Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 2.126
2.
Int Immunopharmacol ; 133: 112069, 2024 May 30.
Article En | MEDLINE | ID: mdl-38643710

Epigallocatechin-3-gallate (EGCG) is an important tea polyphenol with anti-tumor potential. Our previous studies revealed that EGCG was a promising immune checkpoint inhibitor (ICI) as it could downregulate expression of programmed cell death 1 ligand 1 (PD-L1) in tumor cells, thereby resulting tumor killing effect. In particular, EGCG can effectively avoid the inflammatory storm caused by anti-tumor therapy, which is a healthy green capacity absent from many ICIs. However, the relationship between EGCG and programmed cell death 1 (PD-1) of T cells remains unclear. In this work, we explored the effect of EGCG on T cells and found that EGCG suppressed PD-1 via inhibiting NF-κB phosphorylation and nuclear translocation. Furtherly, the capability of EGCG was confirmed in tumor-bearing mice to inhibit PD-1 expression in T cells and enhance apoptosis in tumor cells. These results implied that EGCG could inhibit the expression of PD-1 in T cells, thereby promoting anti-tumor effects of T cells. EGCG will be a promising candidate in anti-tumor therapy.


Catechin , NF-kappa B , Programmed Cell Death 1 Receptor , T-Lymphocytes , Catechin/analogs & derivatives , Catechin/pharmacology , Animals , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/antagonists & inhibitors , NF-kappa B/metabolism , Phosphorylation/drug effects , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Mice , Humans , Apoptosis/drug effects , Cell Line, Tumor , Mice, Inbred C57BL , Female , Cell Nucleus/metabolism , Cell Nucleus/drug effects , Active Transport, Cell Nucleus/drug effects
3.
Eur J Pharmacol ; 972: 176553, 2024 Jun 05.
Article En | MEDLINE | ID: mdl-38574838

Stroke poses a significant risk of mortality, particularly among the elderly population. The pathophysiological process of ischemic stroke is complex, and it is crucial to elucidate its molecular mechanisms and explore potential protective drugs. Ferroptosis, a newly recognized form of programmed cell death distinct from necrosis, apoptosis, and autophagy, is closely associated with the pathophysiology of ischemic stroke. N6022, a selective inhibitor of S-nitrosoglutathione reductase (GSNOR), is a "first-in-class" drug for asthma with potential therapeutic applications. However, it remains unclear whether N6022 exerts protective effects in ischemic stroke, and the precise mechanisms of its action are unknown. This study aimed to investigate whether N6022 mitigates cerebral ischemia/reperfusion (I/R) injury by reducing ferroptosis and to elucidate the underlying mechanisms. Accordingly, we established an oxygen-glucose deprivation/reperfusion (OGD/R) cell model and a middle cerebral artery occlusion/reperfusion (MCAO/R) mouse model to mimic cerebral I/R injury. Our data, both in vitro and in vivo, demonstrated that N6022 effectively protected against I/R-induced brain damage and neurological deficits in mice, as well as OGD/R-induced BV2 cell damage. Mechanistically, N6022 promoted Nrf2 nuclear translocation, enhancing intracellular antioxidant capacity of SLC7A11-GPX4 system. Furthermore, N6022 interfered with the interaction of GSNOR with GSTP1, thereby boosting the antioxidant capacity of GSTP1 and attenuating ferroptosis. These findings provide novel insights, showing that N6022 attenuates microglial ferroptosis induced by cerebral I/R injury through the promotion of Nrf2 nuclear translocation and inhibition of the GSNOR/GSTP1 axis.


Benzamides , Ferroptosis , Microglia , NF-E2-Related Factor 2 , Pyrroles , Reperfusion Injury , Animals , Ferroptosis/drug effects , NF-E2-Related Factor 2/metabolism , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Mice , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Male , Mice, Inbred C57BL , Signal Transduction/drug effects , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/drug therapy , Neuroprotective Agents/pharmacology , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Cell Nucleus/metabolism , Cell Nucleus/drug effects , Disease Models, Animal , Brain Ischemia/metabolism , Brain Ischemia/drug therapy , Brain Ischemia/pathology , Cell Line , Active Transport, Cell Nucleus/drug effects
4.
Exp Cell Res ; 438(2): 114050, 2024 May 15.
Article En | MEDLINE | ID: mdl-38663474

