Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 281
Filter
1.
J Cell Mol Med ; 28(16): e70015, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39159189

ABSTRACT

Pyrocurzerenone is a natural compound found in Curcuma zedoaria and Chloranthus serratus. However, the anticancer effect of pyrocurzerenone in oral cancer remains unclear. Using the MTT assay, wound healing assay, transwell assay and western blot analysis, we investigated the impact of pyrocurzerenone on antimetastatic activity, as well as the critical signalling pathways that underlie the processes of oral cancer cell lines SCC-9, SCC-1 and SAS in this work. Our findings suggested that pyrocurzerenone inhibits cell migration and invasion ability in oral cancer cell lines. Furthermore, phosphorylation of ERK1/2 had significant inhibitory effects in SCC-9 and SCC-1 cell lines. Combining ERK1/2 inhibitors with pyrocurzerenone decreased the migration and invasion activity of SCC-9 and SCC-1 cell lines. We also found that the expressed level of cathepsin S decreased under pyrocurzerenone treatment. This study showed that pyrocurzerenone reduced ERK1/2 expression of the proteins and cathepsin S, suggesting that it could be a valuable treatment to inhibit human oral cancer cell metastasis.


Subject(s)
Cathepsins , Cell Movement , Mouth Neoplasms , Humans , Mouth Neoplasms/pathology , Mouth Neoplasms/drug therapy , Mouth Neoplasms/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cathepsins/metabolism , Cathepsins/antagonists & inhibitors , Phosphorylation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Neoplasm Metastasis , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 1/genetics , Neoplasm Invasiveness , Cell Proliferation/drug effects , Signal Transduction/drug effects
2.
Cell Death Dis ; 15(8): 600, 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39160159

ABSTRACT

Crizotinib carries an FDA hepatotoxicity warning, yet analysis of the FAERS database suggests that the severity of its hepatotoxicity risks, including progression to hepatitis and liver failure, might be underreported. However, the underlying mechanism remains poorly understood, and effective intervention strategies are lacking. Here, mRNA-sequencing analysis, along with KEGG and GO analyses, revealed that DEGs linked to Crizotinib-induced hepatotoxicity predominantly associate with the ferroptosis pathway which was identified as the principal mechanism behind Crizotinib-induced hepatocyte death. Furthermore, we found that ferroptosis inhibitors, namely Ferrostatin-1 and Deferoxamine mesylate, significantly reduced Crizotinib-induced hepatotoxicity and ferroptosis in both in vivo and in vitro settings. We have also discovered that overexpression of AAV8-mediated Nrf2 could mitigate Crizotinib-induced hepatotoxicity and ferroptosis in vivo by restoring the imbalance in glutathione metabolism, iron homeostasis, and lipid peroxidation. Additionally, both Stat1 deficiency and the Stat1 inhibitor NSC118218 were found to reduce Crizotinib-induced ferroptosis. Mechanistically, Crizotinib induces the phosphorylation of Stat1 at Ser727 but not Tyr701, promoting the transcriptional inhibition of Nrf2 expression after its entry into the nucleus to promote ferroptosis. Meanwhile, we found that MgIG and GA protected against hepatotoxicity to counteract ferroptosis without affecting or compromising the anti-cancer activity of Crizotinib, with a mechanism potentially related to the Stat1/Nrf2 pathway. Overall, our findings identify that the phosphorylation activation of Stat1 Ser727, rather than Tyr701, promotes ferroptosis through transcriptional inhibition of Nrf2, and highlight MgIG and GA as potential therapeutic approaches to enhance the safety of Crizotinib-based cancer therapy.


Subject(s)
Chemical and Drug Induced Liver Injury , Crizotinib , Ferroptosis , NF-E2-Related Factor 2 , STAT1 Transcription Factor , Ferroptosis/drug effects , NF-E2-Related Factor 2/metabolism , Humans , Animals , Crizotinib/pharmacology , Crizotinib/adverse effects , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Chemical and Drug Induced Liver Injury/genetics , STAT1 Transcription Factor/metabolism , STAT1 Transcription Factor/genetics , Mice , Signal Transduction/drug effects , Male , Phenylenediamines/pharmacology , Mice, Inbred C57BL , Hepatocytes/metabolism , Hepatocytes/drug effects , Phosphorylation/drug effects
3.
Dev Cell ; 59(16): 2189-2202.e8, 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39089249

ABSTRACT

Targeting wild-type epidermal growth factor receptor (EGFR) using tyrosine kinase inhibitors (TKIs) never achieved its purported success in cancers such as head and neck squamous cell carcinoma, which are largely EGFR-dependent. We had previously shown that exceptional responders to TKIs have a genetic aberration that results in overexpression of an EGFR splice variant, isoform D (IsoD). IsoD lacks an integral transmembrane and kinase domain and is secreted in extracellular vesicles (EVs) in TKI-sensitive patient-derived cultures. Remarkably, the exquisite sensitivity to TKIs could be transferred to TKI-resistant tumor cells, and IsoD protein in the EV is necessary and sufficient to transfer the phenotype in vitro and in vivo across multiple models and drugs. This drug response requires an intact endocytic mechanism, binding to full-length EGFR, and signaling through Src-phosphorylation within the endosomal compartment. We propose a therapeutic strategy using EVs containing EGFR IsoD as a co-drug to expand the use of TKI therapy to EGFR-driven cancers.


