest and cell senescence. The expression of cyclin D1 in megestrol acetate treated cells was downregulated, while the expressions of p21 and p16 were upregulated via PR-B isoform. FOXO1 inhibitor AS1842856 could significantly abrogate megestrol acetate-induced cell senescence, suggesting that FOXO1 was involved in megestrol acetate/PR-B axis. These findings may provide a new understanding for the treatment of human endometrial cancer.


Asunto(s)
Antineoplásicos Hormonales/uso terapéutico , Senescencia Celular/efectos de los fármacos , Neoplasias Endometriales/tratamiento farmacológico , Proteína Forkhead Box O1/efectos de los fármacos , Megestrol/uso terapéutico , Receptores de Progesterona/efectos de los fármacos , Western Blotting , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Femenino , Proteína Forkhead Box O1/metabolismo , Humanos , Receptores de Progesterona/metabolismo , Transducción de Señal/efectos de los fármacos
5.
Aging (Albany NY) ; 13(13): 17370-17379, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34198266

RESUMEN

In this study, we used bioinformatics and an in vitro cellular model of glucocorticoid-induced osteoporosis to investigate mechanisms underlying the beneficial effects of baicalein (BN) against osteoporosis. STITCH database analysis revealed 30 BN-targeted genes, including AKT1, CCND1, MTOR, and PTEN. Functional enrichment analysis demonstrated that BN-targeted genes were enriched in 49 Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. MIRWALK2.0 database analysis identified 110 enriched KEGG pathways related to osteoporosis. A Venn diagram demonstrated that 26 KEGG pathways were common between osteoporosis and BN-targeted genes. The top 5 common KEGG pathways were prostate cancer, bladder cancer, glioma, pathways in cancer, and melanoma. BN-targeted genes in the top 5 shared KEGG pathways were involved in PI3K-AKT, MAPK, p53, ErbB, and mTOR signaling pathways. In addition, glucocorticoid-induced osteoporosis in MC3T3-E1 cells was partially reversed by BN through inhibition of AKT, which, by upregulating FOXO1, enhanced expression of bone turnover markers (ALP, OCN, Runx2, and Col 1) and extracellular matrix mineralization. These findings demonstrate that BN suppresses osteoporosis via an AKT/FOXO1 signaling pathway.


Asunto(s)
Conservadores de la Densidad Ósea/farmacología , Flavanonas/farmacología , Proteína Forkhead Box O1/genética , Osteoporosis/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-akt/genética , Transducción de Señal/efectos de los fármacos , Células 3T3 , Animales , Huesos/efectos de los fármacos , Huesos/metabolismo , Calcificación Fisiológica/efectos de los fármacos , Calcificación Fisiológica/genética , Ciclina D1/genética , Bases de Datos Factuales , Matriz Extracelular/metabolismo , Proteína Forkhead Box O1/efectos de los fármacos , Glucocorticoides , Humanos , Ratones , Osteoporosis/inducido químicamente , Fosfohidrolasa PTEN/genética , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Serina-Treonina Quinasas TOR/genética
6.
Pharmacology ; 106(9-10): 551-563, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34175854

RESUMEN

BACKGROUND: Oral squamous cell carcinoma (OSCC) is among the most prevalent head and neck malignancies globally, and it is associated with high mortality rates. Actein is one of the primary active components extractable from the rhizomes of Cimicifuga foetida. This study aimed to evaluate the anti-OSCC effects of actein and evaluate the potential underlying mechanisms. METHODS AND RESULTS: CCK-8 cell proliferation experiments demonstrated significant dose- and time-dependent anti-OSCC effects of actein, while actein had weak cytotoxic effects on normal oral cell lines. Flow cytometry for cell cycle evaluation revealed that actein could induce cell cycle arrest at the G1 phase among OSCC cell lines. In our Annexin V/PI double staining apoptosis analysis, actein induced significant apoptosis among OSCC cells, with upregulation of Bax and downregulation of Bcl-2. Our mechanistic study implicated the involvement of the Akt/FoxO1 pathway in the anti-OSCC effects of actein. Akt1 and Akt2 expression significantly decreased in association with the FoxO1 upregulation. Furthermore, Bim and p21 were significantly upregulated, while survivin expression was downregulated. Finally, actein treatment was associated with significant p-Akt downregulation and p-FoxO1 upregulation in OSCC cells, demonstrating the validated roles of Akt/FoxO1 in actein-mediated OSCC cell apoptosis and cell cycle arrest. FoxO1 knockdown significantly reversed the anti-OSCC effects of actein. Additionally, a xenograft model indicated that actein could inhibit OSCC cell growth in vivo. CONCLUSIONS: Our findings demonstrated that actein could be a strong anti-OSCC candidate. Further evaluations of its safety and effectiveness are necessary before it can be considered for clinical use.