Myocardial infarction (MI) is a potentially fatal disease that causes a significant number of deaths worldwide. The strategy of increasing fatty acid oxidation in myocytes is considered a therapeutic avenue to accelerate metabolism to meet energy demands. We conducted the study aiming to investigate the effect of KN-93, which induces histone deacetylase (HDAC)4 shuttling to the nucleus, on fatty acid oxidation and the expression of related genes. A mouse model of myocardial infarction was induced by isoprenaline administration. Heart damage was assessed by the detection of cardiac injury markers. The level of fatty acid oxidation level was evaluated by testing the expression of related genes. Both immunofluorescence and immunoblotting in the cytosol or nucleus were utilized to observe the distribution of HDAC4. The interaction between HDAC4 and specificity protein (SP)1 was confirmed by co-immunoprecipitation. The acetylation level of SP1 was tested after KN-93 treatment and HDAC4 inhibitor. Oxygen consumption rate and immunoblotting experiments were used to determine whether the effect of KN-93 on increasing fatty acid oxidation is through HDAC4 and SP1. Administration of KN-93 significantly reduced cardiac injury in myocardial infarction and promoted fatty acid oxidation both in vitro and in vivo. KN-93 was shown to mediate nuclear translocation of HDAC4. HDAC4 was found to interact with SP1 and reduce SP1 acetylation. HDAC4 or SP1 inhibitors attenuated the effect of KN-93 on fatty acid oxidation. In conclusion, KN-93 promotes HDAC4 translocation to the nucleus, thereby potentially enhancing fatty acid oxidation by SP1.


Cell Nucleus , Fatty Acids , Histone Deacetylases , Myocardial Infarction , Oxidation-Reduction , Animals , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Fatty Acids/metabolism , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Mice , Oxidation-Reduction/drug effects , Cell Nucleus/metabolism , Male , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Mice, Inbred C57BL , Humans , Sp1 Transcription Factor/metabolism , Sp1 Transcription Factor/genetics , Acetylation/drug effects , Active Transport, Cell Nucleus/drug effects
5.
Biochem Biophys Res Commun ; 599: 1-8, 2022 04 09.
Article En | MEDLINE | ID: mdl-35158201

Acute lung injury (ALI) is a significant cause of morbidity and mortality worldwide. To search for a new treatment for acute lung injury, we investigated the effect of escitalopram on lipopolysaccharide (LPS)-induced ALI. Our results showed that escitalopram inhibited salt-inducible kinase 2 (SIK2) activity (IC50 = 6.36 ± 0.93 µM) and triggered histone deacetylase 4 (HDAC4) dephosphorylation. Following its dephosphorylation, HDAC4 translocated into the nucleus, promoted deacetylation and cytoplasmic shuttling of p65, thus inhibited LPS-induced pro-inflammatory cytokine production. Moreover, escitalopram markedly ameliorated the inflammatory responses, reduced neutrophils infiltration and attenuated LPS-induced pulmonary injury in mice. Taken together, we identified a previously unexplored role for escitalopram in SIK2/HDAC4/NF-κB pathway, therefore escitalopram may be considered as a new treatment for ALI.


Acute Lung Injury/drug therapy , Escitalopram/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Active Transport, Cell Nucleus/drug effects , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Animals , Drug Repositioning , Histone Deacetylases/metabolism , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/metabolism , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RAW 264.7 Cells , Signal Transduction/drug effects
6.
Oxid Med Cell Longev ; 2022: 2265725, 2022.
Article En | MEDLINE | ID: mdl-35198094

Oxidative stress plays a critical role in age-related macular degeneration (AMD), and epithelial-mesenchymal transition (EMT) is involved in this process. The aim of this study was to investigate the protective effects of luteolin, a natural flavonoid with strong antioxidant activity, on H2O2-induced EMT in ARPE-19 cells. ARPE-19 cells were incubated with H2O2 at 200 µΜ to induce oxidative stress-associated injury. Cell viability assay showed that luteolin at 20 and 40 µM significantly promoted cell survival in H2O2-treated ARPE-19 cells. Luteolin also markedly protected ARPE-19 cells from H2O2-induced apoptosis. Cell migration assay presented that luteolin significantly reduced H2O2-induced migration in APRE-19 cells. EMT in ARPE-19 cells was detected by western blotting and immunofluorescence. The results showed that H2O2 significantly upregulated the expression of α-SMA and vimentin and downregulated the expression of ZO-1 and E-cadherin, while cells pretreated with luteolin showed a reversal. Meanwhile, the assessment of effects of luteolin on the Nrf2 pathway indicated that luteolin promoted Nrf2 nuclear translocation and upregulated the expressions of HO-1 and NQO-1. In addition, luteolin significantly increased the activities of SOD and GSH-PX and decreased intracellular levels of ROS and MDA in H2O2-treated ARPE-19 cells. Meanwhile, we observed that the expression of TGF-ß2, p-AKT, and p-GSK-3ß was upregulated in H2O2-treated ARPE-19 cells and downregulated in luteolin-treated cells, revealing that luteolin inhibited the activation of the AKT/GSK-3ß pathway. However, these effects of luteolin were all annulled by transfecting ARPE-19 cells with Nrf2 siRNA. Our current data collectively indicated that inhibition of luteolin on EMT was induced by oxidative injury in ARPE-19 cell through the Nrf2 and AKT/GSK-3ß pathway, suggesting that luteolin could be a potential drug for the treatment of dry AMD.


Epithelial-Mesenchymal Transition/drug effects , Glycogen Synthase Kinase 3 beta/metabolism , Luteolin/pharmacology , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Active Transport, Cell Nucleus/drug effects , Antioxidants/metabolism , Cell Line , Cell Movement/drug effects , Cell Nucleus/metabolism , Humans , Hydrogen Peroxide/toxicity , Retinal Pigment Epithelium/cytology , Signal Transduction/drug effects
7.
Leukemia ; 36(3): 601-612, 2022 03.
Article En | MEDLINE | ID: mdl-35091658

Novel targeted therapeutics alone or in rational combinations are likely to dominate the future management of various hematological neoplasms. However, the challenges currently faced are the molecular heterogeneity in driver lesions and genetic plasticity leading to multiple resistance pathways. Thus, progress has overall been gradual. For example, despite the advent of targeted agents against actionable drivers like FLT3 in acute myeloid leukemia (AML), the prognosis remains suboptimal in newly diagnosed and dismal in the relapsed/refractory (R/R) setting, due to other molecular abnormalities contributing to inherent and acquired treatment resistance. Nuclear export inhibitors are of keen interest because they can inhibit several active tumorigenic processes simultaneously and also synergize with other targeted drugs and chemotherapy. XPO1 (or CRM1, chromosome maintenance region 1) is one of the most studied exportins involved in transporting critical cargoes, including tumor suppressor proteins like p27, p53, and RB1. Apart from the TSP cargo transport and its role in drug resistance, XPO1 inhibition results in retention of master transcription factors essential for cell differentiation, cell survival, and autophagy, rendering cells more susceptible to the effects of other antineoplastic agents, including targeted therapies. This review will dissect the role of XPO1 inhibition in hematological neoplasms, focusing on mechanistic insights gleaned mainly from work with SINE compounds. Future potential combinatorial strategies will be discussed.


Active Transport, Cell Nucleus/drug effects , Antineoplastic Agents/pharmacology , Hematologic Neoplasms/drug therapy , Animals , Antineoplastic Agents/therapeutic use , Drug Discovery , Hematologic Neoplasms/metabolism , Humans , Karyopherins/metabolism , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Molecular Targeted Therapy , Receptors, Cytoplasmic and Nuclear/metabolism , Exportin 1 Protein
8.
Comput Math Methods Med ; 2022: 3293054, 2022.
Article En | MEDLINE | ID: mdl-35096128

OBJECTIVE: To investigate the effect of dapagliflozin (DAPA) on cardiac hypertrophy induced by type 2 diabetes mellitus (T2DM) and its mechanism. METHODS: SD rats with T2DM were divided into a T2DM group (n = 6) and DAPA group (n = 6). They were, respectively, fed with the same amount of normal saline and 1 mg/kg DAPA. The control group (n = 6) was also fed with normal saline. The hearts were tested by the application of echocardiography and hemodynamics. Subsequently, fasting blood glucose (FBG), serum total cholesterol (TC), and triglyceride (TG) as well as interleukin- (IL-) 10, IL-6, and tumor necrosis factor (TNF)-α in serum were tested. H&E and Masson staining was performed to observe the degree of cardiac tissue lesions, and expression of atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP), calpain-1, p-IκBα, and p65 in myocardial tissue was tested by qRT-PCR and Western blot. RESULTS: Compared with the control group, rats in the T2DM group exhibited significant diabetic symptoms: FBG was significantly elevated, and the levels of TC, TG, IL-6, and TNF-α were significantly increased, while the levels of IL-10 and the calpain activity were evidently decreased. However, DAPA treatment could improve the above changes. At the same time, the damage and fibrosis of the heart tissue in the DAPA group were markedly improved. Additionally, the mRNA expression of ANP and BNP in myocardial tissue of the DAPA group was markedly increased. And DAPA could inhibit the expression of p-IκBα/IκBα in the cytoplasm and p65 in the nucleus as well as the expression of calpain-1 in myocardial tissue. CONCLUSION: DAPA treatment ameliorates the cardiac hypertrophy caused by T2DM by decreasing body blood glucose, while reducing the expression of calpain-1 in cardiomyocytes and inhibiting the nuclear translocation of NF-κB.


Benzhydryl Compounds/pharmacology , Calpain/antagonists & inhibitors , Cardiomegaly/drug therapy , Cardiomegaly/etiology , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Diabetic Cardiomyopathies/drug therapy , Glucosides/pharmacology , NF-kappa B/metabolism , Active Transport, Cell Nucleus/drug effects , Animals , Blood Glucose/metabolism , Calpain/metabolism , Cardiomegaly/metabolism , Computational Biology , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/metabolism , Diabetic Cardiomyopathies/metabolism , Diabetic Cardiomyopathies/pathology , Inflammation Mediators/blood , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Rats , Rats, Sprague-Dawley , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Streptozocin/toxicity
9.
Mol Biol Rep ; 49(2): 1151-1159, 2022 Feb.
Article En | MEDLINE | ID: mdl-35013863

BACKGROUND: Despite commonly use for treatment of type II diabetes, possible effects of glipizide on nuclear transport and DNA damage in cells are unknown. Since clinical response of glipizide may change with aging, the aim of the study was to investigate the effect of glipizide by comparing mature and senescent adipocytes. METHODS AND RESULTS: The effects of glipizide were investigated in 3T3-L1 adipocytes. Effective and lethal doses were determined by real-time monitoring iCELLigence system. Comet assay was performed to determine DNA damage and quantitative PCR was conducted to detect gene expression levels. RAN expressions were found to be up regulated in mature 180 µM glipizide treated adipocytes compared to control group (p < 0.05); whereas down regulated in senescent 180 µM glipizide treated adipocytes compared to their control adipocytes (p < 0.05). Olive Tail Moment values were significantly higher in mature 180 µM glipizide treated adipocytes (MTG) and senescent 180 µM glipizide treated adipocytes (STG) comparing their untreated controls (p < 0.001 and p < 0.001 respectively). Also class 5 comets that shows severe DNA damage were found to be higher in both MTG and STG groups than their controls (p < 0.001 and p < 0.001, respectively). OTM values were higher in STG than MTG (p < 0.001). CONCLUSIONS: This is the first study that reports glipizide caused DNA damage increasing with senescence in adipocytes. As a response to glipizide treatment Ran gene expression increased in mature; and decreased in senescent adipocytes. Further studies are needed to reveal the effect of glipizide on DNA and nuclear interactions in molecular level.


Active Transport, Cell Nucleus/drug effects , DNA Damage/drug effects , Glipizide/pharmacology , 3T3-L1 Cells/drug effects , Active Transport, Cell Nucleus/physiology , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Cell Differentiation , DNA Damage/genetics , Glipizide/adverse effects , Glipizide/metabolism , Mice
10.
Cell Death Dis ; 13(1): 45, 2022 01 10.
Article En | MEDLINE | ID: mdl-35013112

PHY34 is a synthetic small molecule, inspired by a compound naturally occurring in tropical plants of the Phyllanthus genus. PHY34 was developed to have potent in vitro and in vivo anticancer activity against high grade serous ovarian cancer (HGSOC) cells. Mechanistically, PHY34 induced apoptosis in ovarian cancer cells by late-stage autophagy inhibition. Furthermore, PHY34 significantly reduced tumor burden in a xenograft model of ovarian cancer. In order to identify its molecular target/s, we undertook an unbiased approach utilizing mass spectrometry-based chemoproteomics. Protein targets from the nucleocytoplasmic transport pathway were identified from the pulldown assay with the cellular apoptosis susceptibility (CAS) protein, also known as CSE1L, representing a likely candidate protein. A tumor microarray confirmed data from mRNA expression data in public databases that CAS expression was elevated in HGSOC and correlated with worse clinical outcomes. Overexpression of CAS reduced PHY34 induced apoptosis in ovarian cancer cells based on PARP cleavage and Annexin V staining. Compounds with a diphyllin structure similar to PHY34 have been shown to inhibit the ATP6V0A2 subunit of V(vacuolar)-ATPase. Therefore, ATP6V0A2 wild-type and ATP6V0A2 V823 mutant cell lines were tested with PHY34, and it was able to induce cell death in the wild-type at 246 pM while the mutant cells were resistant up to 55.46 nM. Overall, our data demonstrate that PHY34 is a promising small molecule for cancer therapy that targets the ATP6V0A2 subunit to induce autophagy inhibition while interacting with CAS and altering nuclear localization of proteins.


Antineoplastic Agents/pharmacology , Autophagy/drug effects , Cell Nucleus/metabolism , Cellular Apoptosis Susceptibility Protein/metabolism , Cystadenocarcinoma, Serous/metabolism , Ovarian Neoplasms/metabolism , Proton-Translocating ATPases/antagonists & inhibitors , Active Transport, Cell Nucleus/drug effects , Antineoplastic Agents/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Cellular Apoptosis Susceptibility Protein/genetics , Cystadenocarcinoma, Serous/drug therapy , Cystadenocarcinoma, Serous/pathology , Female , Humans , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Phyllanthus/chemistry , Prognosis
11.
Cell Death Dis ; 13(1): 39, 2022 01 10.
Article En | MEDLINE | ID: mdl-35013128

Bladder cancer is a highly heterogeneous and aggressive malignancy with a poor prognosis. EGF/EGFR activation causes the detachment of SHC-binding protein 1 (SHCBP1) from SHC adapter protein 1 (SHC1), which subsequently translocates into the nucleus and promotes cancer development via multiple signaling pathways. However, the role of the EGF-SHCBP1 axis in bladder cancer progression remains unexplored. Herein, we report that SHCBP1 is upregulated in bladder cancer tissues and cells, with cytoplasmic or nuclear localization. Released SHCBP1 responds to EGF stimulation by translocating into the nucleus following Ser273 phosphorylation. Depletion of SHCBP1 reduces EGF-induced cell migration and invasiveness of bladder cancer cells. Mechanistically, SHCBP1 binds to RACGAP1 via its N-terminal domain of amino acids 1 ~ 428, and this interaction is enhanced following EGF treatment. Furthermore, SHCBP1 facilitates cell migration by inhibiting RACGAP-mediated GTP-RAC1 inactivation, whose activity is indispensable for cell movement. Collectively, we demonstrate that the EGF-SHCBP1-RACGAP1-RAC1 axis acts as a novel regulatory mechanism of bladder cancer progression, which offers a new clinical therapeutic strategy to combat bladder cancer.


Cell Nucleus/metabolism , Epidermal Growth Factor/metabolism , GTPase-Activating Proteins/metabolism , Shc Signaling Adaptor Proteins/metabolism , Urinary Bladder Neoplasms/metabolism , rac1 GTP-Binding Protein/metabolism , Active Transport, Cell Nucleus/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation , Disease Progression , Epidermal Growth Factor/pharmacology , Humans , Hydrolysis , Protein Binding , Signal Transduction , Src Homology 2 Domain-Containing, Transforming Protein 1/metabolism , Urinary Bladder Neoplasms/pathology
12.
Gene ; 815: 146178, 2022 Mar 20.
Article En | MEDLINE | ID: mdl-34995733

Frailty develops due to multiple factors, such as sarcopenia, chronic pain, and dementia. Go-sha-jinki-Gan (GJG) is a traditional Japanese herbal medicine used for age-related symptoms. We have reported that GJG improved sarcopenia, chronic pain, and central nervous system function through suppression of tumor necrosis factor-alpha (TNF-α) production. In the present study, GJG was found to reduce the production of TNF-α in the soleus muscle of senescence-accelerated mice at 12 weeks and 36 weeks. GJG did not change the differentiation of C2C12 cells with 2% horse serum. GJG significantly decreased the expression of Muscle atrophy F-box protein (MAFbx) induced by TNF-α in C2C12 cells on real-time PCR. TNF-α significantly decreased the expression of PGC-1α and negated the enhancing effect of GJG for the expression of PGC-1α on digital PCR. Examining 20 chemical compounds derived from GJG, cinnamaldehyde from cinnamon bark and Chikusetsusaponin V (CsV) from Achyrantes Root dose-dependently decreased the production of TNF-⍺ in RAW264.7 cells stimulated by LPS. CsV inhibited the nuclear translocation of nuclear factor-kappa B (NF-κB) p65 in RAW264.7 cells. CsV showed low permeability using Caco-2 cells. However, the plasma concentration of CsV was detected from 30 min to 6 h and peaked at 1 h in the CD1 (ICR) mice after a single dose of GJG. In 8-week-old SAMP8 mice fed 4% (w/w) GJG from one week to four weeks, the plasma CsV concentration ranged from 0.0500 to 10.0 ng/mL. The evidence that CsV plays an important role in various anti-aging effects of GJG via suppression of TNF-⍺ expression is presented.


Aging/drug effects , Drugs, Chinese Herbal/pharmacology , Saponins/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Active Transport, Cell Nucleus/drug effects , Administration, Oral , Animals , Dose-Response Relationship, Drug , Drug Stability , Drugs, Chinese Herbal/chemistry , Male , Mice , Mice, Inbred ICR , Muscle Proteins/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , RAW 264.7 Cells , SKP Cullin F-Box Protein Ligases/metabolism , Saponins/administration & dosage , Saponins/blood , Solubility , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/pharmacology
13.
Lab Invest ; 102(3): 272-280, 2022 03.
Article En | MEDLINE | ID: mdl-34799662

Patients with chronic obstructive pulmonary disease (COPD) are characterized by an imbalance between oxidant enzymes and antioxidant enzymes. In the present study, we explored the protective effect of vitamin E on COPD and the underlying mechanisms. Targets of vitamin E were predicted by bioinformatics analysis. After establishing cigarette smoke (CS)-induced COPD rats, the expression levels of epidermal growth factor receptor (EGFR), cyclooxygenase 2 (COX2), and transcriptional activity of signal transducer and activator of transcription 3 (STAT3) were measured. Additionally, the effects of vitamin E on CS-induced COPD were explored by assessing inflammation, the reactive oxygen species (ROS), the activity of superoxide dismutase (SOD) and the content of malondialdehyde (MDA), viability of human bronchial epithelioid (HBE) cells, and the expression of EGFR/MAPK pathway-related factors after loss- and gain- function assays. Vitamin E alleviated COPD. Vitamin E inhibited MAPK signaling pathway through decreasing EGFR expression. Additionally, vitamin E suppressed CS-induced HBE cell damage. Functionally, vitamin E attenuated CS-induced inflammation, apoptosis, and ROS by inhibiting the EGFR/MAPK axis, thereby inhibiting COX2-mediated p-STAT3 nuclear translocation. Moreover, overexpression of COX2 attenuated the protective effect of vitamin E on COPD rats. The present study shows that vitamin E inhibits the expression of COX2 by negatively regulating the EGFR/MAPK pathway, thereby inhibiting the translocation of phosphorylated STAT3 to the nucleus and relieving COPD.


Cyclooxygenase 2/metabolism , ErbB Receptors/metabolism , MAP Kinase Signaling System/drug effects , Pulmonary Disease, Chronic Obstructive/prevention & control , STAT3 Transcription Factor/metabolism , Vitamin E/pharmacology , Active Transport, Cell Nucleus/drug effects , Animals , Bronchi/cytology , Cell Line , Cell Survival/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Malondialdehyde/metabolism , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/metabolism , Rats , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism , Vitamins/pharmacology
14.
Chem Biol Interact ; 350: 109702, 2021 Dec 01.
Article En | MEDLINE | ID: mdl-34648812

Solid tumors often exhibit hypoxia in their centers, which has been associated with a marked reduction in the sensitivity of the tumor cells to anti-tumor and chemotherapeutic interventions. Here, we found that the occurrence and progress of hypoxic insensitivity to paclitaxel in non-small cell lung cancer (NSCLC) are closely associated with the HIF-1α pathway. The HIF-1α protein upregulated the expression of adipose differentiation-related protein (ADRP), fatty acid synthase (FASN), and sterol regulatory element binding protein 1(SREBP1), while simultaneously downregulating carnitine palmitoyltransferase 1 (CPT1), thereby leading to a more pronounced uptake of lipids and reduced oxidation of fatty acids. Diminished levels of fatty acids led to reduced Wnt pathway activation and ß-catenin nuclear translocation, leading to G2/M cell cycle arrest. In this study, FV-429, a derivative of the natural flavonoid wogonin, reprogrammed metabolism of cancer cells and decreased fatty acid levels. Moreover, paclitaxel-induced G2/M phase arrest in hypoxia-resistant NSCLC was hampered but FV-429 improved the sensitivity of these cancer cells to paclitaxel. FV-429 activated and modulated fatty acid metabolism in NSCLC cells, significantly reduced levels of fatty acids within cells and increased the oxidation of these fatty acids. The results of our study demonstrated that FV-429 could reshape fatty acid metabolism in hypoxia-induced paclitaxel-resistant NSCLC and enhance the sensitivity of NSCLC cells to paclitaxel through G2/M phase arrest deterioration, by inactivating the Wnt pathway, and suggested the possibility of using FV-429 as a promising candidate therapeutic agent for advanced NSCLC.


Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Flavonoids/administration & dosage , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Paclitaxel/administration & dosage , A549 Cells , Active Transport, Cell Nucleus/drug effects , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/toxicity , Antineoplastic Combined Chemotherapy Protocols/toxicity , Cell Line, Tumor , Drug Resistance, Neoplasm , Drug Synergism , Fatty Acids/metabolism , Flavonoids/toxicity , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Paclitaxel/toxicity , Tumor Hypoxia/drug effects , Wnt Signaling Pathway/drug effects , Xenograft Model Antitumor Assays , beta Catenin/metabolism
15.
Int J Mol Sci ; 22(19)2021 Sep 29.
Article En | MEDLINE | ID: mdl-34638882

Persistent inflammatory reactions in microglial cells are strongly associated with neurodegenerative pathogenesis. Additionally, geranylgeraniol (GGOH), a plant-derived isoprenoid, has been found to improve inflammatory conditions in several animal models. It has also been observed that its chemical structure is similar to that of the side chain of menaquinone-4, which is a vitamin K2 sub-type that suppresses inflammation in mouse-derived microglial cells. In this study, we investigated whether GGOH has a similar anti-inflammatory effect in activated microglial cells. Particularly, mouse-derived MG6 cells pre-treated with GGOH were exposed to lipopolysaccharide (LPS). Thereafter, the mRNA levels of pro-inflammatory cytokines were determined via qRT-PCR, while protein expression levels, especially the expression of NF-κB signaling cascade-related proteins, were determined via Western blot analysis. The distribution of NF-κB p65 protein was also analyzed via fluorescence microscopy. Thus, it was observed that GGOH dose-dependently suppressed the LPS-induced increase in the mRNA levels of Il-1ß, Tnf-α, Il-6, and Cox-2. Furthermore, GGOH inhibited the phosphorylation of TAK1, IKKα/ß, and NF-κB p65 proteins as well as NF-κB nuclear translocation induced by LPS while maintaining IκBα expression. We showed that GGOH, similar to menaquinone-4, could alleviate LPS-induced microglial inflammation by targeting the NF-kB signaling pathway.


Diterpenes/pharmacology , Inflammation/prevention & control , Microglia/drug effects , NF-kappa B/metabolism , Signal Transduction/drug effects , Active Transport, Cell Nucleus/drug effects , Animals , Blotting, Western , Cell Line , Cytokines/genetics , Cytokines/metabolism , Gene Expression/drug effects , Inflammation/chemically induced , Lipopolysaccharides , Mice , Microglia/cytology , Microglia/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factor RelA/metabolism
16.
Nat Commun ; 12(1): 6211, 2021 10 27.
Article En | MEDLINE | ID: mdl-34707094

Ribosomal biogenesis has been studied by biochemical, genetic and electron microscopic approaches, but live cell data on the in vivo kinetics are still missing. Here we analyse the export kinetics of the large ribosomal subunit (pre-60S particle) through single NPCs in human cells. We established a stable cell line co-expressing Halo-tagged eIF6 and GFP-fused NTF2 to simultaneously label pre-60S particles and NPCs, respectively. By combining single molecule tracking and super resolution confocal microscopy we visualize the dynamics of single pre-60S particles during export through single NPCs. For export events, maximum particle accumulation is found in the centre of the pore, while unsuccessful export terminates within the nuclear basket. The export has a single rate limiting step and a duration of ∼24 milliseconds. Only about 1/3 of attempted export events are successful. Our results show that the mass flux through a single NPC can reach up to ~125 MDa·s-1 in vivo.


Nuclear Pore/metabolism , Ribosome Subunits, Large, Eukaryotic/metabolism , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/ethics , Cell Nucleus/metabolism , Eukaryotic Initiation Factors/genetics , Eukaryotic Initiation Factors/metabolism , Fatty Acids, Unsaturated/pharmacology , HeLa Cells , Humans , Microscopy, Confocal , Nucleocytoplasmic Transport Proteins/genetics , Nucleocytoplasmic Transport Proteins/metabolism , Organelle Biogenesis , Pregnancy Proteins/genetics , Pregnancy Proteins/metabolism , Single Molecule Imaging
17.
Viruses ; 13(10)2021 10 15.
Article En | MEDLINE | ID: mdl-34696514

Nuclear transport and vesicle trafficking are key cellular functions involved in the pathogenesis of RNA viruses. Among other pleiotropic effects on virus-infected host cells, ivermectin (IVM) inhibits nuclear transport mechanisms mediated by importins and atorvastatin (ATV) affects actin cytoskeleton-dependent trafficking controlled by Rho GTPases signaling. In this work, we first analyzed the response to infection in nasopharyngeal swabs from SARS-CoV-2-positive and -negative patients by assessing the gene expression of the respective host cell drug targets importins and Rho GTPases. COVID-19 patients showed alterations in KPNA3, KPNA5, KPNA7, KPNB1, RHOA, and CDC42 expression compared with non-COVID-19 patients. An in vitro model of infection with Poly(I:C), a synthetic analog of viral double-stranded RNA, triggered NF-κB activation, an effect that was halted by IVM and ATV treatment. Importin and Rho GTPases gene expression was also impaired by these drugs. Furthermore, through confocal microscopy, we analyzed the effects of IVM and ATV on nuclear to cytoplasmic importin α distribution, alone or in combination. Results showed a significant inhibition of importin α nuclear accumulation under IVM and ATV treatments. These findings confirm transcriptional alterations in importins and Rho GTPases upon SARS-CoV-2 infection and point to IVM and ATV as valid drugs to impair nuclear localization of importin α when used at clinically-relevant concentrations.


Active Transport, Cell Nucleus/drug effects , Atorvastatin/pharmacology , COVID-19 Drug Treatment , Ivermectin/pharmacology , SARS-CoV-2/drug effects , alpha Karyopherins/metabolism , A549 Cells , Actin Cytoskeleton/drug effects , Animals , Antiviral Agents/pharmacology , Cell Line, Tumor , Chlorocebus aethiops , Drug Repositioning , HeLa Cells , Humans , NF-kappa B/metabolism , Vero Cells , rho GTP-Binding Proteins/metabolism
18.
Signal Transduct Target Ther ; 6(1): 299, 2021 08 09.
Article En | MEDLINE | ID: mdl-34373448

Aflatoxin exposure is a crucial factor in promoting the development of primary hepatocellular carcinoma (HCC) in individuals infected with the hepatitis virus. However, the molecular pathways leading to its bioactivation and subsequent toxicity in hepatocytes have not been well-defined. Here, we carried out a genome-wide CRISPR-Cas9 genetic screen to identify aflatoxin B1 (AFB1) targets. Among the most significant hits was the aryl hydrocarbon receptor (AHR), a ligand-binding transcription factor regulating cell metabolism, differentiation, and immunity. AHR-deficient cells tolerated high concentrations of AFB1, in which AFB1 adduct formation was significantly decreased. AFB1 triggered AHR nuclear translocation by directly binding to its N-terminus. Furthermore, AHR mediated the expression of P450 induced by AFB1. AHR expression was also elevated in primary tumor sections obtained from AFB1-HCC patients, which paralleled the upregulation of PD-L1, a clinically relevant immune regulator. Finally, anti-PD-L1 therapy exhibited greater efficacy in HCC xenografts derived from cells with ectopic expression of AHR. These results demonstrated that AHR was required for the AFB1 toxicity associated with HCC, and implicate the immunosuppressive regimen of anti-PD-L1 as a therapeutic option for the treatment of AFB1-associated HCCs.


B7-H1 Antigen/genetics , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Receptors, Aryl Hydrocarbon/genetics , Active Transport, Cell Nucleus/drug effects , Aflatoxin B1/pharmacology , Animals , B7-H1 Antigen/antagonists & inhibitors , CRISPR-Cas Systems/drug effects , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/therapy , Carcinoma, Hepatocellular/virology , Cell Line, Tumor , Genome, Human/drug effects , Hepatitis Viruses/pathogenicity , Hepatocytes/drug effects , Humans , Liver Neoplasms/pathology , Liver Neoplasms/therapy , Liver Neoplasms/virology , Mice , Xenograft Model Antitumor Assays
19.
Int J Mol Sci ; 22(13)2021 Jun 22.
Article En | MEDLINE | ID: mdl-34206543

Aberrant nuclear protein transport, often observed in cancer, causes mislocalization-dependent inactivation of critical cellular proteins. Earlier we showed that overexpression of exportin 1 is linked to higher grade and Gleason score in metastatic castration resistant prostate cancer (mCRPC). We also showed that a selective inhibitor of nuclear export (SINE) selinexor and second generation eltanexor (KPT-8602) could suppress mCRPC growth, reduce androgen receptor (AR), and re-sensitize to androgen deprivation therapy. Here we evaluated the combination of KPT-8602 with PARP inhibitors (PARPi) olaparib, veliparib and rucaparib in 22rv1 mCRPC cells. KPT-8602 synergized with PARPi (CI < 1) at pharmacologically relevant concentrations. KPT-8602-PARPi showed superior induction of apoptosis compared to single agent treatment and caused up-regulation of pro-apoptotic genes BAX, TP53 and CASPASE 9. Mechanistically, KPT-8602-PARPi suppressed AR, ARv7, PSA and AR targets FOXA1 and UBE2C. Western blot analysis revealed significant down-regulation of AR, ARv7, UBE2C, SAM68, FOXA1 and upregulation of cleaved PARP and cleaved CASPASE 3. KPT-8602 with or without olaparib was shown to reduce homologous recombination-regulated DNA damage response targets including BRCA1, BRCA2, CHEK1, EXO1, BLM, RAD51, LIG1, XRCC3 and RMI2. Taken together, this study revealed the therapeutic potential of a novel combination of KPT-8602 and PARP inhibitors for the treatment of mCRPC.


Active Transport, Cell Nucleus/drug effects , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Synergism , Humans , Male , Models, Biological , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology
20.
In Vitro Cell Dev Biol Anim ; 57(7): 676-684, 2021 Aug.
Article En | MEDLINE | ID: mdl-34312803

Mammary epithelial cells have been suggested to be central to control the expansion and remodeling of mammary gland. Wnt/ß-catenin signaling modulates cell fate in animals throughout their life span, and represents indispensable roles in tissue homeostasis, cell renewal, and regeneration in organs. Here, we utilized the small molecule 6-bromoindirubin-3'-oxime (BIO), an activator of Wnt/ß-catenin signaling, and investigated whether Wnt/ß-catenin signaling regulated the proliferation and acinus-like structure formation of goat mammary epithelial cells (GMECs). We showed that isolated GMECs displayed the typical epithelial cobblestone morphology and expressed specific markers of mammary epithelial cells. BIO inhibited the proliferation of GMECs and decreased the expression of proliferation marker c-myc and cell cycle protein cyclin D1. However, the ability of GMECs to form spheroids was accelerated, and the level of E-cadherin mRNA was upregulated with BIO treatment. E-cadherin showed a bright cytomembrane with DMSO treatment, yet E-cadherin was present in cytomembrane and cytoplasm in GMECs with BIO treatment. Meanwhile, BIO increased the protein level of ß-catenin and enhanced the translocation of ß-catenin into the nucleus in GMECs. Furthermore, the mRNA level of Axin2 was also upregulated. This study suggested that Wnt/ß-catenin signaling may play an important role in the proliferation and the acinus-like formation of GMECs.


Epithelial Cells/ultrastructure , Mammary Glands, Animal/cytology , Wnt Signaling Pathway , beta Catenin/metabolism , Acinar Cells , Active Transport, Cell Nucleus/drug effects , Animals , Cell Proliferation/drug effects , Cells, Cultured , Female , Gene Expression Regulation/drug effects , Goats , Indoles/pharmacology , Oximes/pharmacology , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/physiology
...