Subject(s)
Carcinoma, Squamous Cell , ErbB Receptors , Extracellular Vesicles , Protein Isoforms , Protein Kinase Inhibitors , ErbB Receptors/metabolism , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Humans , Protein Kinase Inhibitors/pharmacology , Extracellular Vesicles/metabolism , Protein Isoforms/metabolism , Protein Isoforms/genetics , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Animals , Cell Line, Tumor , Mice , Signal Transduction/drug effects , Phosphorylation/drug effects , Drug Resistance, Neoplasm/drug effects , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/genetics , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology , Squamous Cell Carcinoma of Head and Neck/genetics
4.
PLoS One ; 19(8): e0309324, 2024.
Article in English | MEDLINE | ID: mdl-39163364

ABSTRACT

Plasma levels of branched-chain amino acids and their metabolites, the branched-chain ketoacids are increased in insulin resistance. Our previous studies showed that leucine and its metabolite KIC suppress insulin-stimulated glucose uptake in L6 myotubes along with the activation of the S6K1-IRS-1 pathway. Because other tissue and fiber types can be differentially regulated by KIC, we analyzed the effect of KIC gavage on whole-body insulin sensitivity and insulin signaling in vivo. We hypothesized that KIC gavage would reduce whole-body insulin sensitivity and increase S6K1-IRS-1 phosphorylation in various tissues and muscle fibers. Five-week-old male Sprague-Dawley rats were starved for 24 hours and then gavaged with 0.75ml/100g of water, leucine (22.3g/L) or KIC (30g/L) twice, ten minutes apart. They were then euthanized at different time points post-gavage (0.5-3h), and muscle, liver, and heart tissues were dissected. Other sets of gavaged animals underwent an insulin tolerance test. Phosphorylation (ph) of S6K1 (Thr389), S6 (Ser235/6) and IRS-1 (Ser612) was increased at 30 minutes post leucine gavage in skeletal muscles irrespective of fiber type. Ph-S6 (Ser235/6) was also increased in liver and heart 30 minutes after leucine gavage. KIC gavage increased ph-S6 (Ser235/6) in the liver. Neither Leucine nor KIC influenced whole-body insulin tolerance, nor ph-Akt (Ser473) in skeletal muscle and heart. BCKD-E1 α abundance was highest in the heart and liver, while ph-BCKD-E1 α (Ser293) was higher in the gastrocnemius and EDL compared to the soleus. Our data suggests that only leucine activates the S6K1-IRS-1 signaling axis in skeletal muscle, liver and heart, while KIC only does so in the liver. The effect of leucine and KIC on the S6K1-IRS-1 signaling pathway is uncoupled from whole-body insulin sensitivity. These results suggest that KIC and leucine may not induce insulin resistance, and the contributions of other tissues may regulate whole-body insulin sensitivity in response to leucine/KIC gavage.


Subject(s)
Insulin Resistance , Insulin , Keto Acids , Leucine , Rats, Sprague-Dawley , Signal Transduction , Animals , Male , Leucine/metabolism , Leucine/pharmacology , Signal Transduction/drug effects , Insulin/metabolism , Insulin/blood , Rats , Phosphorylation/drug effects , Keto Acids/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/drug effects , Insulin Receptor Substrate Proteins/metabolism , Liver/metabolism , Liver/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/drug effects
5.
Sci Transl Med ; 16(759): eadg1915, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39110779

ABSTRACT

Severe alcohol-associated hepatitis (AH) is a life-threatening form of alcohol-associated liver disease. Liver neutrophil infiltration is a hallmark of AH, yet the effects of alcohol on neutrophil functions remain elusive. Identifying therapeutic targets to reduce neutrophil-mediated liver damage is essential. Bruton's tyrosine kinase (BTK) plays an important role in neutrophil development and function; however, the role of BTK in AH is unknown. Using RNA sequencing of circulating neutrophils, we found an increase in Btk expression (P = 0.05) and phosphorylated BTK (pBTK) in patients with AH compared with healthy controls. In vitro, physiologically relevant doses of alcohol resulted in a rapid, TLR4-mediated induction of pBTK in neutrophils. In a preclinical model of AH, administration of a small-molecule BTK inhibitor (evobrutinib) or myeloid-specific Btk knockout decreased proinflammatory cytokines and attenuated neutrophil-mediated liver damage. We found that pBTK was essential for alcohol-induced bone marrow granulopoiesis and liver neutrophil infiltration. In vivo, BTK inhibition or myeloid-specific Btk knockout reduced granulopoiesis, circulating neutrophils, liver neutrophil infiltration, and liver damage in a mouse model of AH. Mechanistically, using liquid chromatography-tandem mass spectrometry, we identified CD84 as a kinase target of BTK, which is involved in granulopoiesis. In vitro, CD84 promoted alcohol-induced interleukin-1ß and tumor necrosis factor-α in primary human neutrophils, which was inhibited by CD84-blocking antibody treatment. Our findings define the role of BTK and CD84 in regulating neutrophil inflammation and granulopoiesis, with potential therapeutic implications in AH.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase , Liver Diseases, Alcoholic , Neutrophils , Agammaglobulinaemia Tyrosine Kinase/metabolism , Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Animals , Humans , Neutrophils/metabolism , Neutrophils/drug effects , Liver Diseases, Alcoholic/metabolism , Liver Diseases, Alcoholic/pathology , Protein Kinase Inhibitors/pharmacology , Mice , Male , Liver/pathology , Liver/metabolism , Liver/drug effects , Granulocytes/metabolism , Granulocytes/drug effects , Mice, Inbred C57BL , Antigens, CD/metabolism , Mice, Knockout , Toll-Like Receptor 4/metabolism , Phosphorylation/drug effects
6.
Int J Mol Sci ; 25(15)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39125998

ABSTRACT

In the pathological process of Alzheimer's disease, neuronal cell death is closely related to the accumulation of reactive oxygen species. Our previous studies have found that oxidative stress can activate microtubule affinity-regulating kinases, resulting in elevated phosphorylation levels of tau protein specifically at the Ser262 residue in N1E-115 cells that have been subjected to exposure to hydrogen peroxide. This process may be one of the pathogenic mechanisms of Alzheimer's disease. Vitamin E is a fat-soluble, naturally occurring antioxidant that plays a crucial role in biological systems. This study aimed to examine the probable processes that contribute to the inhibiting effect on the abnormal phosphorylation of tau protein and the neuroprotective activity of a particular type of vitamin E, α-tocotrienol. The experimental analysis revealed that α-tocotrienol showed significant neuroprotective effects in the N1E-115 cell line. Our data further suggest that one of the mechanisms underlying the neuroprotective effects of α-tocotrienol may be through the inhibition of microtubule affinity-regulated kinase activation, which significantly reduces the oxidative stress-induced aberrant elevation of p-Tau (Ser262) levels. These results indicate that α-tocotrienol may represent an intriguing strategy for treating or preventing Alzheimer's disease.


Subject(s)
Neurons , Neuroprotective Agents , Oxidative Stress , Vitamin E , tau Proteins , tau Proteins/metabolism , Phosphorylation/drug effects , Neurons/drug effects , Neurons/metabolism , Vitamin E/pharmacology , Vitamin E/analogs & derivatives , Neuroprotective Agents/pharmacology , Animals , Mice , Oxidative Stress/drug effects , Alzheimer Disease/metabolism , Alzheimer Disease/drug therapy , Cell Line, Tumor , Tocotrienols
7.
Neurosci Lett ; 837: 137923, 2024 Aug 10.
Article in English | MEDLINE | ID: mdl-39106918

ABSTRACT

Caffeine, a nonselective adenosine receptor antagonist, is the major component of coffee and the most consumed psychostimulant at nontoxic doses in the world. It has been identified that caffeine consumption reduces the risk of several neurological diseases. However, the mechanisms by which it impacts the pathophysiology of neurological diseases remain to be elucidated. In this study, we investigated whether caffeine exerts anti-inflammatory effects on lipopolysaccharide (LPS)-induced inflammation and depression in vivo and explored the potential mechanism of caffeine through LPS-induced brain injury. Adult male Sprague-Dawley (SD) rats were intraperitoneal injected with various concentrations of LPS to induce the neuroinflammation and depressive-like behavior. Then SD rats were treated with caffeine in the presence or absence of LPS. Open-filed and closed-field tests were applied to detect the behaviors of SD rats, while western blot was performed to measure the phosphorylation level of protein kinase B (p-AKT) and nuclear factor κB (NF-κB) in the cortex after caffeine was orally administered. Our findings indicated that caffeine markedly improved the neuroinflammation and depressive-like behavior of LPS-treated SD rats. Mechanistic investigations demonstrated that caffeine down-regulated the expression of p-AKT and NF-κB in LPS-induced SD rats cortex. Taken together, these results indicated that caffeine, a potential agent for preventing inflammatory diseases, may suppress LPS-induced inflammatory and depressive responses by regulating AKT phosphorylation and NF-κB.


Subject(s)
Caffeine , Depression , Lipopolysaccharides , NF-kappa B , Neuroinflammatory Diseases , Proto-Oncogene Proteins c-akt , Rats, Sprague-Dawley , Animals , NF-kappa B/metabolism , Male , Caffeine/pharmacology , Caffeine/therapeutic use , Proto-Oncogene Proteins c-akt/metabolism , Depression/drug therapy , Depression/chemically induced , Depression/metabolism , Rats , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/chemically induced , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Phosphorylation/drug effects , Inflammation/metabolism , Inflammation/drug therapy , Inflammation/chemically induced
8.
Shanghai Kou Qiang Yi Xue ; 33(3): 229-234, 2024 Jun.
Article in Chinese | MEDLINE | ID: mdl-39104334

ABSTRACT

PURPOSE: To investigate the inhibitory effect of sodium cantharidate (SCA) on human tongue squamous cell carcinoma CAL27 cells and its mechanism. METHODS: CAL27 cells were pretreated with different concentrations of SCA. Cell viability was analyzed by CCK-8 method. The migration and invasion of CAL27 cells were measured by scratch test and Transwell chamber, and the apoptosis rate was measured by flow cytometry. p53 protein and its phosphorylation sites Ser33, Ser37, Ser46, expression of BCL-2, BAX, and cleaved caspase 3 in CAL27 cells were detected by Western blot. Statistical analysis was performed with Graphpad Prism 9.0 software package. RESULTS: Compared with the blank control group, the proliferation, migration and invasion of CAL27 cells in sodium cantharidate group were significantly decreased, and the apoptosis rate was significantly increased(P<0.01) in a dose-dependent manner. The expression of p53 protein and its phosphorylation sites Ser33, Ser37, Ser46 protein was significantly up-regulated(P<0.05 or P<0.01). The expression of BCL-2 protein was down-regulated and the expression of BAX protein was significantly up-regulated(P<0.05 or P<0.01). The ratio of BCL-2/BAX was significantly decreased and the expression of cleaved caspase 3 protein was significantly up-regulated(P<0.05 or P<0.01). CONCLUSIONS: SCA can inhibit the proliferation, migration and invasion of human tongue squamous cell carcinoma CAL27 cells. It also down-regulates the ratio of BCL-2/BAX and up-regulates the expression of cleaved caspase 3 protein by regulating the phosphorylation of p53 protein, which induces apoptosis.


Subject(s)
Apoptosis , Carcinoma, Squamous Cell , Cell Movement , Cell Proliferation , Tongue Neoplasms , Tumor Suppressor Protein p53 , bcl-2-Associated X Protein , Humans , Tongue Neoplasms/drug therapy , Tongue Neoplasms/metabolism , Tongue Neoplasms/genetics , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Apoptosis/drug effects , Cell Proliferation/drug effects , Cell Movement/drug effects , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Phosphorylation/drug effects , bcl-2-Associated X Protein/metabolism , bcl-2-Associated X Protein/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Caspase 3/metabolism , Cell Survival/drug effects , Neoplasm Invasiveness
9.
Int J Biol Macromol ; 276(Pt 1): 133834, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39002899

ABSTRACT

IL-2 regulates the immune response by interacting with different IL-2 receptor (IL-2R) subunits. High dose of IL-2 binds to IL-2Rßγc heterodimer, which induce various side effects while activating immune function. Disrupting IL-2 and IL-2R interactions can block IL-2 mediated immune response. Here, we used a computational approach to de novo design mini-binder proteins against IL-2R ß chain (IL-2Rß) to block IL-2 signaling. The hydrophobic region where IL-2 binds to IL-2Rß was selected and the promising binding mode was broadly explored. Three mini-binders with amino acid numbers ranging from 55 to 65 were obtained and binder 1 showed the best effects in inhibiting CTLL-2 cells proliferation and STAT5 phosphorylation. Molecular dynamics simulation showed that the binding of binder 1 to IL-2Rß was stable; the free energy of binder1/IL-2Rß complex was lower, indicating that the affinity of binder 1 to IL-2Rß was higher than that of IL-2. Free energy decomposition suggested that the ARG35 and ARG131 of IL-2Rß might be the key to improve the affinity of binder. Our efforts provided new insights in developing of IL-2R blocker, offering a potential strategy for ameliorating the side effects of IL-2 treatment.


Subject(s)
Interleukin-2 Receptor beta Subunit , Interleukin-2 , Molecular Dynamics Simulation , Protein Binding , Interleukin-2 Receptor beta Subunit/metabolism , Interleukin-2 Receptor beta Subunit/chemistry , Interleukin-2/metabolism , Interleukin-2/chemistry , Humans , Cell Proliferation/drug effects , STAT5 Transcription Factor/metabolism , Phosphorylation/drug effects , Animals , Molecular Docking Simulation
10.
Int J Biol Macromol ; 276(Pt 1): 133734, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39002903

ABSTRACT

In this study, Kraft lignin was modified by ammonium dihydrogen phosphate (ADP) and urea for achieving phosphorylation and carbamylation, aiming to protect wood against biological and fire attack. Scots pine (Pinus sylvestris L.) sapwood was impregnated with a water solution containing Kraft lignin, ADP, and urea, followed by heat treatment at 150 °C, resulting in changes in the properties of the Kraft lignin as well as the wood matrix. Infrared spectroscopy, 13C cross-polarisation magic-angle-spinning (MAS) nuclear magnetic resonance (NMR), and direct excitation single-pulse 31P MAS NMR analyses suggested the grafting reaction of phosphate and carbamylate groups onto the hydroxyl groups of Kraft lignin. Scanning electron microscopy with energy dispersive X-ray spectroscopy indicated that the condensed Kraft lignin filled the lumen as well as partially penetrating the wood cell wall. The modified Kraft lignin imparted fire-retardancy and increased char residue to the wood at elevated temperature, as confirmed by limiting oxygen index, microscale combustion calorimetry, and thermogravimetric analysis. The modified wood exhibited superior resistance against mold and decay fungi attack under laboratory conditions. The modified wood had a similar modulus of elasticity to the unmodified wood, while experiencing a reduction in the modulus of rupture.


Subject(s)
Lignin , Pinus sylvestris , Wood , Lignin/chemistry , Lignin/pharmacology , Wood/chemistry , Pinus sylvestris/chemistry , Phosphorylation/drug effects , Fires , Magnetic Resonance Spectroscopy
11.
Eur J Pharmacol ; 979: 176852, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-39067565

ABSTRACT

Macrophages polarize into alternatively activated M2 macrophages through interleukin (IL)-4, and they express high levels of arginase-1, which promotes anti-inflammatory responses. Several studies have confirmed the anti-inflammatory effects of cyclin-dependent kinase (CDK) 8/19 inhibition, and hence, numerous CDK8/19 inhibitors, such as BRD6989, have been developed. However, the effects of CDK8/19 inhibitors on arginase-1 expression in macrophages have not yet been elucidated. This study investigated the effects of CDK8/19 inhibitor on arginase-1 expression in IL-4-activated macrophages. The results showed that BRD6989 increased arginase-1 expression transcriptionally in murine peritoneal macrophages and the murine macrophage cell line RAW264.7 in an IL-4-dependent manner. In addition, the results indicated that BRD6989 enhances signal transducer and activator of transcription (STAT) 6 phosphorylation. Meanwhile, BRD6989 exhibited the capability to activate p38 mitogen-activated protein kinase (MAPK) even in the absence of IL-4 stimulation. Moreover, we observed that a p38 MAPK inhibitor suppressed the BRD6989-induced increase in arginase-1 expression. Besides, BRD6989 increased the surface expression of CD206, an M2 macrophage marker. Thus, this study demonstrated for the first time that CDK8/19 inhibition increases arginase-1 expression, suggesting that this mechanism involves the activation of STAT6 and p38 MAPK. This finding implies that CDK8/19 inhibition may facilitate the production of anti-inflammatory M2 macrophages.


Subject(s)
Arginase , Cyclin-Dependent Kinase 8 , Cyclin-Dependent Kinases , Interleukin-4 , STAT6 Transcription Factor , p38 Mitogen-Activated Protein Kinases , Animals , Arginase/metabolism , Arginase/antagonists & inhibitors , STAT6 Transcription Factor/metabolism , Mice , p38 Mitogen-Activated Protein Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , RAW 264.7 Cells , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , Interleukin-4/metabolism , Cyclin-Dependent Kinase 8/antagonists & inhibitors , Cyclin-Dependent Kinase 8/metabolism , Protein Kinase Inhibitors/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Phosphorylation/drug effects , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Enzyme Activation/drug effects , Flavonoids , Piperidines , Cyclin-Dependent Kinase 9
12.
Nutrients ; 16(14)2024 Jul 22.
Article in English | MEDLINE | ID: mdl-39064812

ABSTRACT

Manuka honey (MH) exhibits potential antitumor activity in preclinical models of a number of human cancers. Treatment in vitro with MH at concentrations ranging from 0.3 to 5.0% (w/v) led to significant dose-dependent inhibition of proliferation of human breast cancer MCF-7 cells, but anti-proliferative effects of MH were less pronounced in MDA-MB-231 breast cancer cells. Effects of MH were also tested on non-malignant human mammary epithelial cells (HMECs) at 2.5% w/v, and it was found that MH reduced the proliferation of MCF-7 cells but not that of HMECs. Notably, the antitumor activity of MH was in the range of that exerted by treatment of MCF-7 cells with the antiestrogen tamoxifen. Further, MH treatment stimulated apoptosis of MCF-7 cells in vitro, with most cells exhibiting acute and significant levels of apoptosis that correlated with PARP activation. Additionally, the effects of MH induced the activation of AMPK and inhibition of AKT/mTOR downstream signaling. Treatment of MCF7 cells with increased concentrations of MH induced AMPK phosphorylation in a dose-dependent manner that was accompanied by inhibition of phosphorylation of AKT and mTOR downstream effector protein S6. In addition, MH reduced phosphorylated STAT3 levels in vitro, which may correlate with MH and AMPK-mediated anti-inflammatory properties. Further, in vivo, MH administered alone significantly inhibited the growth of established MCF-7 tumors in nude mice by 84%, resulting in an observable reduction in tumor volume. Our findings highlight the need for further research into the use of natural compounds, such as MH, for antitumor efficacy and potential chemoprevention and investigation of molecular pathways underlying these actions.


Subject(s)
Apoptosis , Breast Neoplasms , Cell Proliferation , Honey , Humans , Breast Neoplasms/pathology , Breast Neoplasms/drug therapy , Female , Animals , Apoptosis/drug effects , MCF-7 Cells , Cell Proliferation/drug effects , Signal Transduction/drug effects , Mice , Xenograft Model Antitumor Assays , Mice, Nude , Leptospermum/chemistry , TOR Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Antineoplastic Agents/pharmacology , STAT3 Transcription Factor/metabolism , Disease Progression , AMP-Activated Protein Kinases/metabolism , Cell Line, Tumor , Phosphorylation/drug effects
13.
Molecules ; 29(14)2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39064957

ABSTRACT

Cholangiocarcinoma (CCA) is a cancer with a poor prognosis due to difficulties in diagnosis and limited treatment options, highlighting the urgent need for new targeted therapies. In a clinical setting, we found that leukotriene levels in bile were higher than in serum. Immunohistochemical analysis of surgically resected samples also revealed that CysLT receptor 1 (CysLTR1) was more highly expressed in CCA than in normal bile duct tissue, prompting us to investigate leukotriene as a potential therapeutic target in CCA. In vitro studies using CCA cell lines expressing CysLTR1 showed that leukotriene D4, a major ligand of CysLTR1, promoted cell proliferation, with increased phosphorylation of AKT and extracellular signal-regulated kinase 1/2 (ERK1/2). Additionally, treatment with two clinically available anti-allergic drugs-zileuton, an inhibitor of CysLT formation, and montelukast, a CysLTR1 inhibitor-had inhibitory effects on cell proliferation and migratory capacity, accompanied by the reduced phosphorylation of AKT and ERK1/2. Furthermore, the simultaneous administration of both drugs synergistically enhanced the inhibitory effect on cell proliferation. Our study suggests that use of these drugs may represent a novel approach to treat CCA through drug repositioning.


Subject(s)
Bile Duct Neoplasms , Cell Proliferation , Cholangiocarcinoma , Hydroxyurea , Leukotriene Antagonists , Quinolines , Receptors, Leukotriene , Sulfides , Humans , Cholangiocarcinoma/drug therapy , Cholangiocarcinoma/metabolism , Cholangiocarcinoma/pathology , Cell Proliferation/drug effects , Receptors, Leukotriene/metabolism , Leukotriene Antagonists/pharmacology , Leukotriene Antagonists/therapeutic use , Cell Line, Tumor , Bile Duct Neoplasms/drug therapy , Bile Duct Neoplasms/metabolism , Bile Duct Neoplasms/pathology , Sulfides/pharmacology , Quinolines/pharmacology , Hydroxyurea/analogs & derivatives , Hydroxyurea/pharmacology , Hydroxyurea/therapeutic use , Acetates/pharmacology , Acetates/chemistry , Male , Cyclopropanes/pharmacology , Cyclopropanes/therapeutic use , Cell Movement/drug effects , Female , Middle Aged , Proto-Oncogene Proteins c-akt/metabolism , Disease Progression , Leukotrienes/metabolism , Phosphorylation/drug effects , Aged , Leukotriene D4/metabolism , MAP Kinase Signaling System/drug effects
14.
Biomed Pharmacother ; 177: 117074, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38972149

ABSTRACT

Glioma, a common and highly malignant central nervous system tumor, markedly influences patient prognosis via interactions with glioma-associated macrophages. Previous research has revealed the anticancer potential of ß-mangostin, a xanthone derivative obtained from the mangosteen fruit. This research investigated the role of ß-mangostin on microglia in the glioma microenvironment and evaluated the efficacy of ß-mangostin combined with anti-PD-1 antibody (αPD-1) in glioma-bearing mice. The results showed that, ß-mangostin attenuated M2 polarization in BV2 cells and promoted M1-related interleukin (IL)-1ß and IL-6 secretion, thereby inhibiting glioma invasion. In addition, ß-mangostin improved the anti-glioma effects of αPD-1 and increased CD8+T cell and M1-type microglia infiltration. Mechanistically, ß-mangostin bound to the stimulator of interferon genes (STING) protein, which is crucial for the anti-tumor innate immune response, and promoted STING phosphorylation in microglia, both in vivo and in vitro. These results provide insights into its mode of action and supporting further investigation into ß-mangostin as a therapeutic agent.


Subject(s)
Glioma , Membrane Proteins , Microglia , Xanthones , Xanthones/pharmacology , Animals , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Glioma/drug therapy , Glioma/pathology , Glioma/metabolism , Mice , Membrane Proteins/metabolism , Cell Line, Tumor , Mice, Inbred C57BL , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Brain Neoplasms/metabolism , Tumor Microenvironment/drug effects , Male , Humans , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Phosphorylation/drug effects
15.
Cell Rep ; 43(7): 114487, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38996071

ABSTRACT

The integrated stress response (ISR) is activated in response to intrinsic and extrinsic stimuli, playing a role in tumor progression and drug resistance. The regulatory role and mechanism of ISR in liver cancer, however, remain largely unexplored. Here, we demonstrate that OTU domain-containing protein 3 (OTUD3) is a deubiquitylase of eukaryotic initiation factor 2α (eIF2α), antagonizing ISR and suppressing liver cancer. OTUD3 decreases interactions between eIF2α and the kinase EIF2ΑK3 by removing K27-linked polyubiquitylation on eIF2α. OTUD3 deficiency in mice leads to enhanced ISR and accelerated progression of N-nitrosodiethylamine-induced hepatocellular carcinoma. Additionally, decreased OTUD3 expression associated with elevated eIF2α phosphorylation correlates with the progression of human liver cancer. Moreover, ISR activation due to decreased OTUD3 expression renders liver cancer cells resistant to sorafenib, while the combined use of the ISR inhibitor ISRIB significantly improves their sensitivity to sorafenib. Collectively, these findings illuminate the regulatory mechanism of ISR in liver cancer and provide a potential strategy to counteract sorafenib resistance.


Subject(s)
Drug Resistance, Neoplasm , Liver Neoplasms , Sorafenib , Ubiquitin-Specific Proteases , Sorafenib/pharmacology , Liver Neoplasms/pathology , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Liver Neoplasms/genetics , Animals , Humans , Drug Resistance, Neoplasm/drug effects , Mice , Ubiquitin-Specific Proteases/metabolism , Ubiquitin-Specific Proteases/genetics , Disease Progression , Stress, Physiological/drug effects , Cell Line, Tumor , Ubiquitination/drug effects , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Eukaryotic Initiation Factor-2/metabolism , Phosphorylation/drug effects , Mice, Inbred C57BL
16.
Exp Cell Res ; 441(1): 114154, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38996959

ABSTRACT

Platelet-derived growth factor (PDGF) is one of the most important cytokines associated with pulmonary vascular remodeling in pulmonary arterial hypertension (PAH). PDGF receptor (PDGFR) inhibition exerted therapeutic effects on PAH in clinical trials, but serious side effects warrant the withdrawal of existing drugs. In this study, a novel highly selective PDGFR inhibitor WQ-C-401 was developed, and its effects on PDGFR signaling pathway and pulmonary vascular remodeling in PAH were investigated. Cell proliferation assays and Western blot analysis of PDGFRα/ß phosphorylation showed that WQ-C-401 inhibited PDGFR-mediated cell proliferation assay and suppressed PDGFR phosphorylation in a concentration-dependent manner. DiscoverX's KinomeScanTM technology confirmed the good kinome selectivity of WQ-C-401 (S score (1) of PDGFR = (0.01)). In monocrotaline (MCT)-induced PAH rats, intragastric administration of WQ-C-401 (25, 50, 100 mg/kg/d) or imatinib (50 mg/kg/d, positive control) significantly decreased right ventricular systolic pressure (RVSP). Histological analysis demonstrated that WQ-C-401 inhibited pulmonary vascular remodeling by reducing muscularization and fibrosis, as well as alleviated right ventricular hypertrophy in MCT-treated rats. In addition, WQ-C-401 suppressed MCT-induced cell hyperproliferation and CD68+ macrophage infiltration around the pulmonary artery. In vitro, WQ-C-401 inhibited PDGF-BB-induced proliferation and migration of human pulmonary arterial smooth muscle cells (PASMCs). Moreover, Western blot analysis showed that WQ-C-401 concertration-dependently inhibited PDGF-BB-induced phosphorylation of ERK1/2 and PDGFRß Y751, decreased collagen Ⅰ synthesis and increased alpha smooth muscle actin (α-SMA) expression in PASMCs. Collectively, our results suggest that WQ-C-401 is a selective and potent PDGFR inhibitor which could be a promising drug for the therapeutics of PAH by preventing pulmonary vascular remodeling.


Subject(s)
Cell Proliferation , Monocrotaline , Pulmonary Arterial Hypertension , Rats, Sprague-Dawley , Vascular Remodeling , Animals , Vascular Remodeling/drug effects , Rats , Cell Proliferation/drug effects , Male , Pulmonary Arterial Hypertension/drug therapy , Pulmonary Arterial Hypertension/chemically induced , Pulmonary Arterial Hypertension/metabolism , Pulmonary Arterial Hypertension/pathology , Humans , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Receptors, Platelet-Derived Growth Factor/metabolism , Phosphorylation/drug effects , Pulmonary Artery/drug effects , Pulmonary Artery/pathology , Pulmonary Artery/metabolism , Signal Transduction/drug effects , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/prevention & control , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/metabolism , Protein Kinase Inhibitors/pharmacology , Receptor, Platelet-Derived Growth Factor beta/metabolism , Receptor, Platelet-Derived Growth Factor beta/antagonists & inhibitors
17.
Int J Mol Sci ; 25(13)2024 Jun 24.
Article in English | MEDLINE | ID: mdl-39000009

ABSTRACT

Yohimbine (YHB) has been reported to possess anti-inflammatory, anticancer, and cardiac function-enhancing properties. Additionally, it has been reported to inhibit the proliferation, migration, and neointimal formation of vascular smooth muscle cells (VSMCs) induced by platelet-derived growth factor (PDGF) stimulation by suppressing the phospholipase C-gamma 1 pathway. However, the transcriptional regulatory mechanism of YHB controlling the behavior of VSMCs is not fully understood. In this study, YHB downregulated the expression of cell cycle regulatory proteins, such as proliferating cell nuclear antigen (PCNA), cyclin D1, cyclin-dependent kinase 4 (CDK4), and cyclin E, by modulating the transcription factor FOXO3a in VSMCs induced by PDGF. Furthermore, YHB decreased p-38 and mTOR phosphorylation in a dose-dependent manner. Notably, YHB significantly reduced the phosphorylation at Y397 and Y925 sites of focal adhesion kinase (FAK), and this effect was greater at the Y925 site than Y397. In addition, the expression of paxillin, a FAK-associated protein known to bind to the Y925 site of FAK, was significantly reduced by YHB treatment in a dose-dependent manner. A pronounced reduction in the migration and proliferation of VSMCs was observed following co-treatment of YHB with mTOR or p38 inhibitors. In conclusion, this study shows that YHB inhibits the PDGF-induced proliferation and migration of VSMCs by regulating the transcription factor FOXO3a and the mTOR/p38/FAK signaling pathway. Therefore, YHB may be a potential therapeutic candidate for preventing and treating cardiovascular diseases such as atherosclerosis and vascular restenosis.


Subject(s)
Cell Movement , Cell Proliferation , Forkhead Box Protein O3 , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Platelet-Derived Growth Factor , Yohimbine , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/drug effects , Forkhead Box Protein O3/metabolism , Cell Proliferation/drug effects , Cell Movement/drug effects , Animals , Platelet-Derived Growth Factor/metabolism , Platelet-Derived Growth Factor/pharmacology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/drug effects , Phosphorylation/drug effects , Yohimbine/pharmacology , Rats , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Focal Adhesion Kinase 1/metabolism , Cells, Cultured , Paxillin/metabolism , Rats, Sprague-Dawley , Male
18.
Phytother Res ; 38(8): 4307-4320, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38973353

ABSTRACT

American ginseng (AG) has been reported to have anti-inflammatory effects in many diseases, but the key molecules and mechanisms are unclear. This study aims to evaluate the anti-inflammatory mechanism of AG and identify the key molecules by in vivo and in vitro models. Zebrafish was employed to assess the anti-inflammatory properties of AG and the compounds. Metabolomics was utilized to identify potential anti-inflammatory molecules in AG, while molecular dynamics simulations were conducted to forecast the interaction capabilities of these compounds with inflammatory targets. Additionally, macrophage cell was employed to investigate the anti-inflammatory mechanisms of the key molecules in AG by enzyme-linked immunosorbent assay and western blotting. Seven potential anti-inflammatory molecules were discovered in AG, with ginsenoside Rg1, ginsenoside Rs3 (G-Rs3), and oleanolic acid exhibiting the strongest affinity for signal transducer and activator of transcription 3. These compounds demonstrated inhibitory effects on macrophage migration in zebrafish models and the ability to regulate ROS levels in both zebrafish and macrophages. The cell experiments found that ginsenoside Rg1, ginsenoside Rs3, and oleanolic acid could promote macrophage M2/M1 polarization ratio and inhibit phosphorylation overexpression of signal transducer and activator of transcription 3. This study revealed the key anti-inflammatory molecules and mechanisms of AG, and provided new evidence of anti-inflammatory for the scientific use of AG.


Subject(s)
Anti-Inflammatory Agents , Ginsenosides , Macrophages , Panax , STAT3 Transcription Factor , Zebrafish , Animals , Panax/chemistry , Anti-Inflammatory Agents/pharmacology , STAT3 Transcription Factor/metabolism , Macrophages/drug effects , Macrophages/metabolism , Ginsenosides/pharmacology , Ginsenosides/chemistry , Phosphorylation/drug effects , RAW 264.7 Cells , Mice , Plant Extracts/pharmacology , Plant Extracts/chemistry , Reactive Oxygen Species/metabolism , Molecular Dynamics Simulation
19.
Sci Rep ; 14(1): 16260, 2024 07 15.
Article in English | MEDLINE | ID: mdl-39009622

ABSTRACT

The aim of this study was to evaluate the effects of C-type natriuretic peptide (CNP) treatment prior to in vitro maturation (IVM) on mitochondria biogenesis in bovine oocyte matured in vitro and explore the related causes. The results showed that treatment with CNP before IVM significantly improved mitochondrial content, elevated the expression of genes related to mitochondria biogenesis, and increased the protein levels of phosphorylation of cAMP-response element binding protein (p-CREB) in bovine oocytes following IVM. However, further studies revealed that treatment with CNP before IVM could not increased the protein levels of p-CREB in bovine oocytes when natriuretic peptide receptor 2 activities was inhibited using the relative specific inhibitor Gö6976. In addition, treatment with CNP before IVM could not improved mitochondrial content or elevated the expression of genes related to mitochondria biogenesis in bovine oocytes when CREB activities was abolished using the specific inhibitor 666-15. In summary, these results provide evidence that treatment of bovine oocytes with CNP before IVM promotes mitochondrial biogenesis in vitro, possibly by activating CREB.


Subject(s)
Cyclic AMP Response Element-Binding Protein , Mitochondria , Natriuretic Peptide, C-Type , Oocytes , Organelle Biogenesis , Animals , Cattle , Natriuretic Peptide, C-Type/pharmacology , Natriuretic Peptide, C-Type/metabolism , Oocytes/metabolism , Oocytes/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Mitochondria/metabolism , Mitochondria/drug effects , Female , In Vitro Oocyte Maturation Techniques/methods , Phosphorylation/drug effects
20.
Eur J Pharmacol ; 979: 176816, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-39038637

ABSTRACT

Alzheimer's disease is a neurodegenerative problem with progressive loss of memory and other cognitive function disorders resulting in the imbalance of neurotransmitter activity and signaling progression, which poses the need of the potential therapeutic target to improve the intracellular signaling cascade brought by kinases. Protein kinase plays a significant and multifaceted role in the treatment of Alzheimer's disease, by targeting pathological mechanisms like tau hyperphosphorylation, neuroinflammation, amyloid-beta production and synaptic dysfunction. In this review, we thoroughly explore the essential protein kinases involved in Alzheimer's disease, detailing their physiological roles, regulatory impacts, and the newest inhibitors and compounds that are progressing into clinical trials. All the findings of studies exhibited the promising role of kinase inhibitors in the management of Alzheimer's disease. However, it still poses the need of addressing current challenges and opportunities involved with this disorder for the future perspective of kinase inhibitors in the management of Alzheimer's disease. Further study includes the development of biomarkers, combination therapy, and next-generation kinase inhibitors with increased potency and selectivity for its future prospects.


Subject(s)
Alzheimer Disease , Protein Kinase Inhibitors , Alzheimer Disease/drug therapy , Humans , Protein Kinase Inhibitors/therapeutic use , Protein Kinase Inhibitors/pharmacology , Animals , tau Proteins/metabolism , tau Proteins/antagonists & inhibitors , Protein Kinases/metabolism , Signal Transduction/drug effects , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Phosphorylation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL