Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
Add more filters










Publication year range
1.
Phytomedicine ; 128: 155551, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38569293

ABSTRACT

BACKGROUND: Because obesity is associated with a hyperplasia-mediated increase in adipose tissue, inhibiting cell proliferation during mitotic clonal expansion (MCE) is a leading strategy for preventing obesity. Although (-)-hydroxycitric acid (HCA) is used to control obesity, the molecular mechanisms underlying its effects on MCE are poorly understood. PURPOSE: This study aimed to investigate the potential effects of HCA on MCE and underlying molecular mechanisms affecting adipogenesis and obesity improvements. METHODS: Preadipocyte cell line, 3T3-L1, were treated with HCA; oil red O, cell proliferation, cell cycle, and related alterations in signaling pathways were examined. High-fat diet (HFD)-fed mice were administered HCA for 12 weeks; body and adipose tissues weights were evaluated, and the regulation of signaling pathways in epidydimal white adipose tissue were examined in vivo. RESULTS: Here, we report that during MCE, HCA attenuates the proliferation of the preadipocyte cell line, 3T3-L1, by arresting the cell cycle at the G0/G1 phase. In addition, HCA markedly inhibits Forkhead Box O1 (FoxO1) phosphorylation, thereby inducing the expression of cyclin-dependent kinase inhibitor 1B and suppressing the levels of cyclin-dependent kinase 2, cyclin E1, proliferating cell nuclear antigen, and phosphorylated retinoblastoma. Importantly, we found that ribosomal protein S6 kinase A1 (RPS6KA1) influences HCA-mediated inactivation of FoxO1 and its nuclear exclusion. An animal model of obesity revealed that HCA reduced high-fat diet-induced obesity by suppressing adipocyte numbers as well as epididymal and mesenteric white adipose tissue mass, which is attributed to the regulation of RPS6KA1, FoxO1, CDKN1B and PCNA that had been consistently identified in vitro. CONCLUSIONS: These findings provide novel insights into the mechanism by which HCA regulates adipogenesis and highlight the RPS6KA1/FoxO1 signaling axis as a therapeutic target for obesity.


Subject(s)
Cell Proliferation , Citrates , Forkhead Box Protein O1 , Obesity , Ribosomal Protein S6 Kinases, 90-kDa , Animals , Mice , 3T3-L1 Cells/drug effects , Adipocytes/drug effects , Adipocytes/metabolism , Adipogenesis/drug effects , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Cell Proliferation/drug effects , Citrates/pharmacology , Citrates/therapeutic use , Diet, High-Fat/adverse effects , Forkhead Box Protein O1/antagonists & inhibitors , Forkhead Box Protein O1/metabolism , Mice, Inbred C57BL , Mitosis/drug effects , Obesity/drug therapy , Obesity/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/antagonists & inhibitors , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Signal Transduction/drug effects
2.
Curr Pharm Des ; 30(6): 440-447, 2024.
Article in English | MEDLINE | ID: mdl-38343056

ABSTRACT

BACKGROUND: It has been reported that inhibition of Fucosyltransferase4 (FUT4) to activate Forkhead box O1 (FOXO1) can lead to apoptosis of cancer cells, however, the mechanism in osteosarcoma is still unclear. OBJECTIVE: To explore the biological significance of the connection between FUT4 and FOXO1 in osteosarcoma growth. METHODS: In vitro tests were conducted using the human osteoblast cell line and the osteosarcoma cell lines. QRT-PCR assay as well as western blot assay were used to ascertain the relative expression levels of FUT4 and FOXO1 in the cells. By using the CCK-8 assay, colony assay, EDU assay, wound healing assay and Transwell assay, osteosarcoma cells' ability to proliferate, migrate and invade were examined in relation to si- FUT4. TUNEL test was used to evaluate Si-impact FUT4's on KHOS and U2OS apoptosis in osteosarcoma cells. Western blot assay was used to identify the expression of proliferative, migrating and apoptosis-related protein markers in osteosarcoma cells KHOS and U2OS and the expression of important proteins in the Wnt/ ß-catenin signaling pathway. RESULTS: In comparison with osteoblasts, osteosarcoma cells expressed more FUT4. The osteosarcoma cells' capacities to proliferate, invade, and migrate were markedly inhibited by the inhibition of FUT4 expression, which also increased osteosarcoma cell apoptosis. The Wnt/ß-catenin signaling pathway was blocked by upregulating FOXO1 expression, which was in turn inhibited by inhibiting FUT4 expression. CONCLUSION: Osteosarcoma cells express more FUT4. The Wnt/ß-catenin signaling pathway has a significant effect on osteosarcoma cell death, and inhibition of FUT4 expression may target FOXO1 activation to decrease osteosarcoma cells' ability to proliferate, invade, and migrate.


Subject(s)
Apoptosis , Cell Proliferation , Forkhead Box Protein O1 , Fucosyltransferases , Osteosarcoma , Humans , Bone Neoplasms/pathology , Bone Neoplasms/metabolism , Bone Neoplasms/genetics , Cell Movement , Forkhead Box Protein O1/metabolism , Forkhead Box Protein O1/antagonists & inhibitors , Forkhead Box Protein O1/genetics , Fucosyltransferases/genetics , Fucosyltransferases/metabolism , Fucosyltransferases/antagonists & inhibitors , Gene Silencing , Osteosarcoma/pathology , Osteosarcoma/metabolism , Osteosarcoma/genetics , Tumor Cells, Cultured
3.
Mol Metab ; 66: 101618, 2022 12.
Article in English | MEDLINE | ID: mdl-36283677

ABSTRACT

OBJECTIVES: Insulin treatment remains the sole effective intervention for Type 1 Diabetes. Here, we investigated the therapeutic potential of converting intestinal epithelial cells to insulin-producing, glucose-responsive ß-like cells by targeted inhibition of FOXO1. We have previously shown that this can be achieved by genetic ablation in gut Neurogenin3 progenitors, adenoviral or shRNA-mediated inhibition in human gut organoids, and chemical inhibition in Akita mice, a model of insulin-deficient diabetes. METHODS: We profiled two novel FOXO1 inhibitors in reporter gene assays, and hepatocyte gene expression studies, and in vivo pyruvate tolerance test (PTT) for their activity and specificity. We evaluated their glucose-lowering effect in mice rendered insulin-deficient by administration of streptozotocin. RESULTS: We provide evidence that two novel FOXO1 inhibitors, FBT432 and FBT374 have glucose-lowering and gut ß-like cell-inducing properties in mice. FBT432 is also highly effective in combination with a Notch inhibitor in this model. CONCLUSION: The data add to a growing body of evidence suggesting that FOXO1 inhibition be pursued as an alternative treatment to insulin administration in diabetes.


Subject(s)
Diabetes Mellitus, Experimental , Forkhead Box Protein O1 , Animals , Mice , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Forkhead Box Protein O1/antagonists & inhibitors , Glucose/metabolism , Insulin/metabolism , Streptozocin
4.
Comput Math Methods Med ; 2022: 4597087, 2022.
Article in English | MEDLINE | ID: mdl-35126623

ABSTRACT

OBJECTIVE: To explore the possibility of microRNA miR-31-3p as a biomarker for bone metastasis of non-small-cell lung cancer (NSCLC) and its molecular mechanism to the invasion and metastasis of NSCLC cells. METHODS: Real-time quantitative PCR (RT-qPCR) was used to detect the expression levels of miR-31-3p and forkhead box 1 (FOXO1) in NSCLC tissues, serum, and cells to analyze the correlation between the expression levels of miR-31-3p and the clinicopathology of NSCLC. After interference with or overexpressing miR-31-3p, NSCLC cell proliferation, apoptosis, invasion ability, and migration ability were detected by MTT, flow cytometry, Transwell, and scratch experiment, respectively. The interaction between miR-31-3p and FOXO1 was further verified by the dual-luciferase reporter experiment. Western blot was performed to detect the protein expression of FOXO1 in tissues and FOXO1, RhoA, p-RhoA, ROCK-2, and p-ROCK-2 in cells. RESULTS: In tissues, serum, and NSCLC cell line A549 of the NSCLC patients, the expression of FOXO1 was notably lower, and the miR-31-3p expression was significantly higher. Overexpression of miR-31-3p could distinctly improve the proliferation, invasion, and migration of A549 cells, meanwhile inhibit cell apoptosis, and activate the RhoA/ROCK-2 signaling pathway, while interfering with the expression of miR-31-3p has the opposite function. Besides, bioinformatics analysis and luciferase reporter assay confirmed that FOXO1 was a target gene of miR-31-3p. Overexpressing FOXO1 could inhibit the proliferation and metastasis of A549 cells, but overexpressing miR-31-3p reverses the results. CONCLUSION: This study confirmed that miR-31-3p promotes the proliferation, invasion, and migration of NSCLC cells and inhibits apoptosis through targeted regulating FOXO1 and be a potential therapeutic targets for the treatment of NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Forkhead Box Protein O1/antagonists & inhibitors , Forkhead Box Protein O1/genetics , Lung Neoplasms/genetics , MicroRNAs/genetics , A549 Cells , Adult , Apoptosis/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/secondary , Case-Control Studies , Cell Movement/genetics , Cell Proliferation/genetics , Computational Biology , Female , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , MicroRNAs/metabolism , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Metastasis/genetics , Signal Transduction/genetics , Young Adult , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism
5.
Int Immunopharmacol ; 101(Pt A): 108339, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34758440

ABSTRACT

Inulae Flos was widely distributed throughout Europe, Africa, and Asia, and was commonly used as a folk medicine in clinic for treating various respiratory diseases, including cough, asthma, bronchitis, pulmonary fibrosis, and pneumonia. However, the ingredients responsible for the pharmacology effects of I. Flos and the underlying mechanisms remain unclear. In this study, the effects of 16 known sesquiterpene lactones and flavonoids from I. Flos on TGF-ß1-induced fibroblast activation were assessed by phenotypic high-content screening. Among those sixteen compounds, 1ß-hydroxy alantolactone (HAL), the main characteristic sesquiterpene lactone from I. Flos, exhibited remarkable inhibitory activity. The further studies showed that HAL significantly inhibited the proliferation and induced the apoptosis of human fibroblast cell lines HELF and MRC-5 in a concentration-dependent manner. It also reduced intracellular ROS production, suppressed the mRNA expressions of E-cad, TGF-ß1, Smad3, Col I, α-SMA and TNF-α, and downregulated protein expressions of α-SMA and F-actin. Furthermore, HAL significantly reduced the levels of HA, LN, PC-III and IV-C in serum, TNF-α and IL-6 in BALF, and TGF-ß1, HYP and Col I in lung tissues of bleomycin (BLM)-treated rats. HAL significantly downregulated the expressions of p-JNK, FOXO1, p-p65, α-SMA, p-smad3 and Col I but upregulated p-FOXO1, which could be reversed by JNK agonist anisomycin. These results demonstrated that HAL induced the apoptosis of lung fibroblast cells activated by TGF-ß1 and improved BLM-induced lung fibrosis in rats via inhibiting JNK/FOXO1/NF-κB pathway.


Subject(s)
Antifibrotic Agents/therapeutic use , Forkhead Box Protein O1/metabolism , MAP Kinase Signaling System/drug effects , Pulmonary Fibrosis/drug therapy , Sesquiterpenes/therapeutic use , Animals , Antifibrotic Agents/isolation & purification , Fibroblasts/drug effects , Fluorescent Antibody Technique , Forkhead Box Protein O1/antagonists & inhibitors , Humans , Inula/chemistry , Male , Plant Extracts/isolation & purification , Plant Extracts/therapeutic use , Pulmonary Fibrosis/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Sesquiterpenes/isolation & purification , Signal Transduction/drug effects
6.
Acta Cir Bras ; 36(8): e360802, 2021.
Article in English | MEDLINE | ID: mdl-34644770

ABSTRACT

PURPOSE: To evaluate the influence of atractylenolide (Atr) III on sepsis-induced lung damage. METHODS: We constructed a mouse sepsis model through cecal ligation and puncture. These mice were allocated to the normal, sepsis, sepsis + Atr III-L (2 mg/kg), as well as Atr III-H (8 mg/kg) group. Lung injury and pulmonary fibrosis were accessed via hematoxylin-eosin (HE) and Masson's staining. We used terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) and flow cytometry for detecting sepsis-induced lung cell apoptosis. The contents of the inflammatory cytokines in lung tissue were measured via enzyme-linked immunosorbent assay (ELISA). RESULTS: Atr III-H did not only reduce sepsis-induced lung injury and apoptosis level, but also curbed the secretion of inflammatory factors. Atr III-H substantially ameliorated lung function and raised Bcl-2 expression. Atr III-H eased the pulmonary fibrosis damage and Bax, caspase-3, Vanin-1 (VNN1), as well as Forkhead Box Protein O1 (FoxO1) expression. CONCLUSIONS: Atr III alleviates sepsis-mediated lung injury via inhibition of FoxO1 and VNN1 protein.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Forkhead Box Protein O1/antagonists & inhibitors , Lung Injury , Sepsis , Sesquiterpenes , Animals , Apoptosis , GPI-Linked Proteins/antagonists & inhibitors , Lactones , Mice , Sepsis/complications , Sepsis/drug therapy , Sesquiterpenes/pharmacology
7.
Comput Math Methods Med ; 2021: 8773423, 2021.
Article in English | MEDLINE | ID: mdl-34552661

ABSTRACT

OBJECTIVE: Prostate cancer (PCa) is considered the most serious cancer in the world. Nevertheless, the accuracy of current biomarkers, such as pathological staging, Gleason's score, and serum prostate-specific antigen (PSA) levels, is limited. FOXO1 is a key downstream effector of PTEN and a tumor suppressor in PCA, which has been reported extensively. However, the clinical relevance of FOXO1 in PCa remains unclear. METHODS: In this study, we first detected its expression in four public databases to explore the clinical role of FOXO1. Verification of the knockdown effect of FOXO1 siRNA was performed by real-time PCR analysis. Changes in cell viability were assessed using cell counting kit-8 (CCK-8) assays. In addition, we verified the effect of FOXO1 on the PCa cell cycle using a cell cycle assay. RESULTS: Herein, we found that FOXO1 was significantly downregulated in PCa tissues and was significantly associated with Gleason's score, age, biochemical recurrence (BCR), and lymph node (LN) status, while FOXO1 expression was independent of pathological staging and preoperative PSA levels. The Kaplan-Meier survival analysis showed that PCA patients with high FOXO1 expression were less likely to develop BCR compared with patients with low FOXO1 expression. In terms of function, FOXO1 inhibition significantly promoted the proliferation and cell cycle progression of PCa cells. CONCLUSIONS: In summary, our study suggests that FOXO1 may be one of the prognostic factors that describe the risk of PCa for BCR. These results suggest that FOXO1 may be a therapeutic target for PCa.


Subject(s)
Forkhead Box Protein O1/metabolism , Prostatic Neoplasms/metabolism , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Computational Biology , Down-Regulation , Forkhead Box Protein O1/antagonists & inhibitors , Forkhead Box Protein O1/genetics , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Kallikreins/metabolism , Male , Middle Aged , Prognosis , Prostate-Specific Antigen/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , RNA, Small Interfering/genetics , Transcriptome , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
8.
Aging Cell ; 20(8): e13430, 2021 08.
Article in English | MEDLINE | ID: mdl-34278704

ABSTRACT

Dental pulp stem cells (DPSCs) play a vital role in tooth restoration, regeneration, and homeostasis. The link between DPSC senescence and tooth aging has been well-recognized. ROR2 plays an important role in aging-related gene expression. However, the expression and function of ROR2 in DPSC aging remain largely unknown. In this study, we found that ROR2 expression was significantly decreased in aged pulp tissues and DPSCs. The depletion of ROR2 in young DPSCs inhibits their self-renewal capacity, while its overexpression in aged DPSCs restores their self-renewal capacity. Interestingly, we found that sphingomyelin (SM) is involved in the senescence of DPSCs regulated by ROR2. Mechanistically, we confirmed that ROR2 inhibited the phosphorylation of STK4, which promoted the translocation of Forkhead Box O1 (FOXO1) to the nucleus. STK4 inhibition or knockdown of FOXO1 markedly increased the proliferation of DPSCs and upregulated the expression of SMS1, which catalyzed SM biogenesis. Moreover, FOXO1 directly bound to the SMS1 promoter, repressing its transcription. Our findings demonstrated the critical role of the ROR2/STK4-FOXO1/SMS1 axis in the regulation of SM biogenesis and DPSC senescence, providing a novel target for antagonizing tooth aging.


Subject(s)
Dental Pulp/metabolism , Forkhead Box Protein O1/antagonists & inhibitors , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Sphingomyelins/biosynthesis , Stem Cells/metabolism , Down-Regulation , Humans
9.
Cell Rep ; 35(1): 108935, 2021 04 06.
Article in English | MEDLINE | ID: mdl-33826891

ABSTRACT

Type 2 diabetes (T2D) increases the risk for diabetic cardiomyopathy and is characterized by diastolic dysfunction. Myocardial forkhead box O1 (FoxO1) activity is enhanced in T2D and upregulates pyruvate dehydrogenase (PDH) kinase 4 expression, which inhibits PDH activity, the rate-limiting enzyme of glucose oxidation. Because low glucose oxidation promotes cardiac inefficiency, we hypothesize that FoxO1 inhibition mitigates diabetic cardiomyopathy by stimulating PDH activity. Tissue Doppler echocardiography demonstrates improved diastolic function, whereas myocardial PDH activity is increased in cardiac-specific FoxO1-deficient mice subjected to experimental T2D. Pharmacological inhibition of FoxO1 with AS1842856 increases glucose oxidation rates in isolated hearts from diabetic C57BL/6J mice while improving diastolic function. However, AS1842856 treatment fails to improve diastolic function in diabetic mice with a cardiac-specific FoxO1 or PDH deficiency. Our work defines a fundamental mechanism by which FoxO1 inhibition improves diastolic dysfunction, suggesting that it may be an approach to alleviate diabetic cardiomyopathy.


Subject(s)
Diabetes Mellitus, Type 2/physiopathology , Diastole/physiology , Forkhead Box Protein O1/metabolism , Myocardium/enzymology , Pyruvate Dehydrogenase Complex/metabolism , Animals , Diabetes Mellitus, Experimental/physiopathology , Diabetic Cardiomyopathies/physiopathology , Fibrosis , Forkhead Box Protein O1/antagonists & inhibitors , Forkhead Box Protein O1/deficiency , Glucose/metabolism , Homeostasis , Lipids/toxicity , Male , Mice, Inbred C57BL
10.
J Cell Biochem ; 122(8): 777-786, 2021 08.
Article in English | MEDLINE | ID: mdl-33644883

ABSTRACT

Lysyl oxidase (LOX) is a copper-containing enzyme and its overexpression in tumor tissues promote tumor metastasis through the crosslinking of extracellular matrix. Our previous report demonstrated that LOX expression is significantly increased in human leukemic THP-1 cell-derived M2-like macrophages, and histone modification plays a key role in its induction. However, the rigorous mechanism of LOX regulation remains unclear. In this study, we investigated the role of functional transcription factors, hypoxia-inducible factor 1α (HIF1α), signal transducer and activator of transcription 3 (STAT3) and forkhead box O1 (FOXO1) in LOX regulation in M2-like macrophages. HIF1α expression was significantly increased in M2-like macrophages, and HIF1α inhibitor, TX402, suppressed LOX induction. The significant STAT3 activation was also observed in M2-like macrophages. Additionally, LOX induction was canceled in the presence of STAT3 inhibitor, S3I-201, suggesting that HIF1α and STAT3 pathways play a critical role in LOX induction. On the other hand, our ChIP results clearly indicated that the enrichment of FOXO1 within the lox promoter region was dramatically decreased in M2-like macrophages. In this context, knockdown of FOXO1 further enhanced LOX induction. LOX induction and HIF1α binding to the lox promoter region were suppressed in FOXO1-overexpressed cells, suggesting that the FOXO1 binding to the lox promoter region counteracted HIF1α binding to that region. Overall, the present data suggested that both of HIF1α and STAT3 were required for LOX induction in M2-like macrophages, and loss of FOXO1 within the lox promoter region facilitated HIF1α binding to that region which promoted LOX induction.


Subject(s)
Gene Expression Regulation, Enzymologic , Macrophages/metabolism , Protein-Lysine 6-Oxidase/biosynthesis , Forkhead Box Protein O1/antagonists & inhibitors , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Protein-Lysine 6-Oxidase/genetics , Response Elements , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , THP-1 Cells
11.
Drug Des Devel Ther ; 15: 245-257, 2021.
Article in English | MEDLINE | ID: mdl-33519190

ABSTRACT

BACKGROUND: Paeoniflorin (Pae) possesses anti-tumor activity in various malignancies. However, it is unclear whether Pae plays a sensitizer role in breast cancer (BC) and the molecular mechanisms involved in this process. Our oligonucleotide microarray revealed that microRNA (miR)-15b is the most significantly downregulated miRNA in MCF-7/4-hydroxytamoxifen (4-OHT) cells treated with Pae. This paper summarized the relevance of Pae in BC cell endocrine resistance to tamoxifen (Tam) and the molecular mechanisms involved miR-15b expression. MATERIALS AND METHODS: 4-OHT-resistant BC cell lines were developed and treated with different concentrations of Pae. Flow cytometry, lactose dehydrogenase activity, caspase-3 activity, colony formation, and EdU assays were carried out to assess the impact of Pae on BC cells. Differentially expressed miRNAs in BC cells treated with Pae were analyzed by microarray. Targeting mRNAs of screened miR-15b as well as the binding of forkhead box O1 (FOXO1) to the cyclin D1 (CCND1) promoter sequence were predicted through bioinformatics analysis. Finally, the expression of ß-catenin signaling-related genes in cells was detected by Western blotting. RESULTS: Pae (100 µg/mL) inhibited the clonality and viability of BC cells, while enhancing apoptosis in vitro. Pae also repressed miR-15b expression. Overexpression of miR-15b restored the growth and resistance of BC cells to 4-OHT. Moreover, Pae promoted FOXO1 expression by downregulating miR-15b, thereby transcriptionally inhibiting CCND1 and subsequently blocking ß-catenin signaling. CONCLUSION: Pae inhibits 4-OHT resistance in BC cells by regulating the miR-15b/FOXO1/CCND1/ß-catenin pathway.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Down-Regulation/drug effects , Glucosides/pharmacology , MicroRNAs/antagonists & inhibitors , Monoterpenes/pharmacology , Apoptosis/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclin D1/antagonists & inhibitors , Cyclin D1/genetics , Cyclin D1/metabolism , Drug Screening Assays, Antitumor , Female , Forkhead Box Protein O1/antagonists & inhibitors , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Tamoxifen/pharmacology , Tumor Cells, Cultured , beta Catenin/antagonists & inhibitors , beta Catenin/genetics , beta Catenin/metabolism
12.
Acta cir. bras ; 36(8): e360802, 2021. graf
Article in English | LILACS, VETINDEX | ID: biblio-1339011

ABSTRACT

ABSTRACT Purpose: To evaluate the influence of atractylenolide (Atr) III on sepsis-induced lung damage. Methods: We constructed a mouse sepsis model through cecal ligation and puncture. These mice were allocated to the normal, sepsis, sepsis + Atr III-L (2 mg/kg), as well as Atr III-H (8 mg/kg) group. Lung injury and pulmonary fibrosis were accessed via hematoxylin-eosin (HE) and Masson's staining. We used terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) and flow cytometry for detecting sepsis-induced lung cell apoptosis. The contents of the inflammatory cytokines in lung tissue were measured via enzyme-linked immunosorbent assay (ELISA). Results: Atr III-H did not only reduce sepsis-induced lung injury and apoptosis level, but also curbed the secretion of inflammatory factors. Atr III-H substantially ameliorated lung function and raised Bcl-2 expression. Atr III-H eased the pulmonary fibrosis damage and Bax, caspase-3, Vanin-1 (VNN1), as well as Forkhead Box Protein O1 (FoxO1) expression. Conclusions: Atr III alleviates sepsis-mediated lung injury via inhibition of FoxO1 and VNN1 protein.


Subject(s)
Animals , Mice , Sesquiterpenes/pharmacology , Sepsis/complications , Sepsis/drug therapy , Lung Injury , Forkhead Box Protein O1/antagonists & inhibitors , Amidohydrolases/antagonists & inhibitors , Apoptosis , GPI-Linked Proteins/antagonists & inhibitors , Lactones
13.
Biochem Pharmacol ; 182: 114246, 2020 12.
Article in English | MEDLINE | ID: mdl-33011160

ABSTRACT

Elevated glucose levels in diabetes mellitus is associated with increased oxidized low density lipoprotein (oxLDL). High glucose (HG) and oxLDL are key inducers of oxidative stress and inflammatory processes responsible for diabetic vascular disorders. Rosmarinic acid is a polyphenol with antioxidant, anti-inflammatory and insulin-sensitizing effects. However, whether rosmarinic acid protects against diabetic atherosclerosis remains unknown. In this study, we aimed to investigate the protective effect of rosmarinic acid against diabetic atherosclerosis and the related signaling pathway. oxLDL-mediated oxidative stress upregulated thioredoxin-interacting protein (TXNIP) and subsequently induced binding of TXNIP to NLRP3 to mediate NLRP3 inflammasome assembly and activation under HG conditions in ECs. Reactive oxygen species (ROS) scavengers, p38 and FOXO1 inhibitors and TXNIP siRNA inhibited TXNIP protein upregulation and NLRP3 inflammasome assembly and activation. Rosmarinic acid abrogated TXNIP protein upregulation and the interaction between TXNIP and NLRP3 to attenuate NLRP3 inflammasome assembly and activation and eventually IL-1ß secretion in ECs through downregulating ROS production, p38 phosphorylation and FOXO1 protein induction in ECs. These findings show that rosmarinic acid inhibits endothelial dysfunction which is shown in diabetic atherosclerosis through downregulating the p38-FOXO1-TXNIP pathway and inhibiting inflammasome activation.


Subject(s)
Carrier Proteins/metabolism , Cinnamates/pharmacology , Depsides/pharmacology , Forkhead Box Protein O1/metabolism , Glucose/toxicity , Inflammasomes/metabolism , Lipoproteins, LDL/toxicity , p38 Mitogen-Activated Protein Kinases/metabolism , Antioxidants/pharmacology , Carrier Proteins/antagonists & inhibitors , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Down-Regulation/physiology , Forkhead Box Protein O1/antagonists & inhibitors , Humans , Inflammasomes/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Rosmarinic Acid
14.
J Biol Chem ; 295(45): 15292-15306, 2020 11 06.
Article in English | MEDLINE | ID: mdl-32868454

ABSTRACT

Muscle atrophy is regulated by the balance between protein degradation and synthesis. FOXO1, a transcription factor, helps to determine this balance by activating pro-atrophic gene transcription when present in muscle fiber nuclei. Foxo1 nuclear efflux is promoted by AKT-mediated Foxo1 phosphorylation, eliminating FOXO1's atrophy-promoting effect. AKT activation can be promoted by insulin-like growth factor 1 (IGF1) or insulin via a pathway including IGF1 or insulin, phosphatidylinositol 3-kinase, and AKT. We used confocal fluorescence time-lapse imaging of FOXO1-GFP in adult isolated living muscle fibers maintained in culture to explore the effects of IGF1 and insulin on FOXO1-GFP nuclear efflux with and without pharmacological inhibitors. We observed that although AKT inhibitor blocks the IGF1- or insulin-induced effect on FOXO1 nuclear efflux, phosphatidylinositol 3-kinase inhibitors, which we show to be effective in these fibers, do not. We also found that inhibition of the protein kinase ACK1 or ATM contributes to the suppression of FOXO1 nuclear efflux after IGF1. These results indicate a novel pathway that has been unexplored in the IGF1- or insulin-induced regulation of FOXO1 and present information useful both for therapeutic interventions for muscle atrophy and for further investigative areas into insulin insensitivity and type 2 diabetes.


Subject(s)
Cell Nucleus/metabolism , Forkhead Box Protein O1/metabolism , Insulin-Like Growth Factor I/metabolism , Insulin/metabolism , Muscle Fibers, Skeletal/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Animals , Cell Nucleus/drug effects , Female , Forkhead Box Protein O1/antagonists & inhibitors , Insulin-Like Growth Factor I/antagonists & inhibitors , Mice , Muscle Fibers, Skeletal/drug effects , Optical Imaging , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Signal Transduction/drug effects
15.
J Microbiol Biotechnol ; 30(9): 1412-1419, 2020 Sep 28.
Article in English | MEDLINE | ID: mdl-32522961

ABSTRACT

Human noroviruses (HuNoVs) are a leading cause of gastroenteritis outbreaks worldwide. However, the paucity of appropriate cell culture model for HuNoV replication has prevented developing effective anti-HuNoV therapy. In this study, first, the replication of the virus at various temperatures in different cells was compared, which showed that lowering the culture temperature from 37°C significantly increased virus replication in Madin-Darby canine kidney (MDCK) cells. Second, the expression levels of autophagy-, immune-, and apoptosis-related genes at 30°C and 37°C were compared to explore factors affecting HuNoV replication. HuNoV cultured at 37°C showed significantly increased autophagy- (ATG5 and ATG7) and immune- (IFNA, IFNB, ISG15, and NFKB) related genes compared to mock. However, the virus cultured at 30°C showed significantly decreased expression of autophagy- (ATG5 and ATG7) and not significantly different in major immune- (IFNA, ISG15, and NFKB) related genes compared to mock. Importantly, expression of the transcription factor FOXO1, which controls autophagy- and immune-related gene expression, was significantly lower at 30°C. Moreover, FOXO1 inhibition in temperature-optimized MDCK cells enhanced HuNoV replication, highlighting FOXO1 inhibition as an approach for successful virus replication. In the temperature-optimized cells, various HuNoV genotypes were successfully replicated, with GI.8 showing the highest replication levels followed by GII.1, GII.3, and GII.4. Furthermore, ultrastructural analysis of the infected cells revealed functional HuNoV replication at low temperature, with increased cellular apoptosis and decreased autophagic vacuoles. In conclusion, temperature-optimized MDCK cells can be used as a convenient culture model for HuNoV replication by inhibiting FOXO1, providing adaptability to different genotypes.


Subject(s)
Forkhead Box Protein O1/metabolism , Norovirus/physiology , Virus Replication , Animals , Dogs , Forkhead Box Protein O1/antagonists & inhibitors , Gastroenteritis/virology , Genotype , Madin Darby Canine Kidney Cells
16.
Drug Des Devel Ther ; 14: 2079-2090, 2020.
Article in English | MEDLINE | ID: mdl-32581510

ABSTRACT

PURPOSE: Osteoarthritis (OA) is associated with chronic low-grade inflammation. Resveratrol exerts protective effects on OA through its anti-inflammatory property; however, the mechanism of resveratrol on anti-inflammatory signaling pathways has not been fully elucidated yet. The aim of the present study was to investigate whether resveratrol-mediated PI3K/Akt expression is linked to TLR4/NF-κB pathway and the role of TLR4/Akt/FoxO1 axis in the anti-osteoarthritic effect of resveratrol. METHODS: SW1353 cells stimulated by IL-1ß (10 ng/mL) were cultured in the presence or absence of resveratrol (50 µM) and then treated with TLR4 siRNA, PI3K inhibitor LY294002 or FoxO1 siRNA, respectively. The associated proteins of TLR4 signaling pathways and TLR4/Akt/FoxO1 axis were evaluated by Western blot. The level of IL-6 in the supernatant was detected by ELISA. RESULTS: IL-1ß treatment increased the expression of TLR4/NF-κB and phosphorylation of PI3K/Akt and FoxO1, while additional resveratrol further upregulated the expression of PI3K/Akt and FoxO1 phosphorylation but downregulated TLR4 signals in SW1353 cells. Further analyses by the inhibition of TLR4, PI3K/Akt and FoxO1 signaling pathways, respectively, showed that the activation of TLR4 can induce PI3K/Akt phosphorylation, which increases the phosphorylation of FoxO1 and inactivates it. Next, inactivated-FoxO1 can reduce the expression of TLR4, which forms a self-limiting mechanism of inflammation. Resveratrol treatment can upregulate PI3K/Akt phosphorylation and inactivate FoxO1, thereby reducing TLR4 and inflammation. CONCLUSION: This study reveals that TLR4/Akt/FoxO1 inflammatory self-limiting mechanism may exist in IL-1ß-stimulated SW1353 cells. This study reveals a novel cross-talk mechanism which is between integrated PI3K/Akt/FoxO1 signaling network and TLR4-driven innate responses in IL-1ß-stimulated SW1353 cells. Resveratrol may exert anti-OA effect by enhancing the self-limiting mechanism of inflammation through TLR4/Akt/FoxO1 axis.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Interleukin-1beta/pharmacology , NF-kappa B/metabolism , Osteoarthritis/drug therapy , Resveratrol/pharmacology , Signal Transduction/drug effects , Toll-Like Receptor 4/antagonists & inhibitors , Cell Line, Tumor , Forkhead Box Protein O1/antagonists & inhibitors , Forkhead Box Protein O1/metabolism , Humans , Osteoarthritis/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Toll-Like Receptor 4/metabolism
17.
J Biol Chem ; 295(13): 4265-4276, 2020 03 27.
Article in English | MEDLINE | ID: mdl-32075913

ABSTRACT

Recent clinical investigations indicate that anthracycline-based chemotherapies induce early decline in heart mass in cancer patients. Heart mass decline may be caused by a decrease in cardiac cell number because of increased cell death or by a reduction in cell size because of atrophy. We previously reported that an anthracycline, doxorubicin (DOX), induces apoptotic death of cardiomyocytes by activating cyclin-dependent kinase 2 (CDK2). However, the signaling pathway downstream of CDK2 remains to be characterized, and it is also unclear whether the same pathway mediates cardiac atrophy. Here we demonstrate that DOX exposure induces CDK2-dependent phosphorylation of the transcription factor forkhead box O1 (FOXO1) at Ser-249, leading to transcription of its proapoptotic target gene, Bcl-2-interacting mediator of cell death (Bim). In cultured cardiomyocytes, treatment with the FOXO1 inhibitor AS1842856 or transfection with FOXO1-specific siRNAs protected against DOX-induced apoptosis and mitochondrial damage. Oral administration of AS1842856 in mice abrogated apoptosis and prevented DOX-induced cardiac dysfunction. Intriguingly, pharmacological FOXO1 inhibition also attenuated DOX-induced cardiac atrophy, likely because of repression of muscle RING finger 1 (MuRF1), a proatrophic FOXO1 target gene. In conclusion, DOX exposure induces CDK2-dependent FOXO1 activation, resulting in cardiomyocyte apoptosis and atrophy. Our results identify FOXO1 as a promising drug target for managing DOX-induced cardiotoxicity. We propose that FOXO1 inhibitors may have potential as cardioprotective therapeutic agents during cancer chemotherapy.


Subject(s)
Atrophy/genetics , Cardiotoxicity/genetics , Cyclin-Dependent Kinase 2/genetics , Forkhead Box Protein O1/genetics , Muscle Proteins/genetics , Tripartite Motif Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Animals , Apoptosis/drug effects , Atrophy/chemically induced , Atrophy/pathology , Cardiotoxicity/etiology , Cardiotoxicity/pathology , Disease Models, Animal , Doxorubicin/adverse effects , Forkhead Box Protein O1/antagonists & inhibitors , Gene Expression Regulation/drug effects , Heart/drug effects , Heart/physiopathology , Humans , Mice , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Neoplasms/complications , Neoplasms/drug therapy , Neoplasms/genetics , Quinolones/pharmacology , Signal Transduction/drug effects
18.
J Mol Cell Cardiol ; 138: 244-255, 2020 01.
Article in English | MEDLINE | ID: mdl-31866378

ABSTRACT

BACKGROUND: Survivin has an anti-apoptotic effect against anthracycline-induced cardiotoxicity. Clinically, statin use is associated with a lower risk for heart failure in breast cancer patients with anthracycline chemotherapy. So, the purpose of our study was to investigate whether survivin mediates the protective effect of statin against anthracycline-induced cardiotoxicity. METHODS: Mice were treated once a week with 5 mg/kg doxorubicin for 4 weeks with or without atorvastatin 20 mg/kg every day then heart tissues were analyzed. Molecular and cellular biology analyses were performed with H9c2 cell lysates. RESULTS: Doxorubicin suppressed survivin expression via activation of FOXO1 in H9c2 cardiomyocytes. Whereas, atorvastatin inhibited FOXO1 by increasing phosphorylation and inhibiting nuclear localization. Doxorubicin induced FOXO1 binding to STAT3 and prevented STAT3 from interacting with Sp1. However, atorvastatin inhibited these interactions and stabilized STAT3/Sp1 transcription complex. Chromatin immunoprecipitation analysis demonstrated that doxorubicin decreased STAT3/Sp1 complex binding to survivin promoter, whereas atorvastatin stabilized this binding. In mouse model, atorvastatin rescued doxorubicin-induced reduction of survivin expression and of heart function measured by cardiac magnetic resonance imaging. CONCLUSIONS: Our study suggested a new pathophysiologic mechanism that survivin mediated protective effect of atorvastatin against doxorubicin-induced cardiotoxicity via FOXO1/STAT3/Sp1 transcriptional network.


Subject(s)
Atorvastatin/pharmacology , Cardiotonic Agents/pharmacology , Cytoprotection , Doxorubicin/toxicity , Forkhead Box Protein O1/antagonists & inhibitors , Myocytes, Cardiac/metabolism , Survivin/metabolism , Animals , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cytoprotection/drug effects , Disease Models, Animal , Forkhead Box Protein O1/metabolism , Male , Mice, Inbred C57BL , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Phosphatidylinositol 3-Kinases/metabolism , Protein Transport/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats , Signal Transduction/drug effects , Sp1 Transcription Factor/metabolism , Transcription, Genetic/drug effects , Transcriptional Activation/drug effects
19.
Br J Pharmacol ; 177(2): 432-448, 2020 01.
Article in English | MEDLINE | ID: mdl-31655022

ABSTRACT

BACKGROUND AND PURPOSE: Growing evidence indicates targeting mitochondrial dynamics and biogenesis could accelerate recovery from renal ischemia-reperfusion (I/R) injury, but the underlying mechanisms remain elusive. Transcription factor forkhead box O1 (FOXO1) is a key regulator of mitochondrial homeostasis and plays a pathological role in the progression of renal disease. EXPERIMENTAL APPROACH: A mouse model of renal I/R injury and a hypoxia/reoxygenation (H/R) injury model for human renal tubular epithelial cells were used. KEY RESULTS: I/R injury up-regulated renal expression of FOXO1 and treatment with FOXO1-selective inhibitor AS1842856 prior to I/R injury decreased serum urea nitrogen, serum creatinine and the tubular damage score after injury. Post-I/R injury AS1842856 treatment could also ameliorate renal function and improve the survival rate of mice following injury. AS1842856 administration reduced mitochondrial-mediated apoptosis, suppressed the overproduction of mitochondrial ROS and accelerated recovery of ATP both in vivo and in vitro. Additionally, FOXO1 inhibition improved mitochondrial biogenesis and suppressed mitophagy. Expression of PPAR-γ coactivator 1α (PGC-1α), a master regulator of mitochondrial biogenesis, was down-regulated in both I/R and H/R injury, which could be abrogated by FOXO1 inhibition. Experiments using integrated bioinformatics analysis and coimmunoprecipitation established that FOXO1 inhibited PGC-1α transcription by competing with cAMP-response element binding protein (CREB) for its binding to transcriptional coactivators CREBBP/EP300 (CBP/P300). CONCLUSION AND IMPLICATIONS: These findings suggested that FOXO1 was critical to maintain mitochondrial function in renal tubular epithelial cells and FOXO1 may serve as a therapeutic target for pharmacological intervention in renal I/R injury.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Forkhead Box Protein O1/antagonists & inhibitors , Kidney Diseases/prevention & control , Kidney Tubules/drug effects , Mitochondria/drug effects , Organelle Biogenesis , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Quinolones/pharmacology , Reperfusion Injury/prevention & control , Animals , Apoptosis/drug effects , Cell Line , Disease Models, Animal , Forkhead Box Protein O1/metabolism , Humans , Kidney Diseases/metabolism , Kidney Diseases/pathology , Kidney Tubules/metabolism , Kidney Tubules/pathology , Male , Mice, Inbred C57BL , Mitochondria/metabolism , Mitochondria/pathology , Mitophagy/drug effects , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Signal Transduction
20.
J Biol Chem ; 294(46): 17555-17569, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31601652

ABSTRACT

The forkhead box O (FOXO) proteins are transcription factors involved in the differentiation of many cell types. Type II collagen (Col2) Cre-Foxo1-knockout and Col2-Cre-Foxo1,3,4 triple-knockout mice exhibit growth plate malformation. Moreover, recent studies have reported that in some cells, the expressions and activities of FOXOs are promoted by transforming growth factor ß1 (TGFß1), a growth factor playing a key role in chondrogenic differentiation. Here, using a murine chondrogenic cell line (ATDC5), mouse embryos, and human mesenchymal stem cells, we report the mechanisms by which FOXOs affect chondrogenic differentiation. FOXO1 expression increased along with chondrogenic differentiation, and FOXO1 inhibition suppressed chondrogenic differentiation. TGFß1/SMAD signaling promoted expression and activity of FOXO1. In ATDC5, FOXO1 knockdown suppressed expression of sex-determining region Y box 9 (Sox9), a master regulator of chondrogenic differentiation, resulting in decreased collagen type II α1 (Col2a1) and aggrecan (Acan) expression after TGFß1 treatment. On the other hand, chemical FOXO1 inhibition suppressed Col2a1 and Acan expression without suppressing Sox9 To investigate the effects of FOXO1 on chondrogenic differentiation independently of SOX9, we examined FOXO1's effects on the cell cycle. FOXO1 inhibition suppressed expression of p21 and cell-cycle arrest in G0/G1 phase. Conversely, FOXO1 overexpression promoted expression of p21 and cell-cycle arrest. FOXO1 inhibition suppressed expression of nascent p21 RNA by TGFß1, and FOXO1 bound the p21 promoter. p21 inhibition suppressed expression of Col2a1 and Acan during chondrogenic differentiation. These results suggest that FOXO1 is necessary for not only SOX9 expression, but also cell-cycle arrest during chondrogenic differentiation via TGFß1 signaling.


Subject(s)
Chondrogenesis/genetics , Forkhead Box Protein O1/genetics , SOX9 Transcription Factor/genetics , Transforming Growth Factor beta1/genetics , Aggrecans/genetics , Animals , Cell Cycle Checkpoints/genetics , Cell Differentiation/genetics , Collagen Type II/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , Forkhead Box Protein O1/antagonists & inhibitors , Gene Expression Regulation, Developmental/genetics , Humans , Mesenchymal Stem Cells/metabolism , Mice , Smad Proteins/genetics , Transforming Growth Factor beta1/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...