Asunto(s)
Carcinoma de Células Escamosas/patología , Medicamentos Herbarios Chinos/farmacología , Proteína Forkhead Box O1/efectos de los fármacos , Neoplasias de la Boca/patología , Saponinas/farmacología , Triterpenos/farmacología , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Femenino , Genes bcl-2/efectos de los fármacos , Humanos , Ratones , Ratones Endogámicos C57BL , Factores de Tiempo , Regulación hacia Arriba , Ensayos Antitumor por Modelo de Xenoinjerto , Proteína X Asociada a bcl-2/efectos de los fármacos
7.
Biofactors ; 47(5): 740-753, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34058789

RESUMEN

Melatonin (Mel.), also known as the magic hormone, is a nocturnally secreted hormone orchestrates the clearance of free radicals that have been built up and cumulated during day. This study aims to detect the impact of pineal gland removal on the incidence of tumor development and to assess the signaling pathways via which exogenous melatonin counteract cancer growth. This goal has been achieved by novel approach for pineal destruction using dental micromotor which validated by melatonin downregulation in blood plasma. Mice were injected sub-cutenously with Ehrlich cells to develop solid tumor as a murine model of breast cancer. The increase at tumor markers carcino embryonic antigen, TNFα, and nuclear factor kappa-light-chain-enhancer of activated B cells was over countered by exogenous melatonin supplementation (20 mg/kg) daily for 1 month. The anticancer effects of melatonin were significantly mediated by scavenging H2 O2 and NO and diminishing of lipid peroxidation marker malondialdehyde. The real-time polymerase chain Rx analyses indicated a significant effect of Melatonin in upregulating the expression of miR215, fork head box protein O1 (foxO1), and downregulation of miR96. Flowcytometric analyses indicated a significant effect of melatonin on induction of cell cycle arrest at G1 phase which was further confirmed by Ki67 downregulation. Immunohistochemical analyses indicated the role of melatonin in upregulating P53-dependent apoptosis and downregulating CD44 signaling for survivin, matrix metallo-protein kinase 2, and vascular endothelial growth factor to inhibit cell survival and metastasis. In conclusion, this study sheds the light on M./P53/miR215/CD44 with an emphasis on M./miR96//foxO1 signaling cascades, as a novel pathway of melatonin signaling in adenocarcinoma to diminish cancer cell growth, survival and metastasis.


Asunto(s)
Adenocarcinoma/genética , Antioxidantes/farmacología , Proteína Forkhead Box O1/genética , Melatonina/farmacología , MicroARNs/genética , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Proteína Forkhead Box O1/efectos de los fármacos , Ratones , MicroARNs/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
8.
Mol Biol Rep ; 48(4): 3089-3096, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33866495

RESUMEN

It has been documented that aging increases the risk of cardiovascular disease including myocardial ischemia/reperfusion (IR) injury and acute myocardial infarction. In this study, we aimed to investigate the individual or combined effects of nicotinamide mononucleotide (NMN) and melatonin (Mel) treatment on apoptotic markers, expression of SIRT3, and FOXO1, and infarct size of the aged myocardium subjected to IR injury. Sixty aged Wistar rats (22-24 months) were assigned to five groups including sham, IR, NMN+IR, Mel+IR, and NMN+Mel+IR (combination therapy). Isolated hearts were exposed to 30-min regional ischemia followed by 60-min reperfusion. NMN (100 mg/kg/day/i.p.) was injected every second day starting on day 28 before IR injury. Melatonin was added to the perfusion solution five minutes prior to and until 15 min after the start of reperfusion. The infarct size was assessed by computerized planimetry. The mRNA levels of SIRT3, FOXO1, and apoptotic genes Bax, Bcl-2, and Caspase-3 were estimated by real-time PCR. All treatments reduced infarct size as compared with the IR group. Melatonin and NMN upregulated the gene expression of Bcl-2, SIRT3, and FOXO1 and downregulated the gene expression of Bax, and Caspase-3, in comparison to the IR group. Also, the protein levels of SIRT3, quantified by Western blotting, were upregulated by the interventions. The effects of combination therapy were significantly greater than those of melatonin or NMN alone. These findings indicate that the combined administration of NMN and melatonin can protect the aged heart against IR injury by decreasing apoptosis and activating the SIRT3/FOXO1 pathway.


Asunto(s)
Apoptosis/efectos de los fármacos , Melatonina/farmacología , Daño por Reperfusión Miocárdica , Mononucleótido de Nicotinamida/farmacología , Envejecimiento/efectos de los fármacos , Animales , Combinación de Medicamentos , Proteína Forkhead Box O1/efectos de los fármacos , Proteína Forkhead Box O1/metabolismo , Corazón/fisiopatología , Masculino , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Proteínas del Tejido Nervioso/efectos de los fármacos , Proteínas del Tejido Nervioso/metabolismo , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Sirtuina 3/efectos de los fármacos , Sirtuina 3/metabolismo
9.
Histol Histopathol ; 36(7): 785-794, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33876419

RESUMEN

BACKGROUND: ß-Ecdysone has been reported to perform a protective effect to prevent interleukin 1ß (IL-1ß)-induced apoptosis and inflammatory response in chondrocytes. In our study, the chondroprotective effects of ß-Ecdysone were explored in a mouse model of collagenase-induced osteoarthritis (OA). METHODS: Injection of collagenase in the left knee was implemented to establish a mouse model of OA. The histomorphological analysis was detected using safranine O staining. Serum pro-inflammatory cytokines were measured by ELISA assays. Protein expression in the femur and chondrocytes was analyzed using western blot. Chondrocyte apoptosis was evaluated by terminal-deoxynucleoitidyl transferase mediated nick end labeling (TUNEL) staining. RESULTS: Treatment of OA mice with ß-Ecdysone supplementation significantly inhibited the production of pro-inflammatory cytokines. Histologic examination exhibited that the degradation of proteoglycans and the loss of trabecular bone were observed in collagenase-injected mice. However, OA-like changes were attenuated by ß-Ecdysone administration in collagenase-injected mice. Both in vivo and in vitro models, nuclear forkhead box O1 (FOXO1) protein expression was significantly reduced in the femur of collagenase-treated mice and IL-1ß-stimulated chondrocytes. However, ß-Ecdysone treatment was able to rescue FOXO1 protein expression in the nucleus to inhibit the transcription and translation of a disintegrin-like and metallopeptidase (reprolysin type) with thrombospondin type 1 motif, 4 (ADAMTS-4) and ADAMTS-5. CONCLUSION: The findings suggested that ß-Ecdysone functioned as a FOXO1 activator to protect collagenase-induced cartilage damage. FOXO1 might be a potential molecular target of ß-Ecdysone for the effective prevention and treatment of OA.


Asunto(s)
Artritis Experimental/patología , Cartílago Articular/efectos de los fármacos , Ecdisterona/farmacología , Osteoartritis de la Rodilla/patología , Transducción de Señal/efectos de los fármacos , Proteína ADAMTS4/efectos de los fármacos , Proteína ADAMTS4/metabolismo , Proteína ADAMTS5/efectos de los fármacos , Proteína ADAMTS5/metabolismo , Animales , Artritis Experimental/metabolismo , Cartílago Articular/metabolismo , Cartílago Articular/patología , Proteína Forkhead Box O1/efectos de los fármacos , Proteína Forkhead Box O1/metabolismo , Ratones , Osteoartritis de la Rodilla/metabolismo , Transducción de Señal/fisiología
10.
Mol Metab ; 49: 101187, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33577983

RESUMEN

OBJECTIVE: Forkhead box protein O1 (FOXO1) plays a key role in regulating hepatic glucose production, but investigations of FOXO1 inhibition as a potential therapeutic approach have been hampered by a lack of selective chemical inhibitors. By profiling structurally diverse FOXO1 inhibitors, the current study validates FOXO1 as a viable target for the treatment of diabetes. METHODS: Using reporter gene assays, hepatocyte gene expression studies, and in vivo studies in mice, we profiled our leading tool compound 10 and a previously characterized FOXO1 inhibitor, AS1842856 (AS). RESULTS: We show that AS has significant FOXO1-independent effects, as demonstrated by testing in FOXO1-deficient cell lines and animals, while compound 10 is highly selective for FOXO1 both in vitro and in vivo and fails to elicit any effect in genetic models of FOXO1 ablation. Chronic administration of compound 10 improved insulin sensitivity and glucose control in db/db mice without causing weight gain. Furthermore, chronic compound 10 treatment combined with FGF21 led to synergistic glucose lowering in lean, streptozotocin-induced diabetic mice. CONCLUSIONS: We show that the widely used AS compound has substantial off-target activities and that compound 10 is a superior tool molecule for the investigation of FOXO1 function. In addition, we provide preclinical evidence that selective FOXO1 inhibition has potential therapeutic benefits for diabetes as a monotherapy or in combination with FGF21.


Asunto(s)
Glucemia/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Proteína Forkhead Box O1/metabolismo , Animales , Diabetes Mellitus Tipo 2/metabolismo , Factores de Crecimiento de Fibroblastos/genética , Proteína Forkhead Box O1/efectos de los fármacos , Proteína Forkhead Box O1/genética , Glucosa/metabolismo , Hepatocitos/metabolismo , Resistencia a la Insulina , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Noqueados , Quinolonas/farmacología
11.
Hum Exp Toxicol ; 40(5): 882-894, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33233951

RESUMEN

Exogenous and endogenous formaldehyde (FA) both play an important role in cell growth and migration; however, their potential role in osteoblasts remains largely unclear. Cell counting kit-8 (CCK-8) and wound-healing assays revealed that FA exposure at naturally occurring concentrations inhibited the proliferation and migration of mouse preosteoblast MC3T3-E1 cells. Moreover, RNA sequencing (RNA-seq) analysis revealed that FoxO1 signaling pathway components displayed distinct expression patterns upon FA exposure, reflected through significant enrichment of cell migration. In particular, FoxO1-, Sirt1-, and FA-induced protein expression, which was closely associated with cell proliferation and migration, was confirmed by western blotting. The results obtained indicated that the FoxO1 pathway is involved in FA-induced inhibition of cell growth and migration.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Proteína Forkhead Box O1/efectos de los fármacos , Formaldehído/toxicidad , Osteoblastos/efectos de los fármacos , Cicatrización de Heridas/efectos de los fármacos , Animales , Células Cultivadas/efectos de los fármacos , Humanos , Ratones , Modelos Animales
12.
Curr Pharm Biotechnol ; 22(5): 609-621, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33198615

RESUMEN

BACKGROUND: Osteoporosis, characterized by bone loss, usually occurs with the increased bone resorption and decreased bone formation. H2O2-induced MC3T3-E1 cells are commonly used for the study of osteoblastic activities, which play a crucial role in bone formation. OBJECTIVE: This study aimed to investigate the effects of Phosphocreatine (PCr) on the osteoblastic activities in H2O2-induced MC3T3-E1 cells and elaborate on the possible molecular mechanism. METHODS: The Osteoprotegerin (OPG)/Receptor Activator of NF-κB Ligand (RANKL) ratio and osteogenic markers were detected to investigate the effects of PCr on osteoblastic activities, and the osteoblastic apoptosis was detected using Hochest staining. Moreover, oxidative stress, Adenosine Triphosphate (ATP) generation and the expression of Sirtuin 1 (SIRT1), Forkhead Box O 1 (FOXO1) and Peroxisome Proliferator-Activated Receptor Γ Coactivator-1α (PGC-1α) were also examined to uncover the possible molecular mechanism in H2O2-induced MC3T3-E1 cells. RESULT: The results showed that PCr promoted the osteoblastic differentiation by increasing the expression levels of osteogenic markers of Alkaline Phosphatase (ALP) and Runt-related transcription factor 2 (Runx2), as well as increased the OPG/RANKL ratio and suppressed the osteoblastic apoptosis in H2O2-induced MC3T3-E1 cells. Moreover, treatment with PCr suppressed reactive oxygen species (ROS) over-generation and promoted the ATP production as well as increased the PGC-1α, FOXO1 and SIRT1 protein expression levels in H2O2-induced MC3T3-E1 cells. CONCLUSION: PCr treatment could promote osteoblastic activities via suppressing oxidative stress and increasing the ATP generation in H2O2-induced MC3T3-E1 cells. In addition, the positive effects of PCr on osteoblasts might be regulated by SIRT1/FOXO1/ PGC-1α signaling pathway.


Asunto(s)
Proteína Forkhead Box O1/efectos de los fármacos , Peróxido de Hidrógeno/farmacología , Osteoblastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/efectos de los fármacos , Fosfocreatina/farmacología , Transducción de Señal/efectos de los fármacos , Sirtuina 1/efectos de los fármacos , Células 3T3 , Fosfatasa Alcalina/biosíntesis , Fosfatasa Alcalina/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Subunidad alfa 1 del Factor de Unión al Sitio Principal/biosíntesis , Subunidad alfa 1 del Factor de Unión al Sitio Principal/efectos de los fármacos , Ratones , Osteoprotegerina/efectos de los fármacos , Osteoprotegerina/metabolismo , Estrés Oxidativo , Ligando RANK/efectos de los fármacos , Ligando RANK/metabolismo , Especies Reactivas de Oxígeno
13.
Nutrients ; 12(9)2020 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-32842462

RESUMEN

Obesity is a notable risk factor for developing type 2 diabetes, augmenting the concern of obese diabetes (ObD). Anti-obesity and antioxidant effects of red pepper seeds extract (RPSE) have increased our expectations that RPSE would also improve the pathological phenotypes of obese diabetes. Therefore, we hypothesized that RPSE would have an anti-diabetic effect in ObD mice. Animals were assigned either as follows: (1) db/+, (2) db/db control, (3) RPSE (200 mg/kg bw), or (4) a comparative control (metformin 150 mg/kg bw). RPSE was orally administered daily for 8 weeks. As a result, RPSE supplementation improved diabetic phenotypes, including fasting glucose, hemoglobin (HbA1c), and insulin levels. Pro-inflammatory cytokines, tumor necrosis factor-alpha (TNF-α) and interleukin 6 (IL-6), and triglycerides were reduced in RPSE-treated mice. RPSE supplementation also diminished the rate-limiting enzymes of gluconeogenesis, including glucose 6-phosphatas (G6Pase) and phosphoenolpyruvate carboxykinase (PEPCK), in the liver. RPSE supplementation increased the phosphorylation of forkhead box protein O1 (FOXO1) and AMP-activated protein kinase (AMPK), which underlined the mechanism of the anti-diabetic effects of RPSE. Taken together, RPSE has the potential to improve glycemic control by repressing hepatic gluconeogenesis via the phosphorylation of FOXO1 and AMPK in ObD mice.


Asunto(s)
Proteínas Quinasas Activadas por AMP/efectos de los fármacos , Glucemia/efectos de los fármacos , Capsicum , Diabetes Mellitus Experimental/sangre , Proteína Forkhead Box O1/efectos de los fármacos , Gluconeogénesis/efectos de los fármacos , Extractos Vegetales/farmacología , Proteínas Quinasas Activadas por AMP/sangre , Animales , Diabetes Mellitus Experimental/complicaciones , Modelos Animales de Enfermedad , Proteína Forkhead Box O1/sangre , Control Glucémico , Hígado/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/sangre , Obesidad/complicaciones , Fosforilación , Semillas
14.
J Neuroinflammation ; 17(1): 217, 2020 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-32698899

RESUMEN

BACKGROUND: Microglia, the principal sentinel immune cells of the central nervous system (CNS), play an extensively vital role in neuroinflammation and perioperative neurocognitive disorders (PND). Histamine, a potent mediator of inflammation, can both promote and prevent microglia-related neuroinflammation by activating different histamine receptors. Rat microglia express four histamine receptors (H1R, H2R, H3R, and H4R), among which the histamine 1 and 4 receptors can promote microglia activation, whereas the role and cellular mechanism of the histamine 2 and 3 receptors have not been elucidated. Therefore, we evaluated the effects and potential cellular mechanisms of histamine 2/3 receptors in microglia-mediated inflammation and PND. METHODS: This study investigated the role of histamine 2/3 receptors in microglia-induced inflammation and PND both in vivo and in vitro. In the in vivo experiments, rats were injected with histamine 2/3 receptor agonists in the right lateral ventricle and were then subjected to exploratory laparotomy. In the in vitro experiments, primary microglia were pretreated with histamine 2/3 receptor agonists before stimulation with lipopolysaccharide (LPS). Cognitive function, microglia activation, proinflammatory cytokine production, NF-κb expression, M1/M2 phenotypes, cell migration, and Toll-like receptor-4 (TLR4) expression were assessed. RESULTS: In our study, the histamine 2/3 receptor agonists inhibited exploratory laparotomy- or LPS-induced cognitive decline, microglia activation, proinflammatory cytokine production, NF-κb expression, M1/M2 phenotype transformation, cell migration, and TLR4 expression through the PI3K/AKT/FoxO1 pathway. CONCLUSION: Based on our findings, we conclude that histamine 2/3 receptors ameliorate PND by inhibiting microglia activation through the PI3K/AKT/FoxO1 pathway. Our results highlight histamine 2/3 receptors as potential therapeutic targets to treat neurological conditions associated with PND.


Asunto(s)
Agonistas de los Receptores Histamínicos/farmacología , Microglía/efectos de los fármacos , Complicaciones Cognitivas Postoperatorias/inmunología , Complicaciones Cognitivas Postoperatorias/metabolismo , Envejecimiento , Animales , Método Doble Ciego , Proteína Forkhead Box O1/efectos de los fármacos , Inyecciones Intraventriculares , Masculino , Metilhistaminas/farmacología , Fosfatidilinositol 3-Quinasas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores Histamínicos , Transducción de Señal/efectos de los fármacos , Tiazoles/farmacología
15.
J Diabetes Res ; 2019: 6198495, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31828164

RESUMEN

The angiopoietin-Tie-2 system plays a crucial role in the maintenance of endothelial integrity. Hyperglycemia and advanced glycation end-products (AGEs) are involved in endothelial cell dysfunction responsible of the pathogenesis of microvascular complications of diabetes. Here, we investigated whether glycated serum (GS) or hyperglycemia (HG) affect the angiopoietin-Tie-2 system in the microvascular endothelial cells HMEC-1. We found that culture for 5 days in the presence of AGEs and HG (alone or in combination) decreased cell proliferation, increased reactive oxygen species (ROS) production, and reduced ratio between the oxidized and the reduced form of glutathione. Since angiopoietin-1 (Ang-1) signaling regulates angiopoietin-2 (Ang-2) expression through inactivation of the forkhead transcription factor FoxO1, we investigated intracellular signaling of Ang-1 and expression of Ang-2. HG and AGEs reduced phosphorylation of Akt and abrogated phosphorylation of FoxO1 induced by Ang-1 without affecting neither Tie-2 expression nor its activation. Furthermore, AGEs and/or HG induced nuclear translocation of FoxO1 and increased Ang-2 production. In conclusion, we demonstrated that both hyperglycemia and AGEs affect the angiopoietin-Tie-2 system by impairing Ang-1/Tie-2 signaling and by increasing Ang-2 expression. These results suggest that therapeutic strategies useful in preventing or delaying the onset of diabetic vascular complications should be aimed to preserve Ang-1 signaling.


Asunto(s)
Angiopoyetina 1/metabolismo , Angiopoyetina 2/biosíntesis , Células Endoteliales/efectos de los fármacos , Glucosa/farmacología , Productos Finales de Glicación Avanzada/farmacología , Hiperglucemia/metabolismo , Receptor TIE-2/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Endoteliales/metabolismo , Proteína Forkhead Box O1/efectos de los fármacos , Proteína Forkhead Box O1/metabolismo , Glutatión/efectos de los fármacos , Glutatión/metabolismo , Humanos , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptor TIE-2/metabolismo , Transducción de Señal
16.
Biol Res ; 52(1): 57, 2019 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-31767027

RESUMEN

BACKGROUND: Gastric cancer is a common malignant tumor with high morbidity and mortality worldwide, which seriously affects human health. Gramicidin is a short peptide antibiotic which could be used for treating infection induced by bacteria or fungi. However, the anti-cancer effect of gramicidin on gastric cancer cells and its underlying mechanism remains largely unknown. RESULTS: Gastric cancer cells SGC-7901, BGC-823 and normal gastric mucosal cells GES-1 were treated with different concentrations of gramicidin respectively. The results of CCK-8 experiment revealed cellular toxicity of gramicidin to cancer cells while cell colony formation assay showed that gramicidin significantly inhibited the proliferation of gastric cancer cells, but had little effect on normal gastric mucosal cells. In addition, the wound healing assay showed that gramicidin inhibited the migration of SGC-7901 cell. Meanwhile, apoptosis and cell cycle analysis revealed that gramicidin induced cell apoptosis with G2/M cell cycle inhibition. Furthermore, western blot analysis demonstrated that gramicidin down-regulated the expression of cyclinD1 and Bcl-2 as well as the FoxO1 phosphorylation. CONCLUSIONS: The current study illustrated the anti-tumor activity of gramicidin on gastric cancer cells, providing a possibility for gramicidin to be applied in clinical practice for the treatment of gastric cancer.


Asunto(s)
Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Gramicidina/farmacología , Neoplasias Gástricas/patología , Línea Celular Tumoral , Ciclina D1/efectos de los fármacos , Ciclina D1/metabolismo , Regulación hacia Abajo , Proteína Forkhead Box O1/efectos de los fármacos , Proteína Forkhead Box O1/metabolismo , Humanos , Fosforilación , Proteínas Proto-Oncogénicas c-bcl-2/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo
18.
Muscle Nerve ; 60(2): 192-201, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31093982

RESUMEN

INTRODUCTION: We recently demonstrated the beneficial effects of 4-aminopyridine (4-AP), a potassium channel blocker, in enhancing remyelination and recovery of nerve conduction velocity and motor function after sciatic nerve crush injury in mice. Although muscle atrophy occurs very rapidly after nerve injury, the effect of 4-AP on muscle atrophy and intrinsic muscle contractile function is largely unknown. METHODS: Mice were assigned to sciatic nerve crush injury and no-injury groups and were followed for 3, 7, and 14 days with/without 4-AP or saline treatment. Morphological, functional, and transcriptional properties of skeletal muscle were assessed. RESULTS: In addition to improving in vivo function, 4-AP significantly reduced muscle atrophy with increased muscle fiber diameter and contractile force. Reduced muscle atrophy was associated with attenuated expression of atrophy-related genes and increased expression of proliferating stem cells. DISCUSSION: These findings provide new insights into the potential therapeutic benefits of 4-AP against nerve injury-induced muscle atrophy and dysfunction. Muscle Nerve 60: 192-201, 2019.


Asunto(s)
4-Aminopiridina/farmacología , Lesiones por Aplastamiento/fisiopatología , Músculo Esquelético/efectos de los fármacos , Atrofia Muscular/patología , Traumatismos de los Nervios Periféricos/fisiopatología , Bloqueadores de los Canales de Potasio/farmacología , Remielinización/efectos de los fármacos , Nervio Ciático/efectos de los fármacos , Animales , Lesiones por Aplastamiento/metabolismo , Lesiones por Aplastamiento/patología , Proteína Forkhead Box O1/efectos de los fármacos , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O3/efectos de los fármacos , Proteína Forkhead Box O3/genética , Ratones , Proteínas Musculares/efectos de los fármacos , Proteínas Musculares/genética , Músculo Esquelético/inervación , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Atrofia Muscular/genética , Traumatismos de los Nervios Periféricos/genética , Traumatismos de los Nervios Periféricos/patología , Regeneración/efectos de los fármacos , Nervio Ciático/lesiones , Nervio Ciático/patología , Nervio Ciático/fisiopatología , Proteínas de Motivos Tripartitos/efectos de los fármacos , Proteínas de Motivos Tripartitos/genética , Ubiquitina-Proteína Ligasas/efectos de los fármacos , Ubiquitina-Proteína Ligasas/genética
19.
Biol. Res ; 52: 57-57, 2019. ilus, graf
Artículo en Inglés | LILACS | ID: biblio-1505777

RESUMEN

BACKGROUND: Gastric cancer is a common malignant tumor with high morbidity and mortality worldwide, which seriously affects human health. Gramicidin is a short peptide antibiotic which could be used for treating infection induced by bacteria or fungi. However, the anti-cancer effect of gramicidin on gastric cancer cells and its underlying mechanism remains largely unknown. RESULTS: Gastric cancer cells SGC-7901, BGC-823 and normal gastric mucosal cells GES-1 were treated with different concentrations of gramicidin respectively. The results of CCK-8 experiment revealed cellular toxicity of gramicidin to cancer cells while cell colony formation assay showed that gramicidin significantly inhibited the proliferation of gastric cancer cells, but had little effect on normal gastric mucosal cells. In addition, the wound healing assay showed that gramicidin inhibited the migration of SGC-7901 cell. Meanwhile, apoptosis and cell cycle analysis revealed that gramicidin induced cell apoptosis with G2/M cell cycle inhibition. Furthermore, western blot analysis demonstrated that gramicidin down-regulated the expression of cyclinD1 and Bcl-2 as well as the FoxO1 phosphorylation. CONCLUSIONS: The current study illustrated the anti-tumor activity of gramicidin on gastric cancer cells, providing a possibility for gramicidin to be applied in clinical practice for the treatment of gastric cancer.


Asunto(s)
Humanos , Neoplasias Gástricas/patología , Ciclo Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Gramicidina/farmacología , Fosforilación , Regulación hacia Abajo , Proteínas Proto-Oncogénicas c-bcl-2/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ciclina D1/efectos de los fármacos , Ciclina D1/metabolismo , Línea Celular Tumoral , Proteína Forkhead Box O1/efectos de los fármacos , Proteína Forkhead Box O1/metabolismo
20.
Metabolism ; 87: 36-47, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29928895

RESUMEN

BACKGROUND: High Density Lipoprotein (HDL) and its main protein component, apolipoprotein A-I (apoA-I), have numerous atheroprotective functions on various tissues including the endothelium. Therapies based on reconstituted HDL containing apoA-I (rHDL-apoA-I) have been used successfully in patients with acute coronary syndrome, peripheral vascular disease or diabetes but very little is known about the genomic effects of rHDL-apoA-I and how they could contribute to atheroprotection. OBJECTIVE: The present study aimed to understand the endothelial signaling pathways and the genes that may contribute to rHDL-apoA-I-mediated atheroprotection. METHODS: Human aortic endothelial cells (HAECs) were treated with rHDL-apoA-I and their total RNA was analyzed with whole genome microarrays. Validation of microarray data was performed using multiplex RT-qPCR. The expression of ANGPTL4 in EA.hy926 endothelial cells was determined by RT-qPCR and Western blotting. The contribution of signaling kinases and transcription factors in ANGPTL4 gene regulation by HDL-apoA-I was assessed by RT-qPCR, Western blotting and immunofluorescence using chemical inhibitors or siRNA-mediated gene silencing. RESULTS: It was found that 410 transcripts were significantly changed in the presence of rHDL-apoA-I and that angiopoietin like 4 (ANGPTL4) was one of the most upregulated and biologically relevant molecules. In validation experiments rHDL-apoA-I, as well as natural HDL from human healthy donors or from transgenic mice overexpressing human apoA-I (TgHDL-apoA-I), increased ANGPTL4 mRNA and protein levels. ANGPTL4 gene induction by HDL was direct and was blocked in the presence of inhibitors for the AKT or the p38 MAP kinases. TgHDL-apoA-I caused phosphorylation of the transcription factor forkhead box O1 (FOXO1) and its translocation from the nucleus to the cytoplasm. Importantly, a FOXO1 inhibitor or a FOXO1-specific siRNA enhanced ANGPTL4 expression, whereas administration of TgHDL-apoA-I in the presence of the FOXO1 inhibitor or the FOXO1-specific siRNA did not induce further ANGPTL4 expression. These data suggest that FOXO1 functions as an inhibitor of ANGPTL4, while HDL-apoA-I blocks FOXO1 activity and induces ANGPTL4 through the activation of AKT. CONCLUSION: Our data provide novel insights into the global molecular effects of HDL-apoA-I on endothelial cells and identify ANGPTL4 as a putative mediator of the atheroprotective functions of HDL-apoA-I on the artery wall, with notable therapeutic potential.


Asunto(s)
Proteína 4 Similar a la Angiopoyetina/biosíntesis , Apolipoproteína A-I/farmacología , Células Endoteliales/metabolismo , Proteína Forkhead Box O1/metabolismo , Lipoproteínas HDL/farmacología , Proteína Oncogénica v-akt/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal/efectos de los fármacos , Proteína 4 Similar a la Angiopoyetina/efectos de los fármacos , Proteína 4 Similar a la Angiopoyetina/genética , Animales , Proteína Forkhead Box O1/efectos de los fármacos , Proteína Forkhead Box O1/genética , Expresión Génica/efectos de los fármacos , Silenciador del Gen , Voluntarios Sanos , Humanos , Ratones , Ratones Transgénicos , Análisis por Micromatrices , Proteína Oncogénica v-akt/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , ARN Interferente Pequeño/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA