Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 336
Filter
Add more filters

Publication year range
1.
Biochem Biophys Res Commun ; 695: 149451, 2024 Feb 05.
Article in English | MEDLINE | ID: mdl-38176173

ABSTRACT

BACKGROUND/OBJECTIVE: DT-13, the principal active component of Mysidium shortscapes from the Liliaceae family, has garnered substantial interest in cancer therapy owing to its potential anticancer properties. This study investigated the effects of DT-13 on the proliferation and apoptosis of human pancreatic cancer cell lines and aimed to elucidate the underlying mechanisms. METHODS: PANC1 and CFPAC1 cells were exposed to DT-13 and their proliferation was assessed using RTCA and clone formation assays. Apoptotic protein expression was analyzed by western blotting, and apoptotic cells were identified by flow cytometry. RNA was extracted from DT-13 treated and untreated PANC1 cells for RNA sequencing. Differentially expressed genes were identified and subjected to GO bioprocess, KEGG pathway analysis, and western blotting. Finally, to evaluate tumor growth, CFPAC1 cells were subcutaneously injected into BALB/c nude mice. RESULTS: DT-13 inhibited proliferation and induced apoptosis of PANC1 and CFPAC1 cells by activating the AMPK/mTOR pathway and suppressing p70 S6K. Moreover, DT-13 hindered the growth of CFPAC1 xenograft tumors in nude mice. CONCLUSIONS: DT-13 effectively inhibited the growth of human pancreatic cancer cells.


Subject(s)
AMP-Activated Protein Kinases , Pancreatic Neoplasms , Saponins , Animals , Humans , Mice , AMP-Activated Protein Kinases/drug effects , AMP-Activated Protein Kinases/metabolism , Apoptosis , Cell Line, Tumor , Cell Proliferation , Mice, Inbred BALB C , Mice, Nude , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , TOR Serine-Threonine Kinases/drug effects , TOR Serine-Threonine Kinases/metabolism , Xenograft Model Antitumor Assays , Saponins/pharmacology , Saponins/therapeutic use
2.
Biochem Biophys Res Commun ; 704: 149688, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38387327

ABSTRACT

BACKGROUND: Multi-territory perforator flap reconstruction has been proven effective in treating large skin and soft tissue defects in clinical settings. However, in view of that the multi-territory perforator flap is prone to partial postoperative necrosis, increasing its survival is the key to the success of reconstruction. In this study, we aimed to clarify the effect of emodin on multi-territory perforator flap survival. METHODS: Flap survival was assessed by viability area analysis, infrared laser imaging detector, HE staining, immunohistochemistry, and angiography. Western blotting, immunofluorescence assays, and real-time fluorescent quantitative PCR were performed to detect the indicators of oxidative stress, pyroptosis and autophagy. RESULTS: After emodin treatment, the multi-territory perforator flap showed a significantly increased survival rate, which was shown to be closely related to the inhibition of oxidative stress and pyroptosis and enhanced autophagy. Meanwhile, the use of autophagy inhibitor 3 MA was found to reverse the inhibitory effects of emodin on oxidative stress and pyroptosis and weaken the improving effect of emodin on flap survival, suggesting that autophagy plays a critical role in emodin-treated flaps. Interestingly, our mechanistic investigations revealed that the positive effect of emodin on multi-territory perforator flap was attributed to the mTOR-ULK1 signaling pathway activation. CONCLUSIONS: Emodin can inhibit oxidative stress and pyroptosis by activating autophagy via the mTOR-ULK1 pathway, thereby improving the multi-territory perforator flap survival.


Subject(s)
Emodin , Perforator Flap , Autophagy/drug effects , Emodin/pharmacology , Oxidative Stress/drug effects , Pyroptosis/drug effects , Signal Transduction , TOR Serine-Threonine Kinases/drug effects , Autophagy-Related Protein-1 Homolog/drug effects
3.
Biochem Biophys Res Commun ; 715: 149999, 2024 Jun 30.
Article in English | MEDLINE | ID: mdl-38678787

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD), a chronic liver condition and metabolic disorder, has emerged as a significant health issue worldwide. D-mannose, a natural monosaccharide widely existing in plants and animals, has demonstrated metabolic regulatory properties. However, the effect and mechanism by which D-mannose may counteract NAFLD have not been studied. In this study, network pharmacology followed by molecular docking analysis was utilized to identify potential targets of mannose against NAFLD, and the leptin receptor-deficient, genetically obese db/db mice was employed as an animal model of NAFLD to validate the regulation of D-mannose on core targets. As a result, 67 targets of mannose are predicted associated with NAFLD, which are surprisingly centered on the mechanistic target of rapamycin (mTOR). Further analyses suggest that mTOR signaling is functionally enriched in potential targets of mannose treating NAFLD, and that mannose putatively binds to mTOR as a core mechanism. Expectedly, repeated oral gavage of supraphysiological D-mannose ameliorates liver steatosis of db/db mice, which is based on suppression of hepatic mTOR signaling. Moreover, daily D-mannose administration reduced hepatic expression of lipogenic regulatory genes in counteracting NAFLD. Together, these findings reveal D-mannose as an effective and potential NAFLD therapeutic through mTOR suppression, which holds translational promise.


Subject(s)
Mannose , Network Pharmacology , Non-alcoholic Fatty Liver Disease , TOR Serine-Threonine Kinases , Animals , Mice , Liver/metabolism , Liver/drug effects , Mannose/pharmacology , Mannose/metabolism , Mice, Inbred C57BL , Molecular Docking Simulation , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/drug effects , TOR Serine-Threonine Kinases/metabolism
4.
Exp Cell Res ; 433(2): 113829, 2023 12 15.
Article in English | MEDLINE | ID: mdl-37879548

ABSTRACT

Chronic subdural hematoma (CSDH) remains a neurosurgical condition and a healthy burden especially in elderly patients. This study focuses on the functions of rapamycin and its related molecular mechanisms in CSDH management. A rat model of CSDH was induced, which developed significant hematoma on day 5 after operation. The rats were treated with rapamycin or atorvastatin, a drug with known effect on hematoma alleviation, or treated with rapamycin and atorvastatin in combination. The atorvastatin or rapamycin treatment reduced the hematoma development, blood-brain barrier permeability, neurological dysfunction in CSDH rats, and the combination treatment showed more pronounced effects. Human brain microvascular endothelial cells hCMEC/D3 were stimulated by hematoma samples to mimic a CSDH condition in vitro. The drug treatments elevated the cell junction-related factors and reduced the pro-inflammatory cytokines both in rat hematoma tissues and in hCMEC/D3 cells. Rapamycin suppressed the mTOR and STAT3 signaling pathways. Overexpression of mTOR or the STAT3 agonist suppressed the alleviating effects of rapamycin on CSDH. In summary, this study demonstrates that rapamycin promotes hematoma resorption and enhances endothelial cell function by suppressing the mTOR/STAT3 signaling.


Subject(s)
Hematoma, Subdural, Chronic , Sirolimus , Aged , Animals , Humans , Rats , Atorvastatin/pharmacology , Atorvastatin/therapeutic use , Endothelial Cells/metabolism , Hematoma, Subdural, Chronic/drug therapy , Hematoma, Subdural, Chronic/metabolism , Signal Transduction , STAT3 Transcription Factor/drug effects , STAT3 Transcription Factor/metabolism , TOR Serine-Threonine Kinases/drug effects , TOR Serine-Threonine Kinases/metabolism , Sirolimus/pharmacology , Sirolimus/therapeutic use
5.
Semin Cancer Biol ; 73: 302-309, 2021 08.
Article in English | MEDLINE | ID: mdl-33152487

ABSTRACT

Research over decades has enabled us in developing a better understanding of the multifaceted and heterogeneous nature of cancer. High-throughput technologies have helped the researchers in unraveling of the underlying mechanisms which centrally regulate cancer onset, metastasis and drug resistance. Our rapidly expanding knowledge about signal transduction cascade has added another layer of complexity to already complicated nature of cancer. Deregulation of cell signaling pathways played a linchpin role in carcinogenesis and metastasis. Cucurbitacins have gained tremendous attention because of their remarkable pharmacological properties and considerable ability to mechanistically modulate myriad of cell signaling pathways in different cancers. In this review, we have attempted to provide a mechanistic and comprehensive analysis of regulation of oncogenic pathways by cucurbitacins in different cancers. We have partitioned this review into separate sections for exclusive analysis of each signaling pathway and critical assessment of the knowledge gaps. In this review, we will summarize most recent and landmark developments related to regulation of Wnt/ß-catenin, JAK/STAT, mTOR, VEGFR, EGFR and Hippo pathway by cucurbitacins. Moreover, we will also address how cucurbitacins regulate DNA damage repair pathway and TRAIL-driven signaling in various cancers. However, there are still outstanding questions related to regulation of SHH/GLI, TGF/SMAD and Notch-driven pathway by cucurbitacins in different cancers. Future studies must converge on the analysis of full-fledge potential of cucurbitacins by in-depth analysis of these pathways and how these pathways can be therapeutically targeted by cucurbitacins.


Subject(s)
Cucurbitacins/pharmacology , Neoplasms , Phytochemicals/pharmacology , Signal Transduction/drug effects , Animals , Humans , Janus Kinases/drug effects , RNA, Untranslated/drug effects , STAT Transcription Factors/drug effects , TNF-Related Apoptosis-Inducing Ligand/drug effects , TOR Serine-Threonine Kinases/drug effects , Wnt Signaling Pathway/drug effects
6.
Int J Exp Pathol ; 103(1): 23-30, 2022 02.
Article in English | MEDLINE | ID: mdl-34802165

ABSTRACT

The aim of this study was to investigate the effect of nucleotide-binding oligomerization domain (NOD)-like receptor family CARD domain containing 5 (NLRC5) in cardiac hypertrophy, and to explore the mechanism implicated in this effect Cardiac hypertrophy was induced in neonatal rat cardiac myocytes using 1 µM of angiotensin II (Ang II) for 12, 24 and 48 h. Overexpression of NLRC5 was induced in H9C2 cells, and the NLRC5 + Ang II-treated cells were exposed to SC9 and 3-methyladenine (3MA). An immunofluorescence assay was used for α-actinin staining, and quantitative real-time reverse transcriptase-polymerase chain reaction (qRT-PCR) was performed for NLRC5, atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP) determination. Western blot analysis was applied to measure the levels of NLRC5, microtubule-associated protein 1A/1B-light chain 3 type I (LC3I), LC3II, sequestosome 1 (p62), protein kinase B (AKT), phosphorylated Akt (pAKT), mammalian target of rapamycin (mTOR) and phosphorylated mTOR (pmTOR). The level of NLRC5 was significantly decreased after Ang II treatment in cardiomyocytes, but the levels of ANP and BNP were increased. Overexpression of NLRC5 reduced the cell size, downregulated the levels of ANP and BNP, increased LC3II / LC3I, but decreased p62 in Ang II-induced cardiomyocyte hypertrophy. In addition, the results from Western blot showed that overexpression of NLRC5 distinctly decreased the ratios of pAKT/AKT and pmTOR/mTOR in cardiomyocyte hypertrophy. SC79 and 3MA significantly downregulated the ratio of LC3I/LC3II but increased the level of p62 in NLRC5 + Ang II-treated cells. These results provide a possible novel therapeutic strategy for cardiac hypertrophy that might be useful in a clinical setting.


Subject(s)
Autophagy/drug effects , Cardiomegaly/metabolism , NLR Proteins/pharmacology , TOR Serine-Threonine Kinases/drug effects , Angiotensin II/pharmacology , Animals , Autophagy/physiology , Myocytes, Cardiac/metabolism , NLR Proteins/metabolism , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats
7.
FASEB J ; 35(4): e21485, 2021 04.
Article in English | MEDLINE | ID: mdl-33709562

ABSTRACT

Cognitive dysfunction often occurs in diabetes mellitus patients. This study aimed to investigate the efficacy of melatonin (MLT) in improving diabetes-associated cognitive decline and the underlying mechanism involved. Type 2 diabetic mice and palmitic acid (PA)-stimulated BV-2 cells were treated by MLT, and the potential mechanisms among MLT, cognition, and autophagy were explored. The results showed that type 2 diabetic mice showed obvious learning and memory impairments in the Morris water maze test compared with normal controls, which could be ameliorated by MLT treatment. Meanwhile, MLT administration significantly improved neuroinflammation and regulated microglial apoptosis. Furthermore, autophagy inhibitor 3-methyladenine (3-MA) increased the microglial inflammation and apoptosis, indicating that the treatment effect of MLT was mediated by autophagy. Lastly, MLT treatment significantly decreased the levels of toll-like receptors 4 (TLR4), phosphorylated-protein kinase B (Akt), and phosphorylated-mechanistic target of rapamycin (mTOR), indicating that blocking TLR4/Akt/mTOR pathway might be an underlying basis for the anti-inflammatory and anti-apoptosis effects of MLT. Collectively, our study suggested that MLT could improve learning and memory in type 2 diabetic mice by activating autophagy via the TLR4/Akt/mTOR pathway, thereby inhibiting neuroinflammation and microglial apoptosis.


Subject(s)
Cognitive Dysfunction/prevention & control , Melatonin/pharmacology , Microglia/drug effects , Proto-Oncogene Proteins c-akt/drug effects , Animals , Anti-Inflammatory Agents/pharmacology , Cognitive Dysfunction/metabolism , Diabetes Mellitus, Experimental/metabolism , Inflammation/metabolism , Male , Mice, Inbred C57BL , Microglia/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/drug effects
8.
Am J Respir Crit Care Med ; 204(3): 312-325, 2021 08 01.
Article in English | MEDLINE | ID: mdl-33784491

ABSTRACT

Rationale: CD148/PTRJ (receptor-like protein tyrosine phosphatase η) exerts antifibrotic effects in experimental pulmonary fibrosis via interactions with its ligand syndecan-2; however, the role of CD148 in human pulmonary fibrosis remains incompletely characterized.Objectives: We investigated the role of CD148 in the profibrotic phenotype of fibroblasts in idiopathic pulmonary fibrosis (IPF).Methods: Conditional CD148 fibroblast-specific knockout mice were generated and exposed to bleomycin and then assessed for pulmonary fibrosis. Lung fibroblasts (mouse lung and human IPF lung), and precision-cut lung slices from human patients with IPF were isolated and subjected to experimental treatments. A CD148-activating 18-aa mimetic peptide (SDC2-pep) derived from syndecan-2 was evaluated for its therapeutic potential.Measurements and Main Results: CD148 expression was downregulated in IPF lungs and fibroblasts. In human IPF lung fibroblasts, silencing of CD148 increased extracellular matrix production and resistance to apoptosis, whereas overexpression of CD148 reversed the profibrotic phenotype. CD148 fibroblast-specific knockout mice displayed increased pulmonary fibrosis after bleomycin challenge compared with control mice. CD148-deficient fibroblasts exhibited hyperactivated PI3K/Akt/mTOR signaling, reduced autophagy, and increased p62 accumulation, which induced NF-κB activation and profibrotic gene expression. SDC2-pep reduced pulmonary fibrosis in vivo and inhibited IPF-derived fibroblast activation. In precision-cut lung slices from patients with IPF and control patients, SDC2-pep attenuated profibrotic gene expression in IPF and normal lungs stimulated with profibrotic stimuli.Conclusions: Lung fibroblast CD148 activation reduces p62 accumulation, which exerts antifibrotic effects by inhibiting NF-κB-mediated profibrotic gene expression. Targeting the CD148 phosphatase with activating ligands such as SDC2-pep may represent a potential therapeutic strategy in IPF.


Subject(s)
Fibroblasts/metabolism , Idiopathic Pulmonary Fibrosis/genetics , Lung/metabolism , Animals , Antibiotics, Antineoplastic/toxicity , Autophagy/drug effects , Autophagy/genetics , Bleomycin/toxicity , Disease Models, Animal , Fibroblasts/drug effects , Fibroblasts/pathology , Humans , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/pathology , In Vitro Techniques , Lung/drug effects , Lung/pathology , Mice , Mice, Knockout , NF-kappa B/drug effects , NF-kappa B/metabolism , Peptide Fragments/pharmacology , Phenotype , Phosphatidylinositol 3-Kinases/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Primary Cell Culture , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism , Signal Transduction , Syndecan-2/pharmacology , TOR Serine-Threonine Kinases/drug effects , TOR Serine-Threonine Kinases/metabolism
9.
J Cell Physiol ; 236(12): 8110-8121, 2021 12.
Article in English | MEDLINE | ID: mdl-34101831

ABSTRACT

Even though aberrant mechanistic target of rapamycin (mTOR) signaling is known to cause cardiomyopathy, its underlying mechanism remains poorly understood. Because augmentation of αB-crystallin and hspB2 was presented in the cortical tubers and lymphangioleiomyomatosis of tuberous sclerosis complex patients, we deciphered the role of αB-crystallin and its adjacent duplicate gene, hspB2, in hyperactive mTOR-induced cardiomyopathy. Cardiac Tsc1 deletion (T1-hKO) caused mouse mTOR activation and cardiomyopathy. Overexpression of αB-crystallin and hspB2 was presented in the hearts of these mice. Knockout of αB-crystallin/hspB2 reversed deficient Tsc1-mediated fetal gene expression, mTOR activation, mitochondrial damage, cardiomyocyte vacuolar degeneration, cardiomyocyte size, and fibrosis of T1-hKO mice. These cardiac-Tsc1; αB-crystallin; hspB2 triple knockout (tKO) mice had improved cardiac function, smaller heart weight to body weight ratio, and reduced lethality compared with T1-hKO mice. Even though activated mTOR suppressed autophagy in T1-hKO mice, ablation of αB-crystallin and hspB2 failed to restore autophagy in tKO mice. mTOR inhibitors suppressed αB-crystallin expression in T1-hKO mice and rat cardiomyocyte line H9C2. Starvation of H9C2 cells activated autophagy and suppressed αB-crystallin expression. Since inhibition of autophagy restored αB-crystallin expression in starved H9C2 cells, autophagy is a negative regulator of αB-crystallin expression. mTOR thus stimulates αB-crystallin expression through suppression of autophagy. In conclusion, αB-crystallin and hspB2 play a pivotal role in Tsc1 knockout-related cardiomyopathy and are therapeutic targets of hyperactive mTOR-associated cardiomyopathy.


Subject(s)
Cardiomyopathies/metabolism , Crystallins/metabolism , HSP27 Heat-Shock Proteins/metabolism , Heat-Shock Proteins/metabolism , Myocytes, Cardiac/metabolism , Animals , Cardiomyopathies/drug therapy , Cardiomyopathies/genetics , HSP27 Heat-Shock Proteins/drug effects , HSP27 Heat-Shock Proteins/genetics , Heat-Shock Proteins/drug effects , MTOR Inhibitors/pharmacology , Mice, Knockout , Myocytes, Cardiac/drug effects , Promoter Regions, Genetic/genetics , TOR Serine-Threonine Kinases/drug effects , TOR Serine-Threonine Kinases/metabolism
10.
Apoptosis ; 26(3-4): 195-208, 2021 04.
Article in English | MEDLINE | ID: mdl-33550458

ABSTRACT

Acute promyelocytic leukemia (APL) is a blood system disease caused by the accumulation of a large number of immature blood cells in bone marrow. Although the introduction of all-trans retinoic acid (ATRA) and arsenic has reached a high level of complete remission rate and 5-year disease-free survival rate, the occurrence of various adverse reactions still severely affects the quality of life of patients. As a natural product, honokiol (HNK) has the advantages of low toxicity and high efficiency, and it is a potential drug for the treatment of cancer. Since cancer cells can escape apoptotic cell death through multiple adaptive mechanisms, HNK, a drug that induces cancer cell death in a nonapoptotic way, has attracted much interest. We found that HNK reduced the viability of human APL cell line (NB4 cells) by inducing paraptosis-like cell death. The process was accompanied by excessive reactive oxygen species (ROS), mitochondrial damage, endoplasmic reticulum stress, and increased microtubule-associated protein 1 light chain 3 (LC3) processing. The inactivation of proteasome activity was the main cause of misfolded and unfolded protein accumulation in endoplasmic reticulum, such as LC3II/I and p62. This phenomenon could be alleviated by adding cycloheximide (CHX), a protein synthesis inhibitor. We found that mTOR signaling pathway participated in paraptosis-like cell death induced by HNK in an autophagy-independent process. Moreover, the mitogen-activated protein kinase (MAPK) signaling pathway induced paraptosis of NB4 cells by promoting endoplasmic reticulum stress. In summary, these findings indicate that paraptosis may be a new way to treat APL, and provide novel insights into the potential mechanism of paraptosis-like cell death.


Subject(s)
Apoptosis/drug effects , Biphenyl Compounds/pharmacology , Leukemia, Promyelocytic, Acute , Lignans/pharmacology , Signal Transduction/drug effects , Biological Products/pharmacology , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Endoplasmic Reticulum Stress/drug effects , Humans , In Vitro Techniques , Leukemia, Promyelocytic, Acute/drug therapy , Leukemia, Promyelocytic, Acute/metabolism , MAP Kinase Signaling System/drug effects , Proteasome Endopeptidase Complex/drug effects , Reactive Oxygen Species/metabolism , TOR Serine-Threonine Kinases/drug effects , TOR Serine-Threonine Kinases/metabolism
11.
BMC Cancer ; 21(1): 125, 2021 Feb 06.
Article in English | MEDLINE | ID: mdl-33549034

ABSTRACT

BACKGROUND: Side population (SP) cells, which have similar features to those of cancer stem cells, show resistance to dexamethasone (Dex) treatment. Thus, new drugs that can be used in combination with Dex to reduce the population of SP cells in multiple myeloma (MM) are required. Diallyl thiosulfinate (DATS, allicin), a natural organosulfur compound derived from garlic, has been shown to inhibit the proliferation of SP cells in MM cell lines. Therefore, we investigated the effect of a combination of DATS and Dex (DAT + Dex) on MM SP cells. METHODS: SP cells were sorted from MM RPMI-8226 and NCI-H929 cell lines using Hoechst 33342-labeled fluorescence-activated cell sorting. The growth of SP cells was evaluated using the cell counting kit-8 assay. Cell cycle and apoptosis assays were conducted using a BD Calibur flow cytometer. miRNA expression was measured using quantitative reverse transcription-polymerase chain reaction. Phosphoinositide 3-kinase (PI3K), phosphorylated AKT (p-AKT), AKT, p-mechanistic target of rapamycin (mTOR), and mTOR levels were measured using western blot analysis. RESULTS: Our results showed that the combination of DATS+Dex inhibited sphere formation, colony formation, and proliferation of MM SP cells by inducing apoptosis and cell cycle arrest in the G1/S phase. In addition, the combination of DATS+Dex promoted miR-127-3p expression and inhibited PI3K, p-AKT, and p-mTOR expression in SP cells. Knockdown of miR-127-3p expression weakened the effect of DATS+Dex on cell proliferation, colony formation, apoptosis, and cell cycle of MM SP cells. Additionally, knockdown of miR-127-3p activated the PI3K/AKT/mTOR signaling pathway in MM SP cells cotreated with DATS+Dex. CONCLUSION: We demonstrated that cotreatment with DATS+Dex reduced cell proliferation, promoted apoptosis, and caused cell cycle arrest of MM SP cells by promoting miR-127-3p expression and deactivating the PI3K/AKT/mTOR signaling pathway.


Subject(s)
Antineoplastic Agents/pharmacology , Dexamethasone/pharmacology , Disulfides/pharmacology , MicroRNAs/drug effects , Multiple Myeloma/drug therapy , Phosphatidylinositol 3-Kinase/drug effects , Proto-Oncogene Proteins c-akt/drug effects , Side-Population Cells/drug effects , Sulfinic Acids/pharmacology , Aldehyde Dehydrogenase 1 Family/metabolism , Apoptosis/drug effects , Apoptosis/physiology , Cell Line, Tumor , Cell Proliferation/drug effects , Databases, Genetic , Drug Resistance, Neoplasm , Drug Synergism , G1 Phase Cell Cycle Checkpoints , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Neoplastic Stem Cells/drug effects , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/pharmacology , S Phase Cell Cycle Checkpoints , Sex-Determining Region Y Protein/metabolism , Side-Population Cells/metabolism , Side-Population Cells/pathology , Signal Transduction/drug effects , Spheroids, Cellular/pathology , TOR Serine-Threonine Kinases/drug effects , TOR Serine-Threonine Kinases/metabolism
12.
Cell Mol Neurobiol ; 41(7): 1441-1452, 2021 Oct.
Article in English | MEDLINE | ID: mdl-32725456

ABSTRACT

The lack of an effective pharmaceutical agent for spinal cord injury (SCI) is a current problematic situation for clinicians, as the rate of motor vehicle accidents among young adults is on the rise. SCI contributes to the high disability rate. Presently, evidences detailing the precise pathological mechanisms in SCI are limited, compounding to the unavailability of an effective treatment method. Surgery, though not a complete curative method, is useful in managing some of the associated symptoms of secondary SCI. Autophagy and inflammation are contributive factors to both exacerbation and improvement of SCI. The mammalian target of rapamycin (mTOR) signaling pathway is a key player in the regulation of inflammatory response and autophagy. Valproic acid (VPA), a clinically used antiepileptic drug, has been suggested to improve neurological conditions, including SCI. This report reviewed the correlation between mTOR and autophagy, as well as autophagy's role and the therapeutic effects of VPA in SCI. VPA regulates autophagy by potentially inhibiting mTORC1, a complex of mTOR, while also hindering inflammatory response. Conclusively, an effective treatment for SCI could lie in the timely regulation of mTOR signaling pathway, and VPA could be the potential drug that improves SCI owing to its propensity to regulate the mTOR signaling pathway.


Subject(s)
Recovery of Function/drug effects , Spinal Cord Injuries/drug therapy , TOR Serine-Threonine Kinases/drug effects , Valproic Acid/pharmacology , Animals , Autophagy/drug effects , Spinal Cord/drug effects , Spinal Cord/metabolism , Spinal Cord Injuries/pathology , TOR Serine-Threonine Kinases/metabolism , Valproic Acid/metabolism
13.
Anticancer Drugs ; 32(8): 793-801, 2021 09 01.
Article in English | MEDLINE | ID: mdl-33675613

ABSTRACT

Sertraline and fluoxetine are the two most commonly used selective serotonin reuptake inhibitors (SSRIs) in the treatment of depression. Accumulating evidence has revealed that SSRIs can reduce the risk of hepatocellular carcinoma (HCC), but their therapeutic effects in HCC have not yet been elucidated. Previous studies have reported that sertraline and fluoxetine can suppress the growth of gastric carcinoma, melanoma and nonsmall cell lung cancers by inhibiting the mammalian target rapamycin (mTOR) activity. In this study, we found that sertraline and fluoxetine blocked the protein kinase B (AKT)/mTOR pathway and suppressed the growth of HCC cells in vitro, in xenografts and in diethylnitrosamine/carbon tetrachloride (DEN/CCL4)-induced primary liver mouse model. Sertraline and fluoxetine can synergize with sorafenib, the first approved standard therapy for advanced HCC, to inhibit the viability of HCC cells in vitro and in vivo. In addition, the combination of sorafenib and SSRIs synergistically inhibited the effects of the AKT/mTOR pathway. These results reveal novel therapeutic effects of a combination of SSRIs and sorafenib in HCC.


Subject(s)
Carcinoma, Hepatocellular/pathology , Floxuridine/pharmacology , Liver Neoplasms/pathology , Selective Serotonin Reuptake Inhibitors/pharmacology , Sertraline/pharmacology , Sorafenib/pharmacology , Animals , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Combinations , Humans , Male , Mice , Mice, Inbred BALB C , Proto-Oncogene Proteins c-akt/drug effects , TOR Serine-Threonine Kinases/drug effects , Xenograft Model Antitumor Assays
14.
Anticancer Drugs ; 32(8): 822-828, 2021 09 01.
Article in English | MEDLINE | ID: mdl-33783376

ABSTRACT

The clinical efficacy of sorafenib in hepatocellular carcinoma (HCC) is disappointing due to its low response rate and high rates of adverse effects. The eukaryotic translation initiation factor 4F (eIF4F) complex, mainly consisting of eIF4E-eukaryotic translation initiation factor 4G (eIF4G) interaction, is involved in the induction of drug resistance. Herein, we aimed to demonstrate that eIF4E-eIF4G complex inhibition enhanced the effect of sorafenib. The antiproliferation effect of combined treatment was evaluated by MTT assay and colony formation assay. Flow cytometry was used to detect the early cell apoptosis and cell cycle. The specific mechanism was demonstrated using western blot and lentivirus transfection. The combination of sorafenib with eIF4E-eIF4G inhibitors 4E1RCat (structural) or 4EGI-1 (competitive) synergistically inhibited the cell viability and colony formation ability of HCC cells. Moreover, the combined treatment induced more early apoptosis than sorafenib alone through downregulating the Bcl-2 expression. Besides, the coadministration of sorafenib and 4E1RCat or 4EGI-1 synergistically inhibited the expressions of eIF4E, eIF4G and phospho-4E-BP1 in HCC cells while blocking the phosphorylation of 4E-BP1 with lentiviral transfection failed to increase the sensitivity of HCC cells to sorafenib treatment. PI3K-AKT-mTOR signaling was also inhibited by the combined treatment. In a word, eIF4E-eIF4G complex inhibition synergistically enhances the effect of sorafenib in HCC treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/pathology , Eukaryotic Initiation Factor-4F/antagonists & inhibitors , Liver Neoplasms/pathology , Sorafenib/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Down-Regulation , Drug Combinations , Eukaryotic Initiation Factor-4E/antagonists & inhibitors , Eukaryotic Initiation Factor-4G/antagonists & inhibitors , Humans , Phosphatidylinositol 3-Kinases/drug effects , Proto-Oncogene Proteins c-bcl-2/drug effects , TOR Serine-Threonine Kinases/drug effects
15.
Anticancer Drugs ; 32(8): 786-792, 2021 09 01.
Article in English | MEDLINE | ID: mdl-33675608

ABSTRACT

Stachydrine is a bioactive alkaloid that has been found to exert tumor-suppressive potential. However, the effect of stachydrine on hepatocellular carcinoma (HCC) has not been previously investigated. In the present study, we investigated the effect of transforming growth factor-ß1 (TGF-ß1)-induced epithelial-mesenchymal transition (EMT) in HepG2 cells. Our results showed that stachydrine significantly suppressed TGF-ß1-induced HepG2 cell migration and invasion in a dose-dependent manner. Stachydrine prevented TGF-ß1-induced EMT in HepG2 cells, as proved by the increased expression level of E-cadherin and decreased expression levels of N-cadherin and vimentin. In addition, stachydrine attenuated TGF-ß1-induced upregulation of TGF-ß receptor I (TßRI) in both protein and mRNA levels. Further mechanism investigations proved that stachydrine prevented TGF-ß1-induced activation of Smad2/3 and phosphoinositol-3-kinase (PI3K)/Akt/mTOR signaling pathways in HepG2 cells. In conclusion, these findings demonstrated that stachydrine prevented TGF-ß1-induced EMT in HCC cells through Smad2/3 and PI3K/Akt/mTOR signaling pathways. Thus, stachydrine might be a potential therapeutic agent for the treatment of HCC.


Subject(s)
Carcinoma, Hepatocellular/pathology , Epithelial-Mesenchymal Transition/drug effects , Liver Neoplasms/pathology , Proline/analogs & derivatives , Transforming Growth Factor beta1/drug effects , Cadherins/drug effects , Cell Movement/drug effects , Cell Survival , Dose-Response Relationship, Drug , Hep G2 Cells , Humans , Phosphatidylinositol 3-Kinases , Proline/pharmacology , Signal Transduction/drug effects , Smad2 Protein/drug effects , TOR Serine-Threonine Kinases/drug effects , Vimentin/drug effects
16.
Anticancer Drugs ; 32(8): 864-874, 2021 09 01.
Article in English | MEDLINE | ID: mdl-33929996

ABSTRACT

Actein, a triterpene glycoside, isolated from rhizomes of Cimicifuga foetida, was reported to exhibit anticancer effects in vitro and in vivo. However, the effects of actein on colorectal cancer (CRC) remains unclear. As one of the most popular cancers all over the world, CRC ranked third place in both men and women. Recently, we investigated the potential anti-CRC effects of actein and its mechanisms. The Cell counting kit-8 cell proliferation assays, cell cycle detection, apoptosis detection, reactive oxygen species and mitochondrial membrane potential evaluation, western blot, as well as SW480 xenograft mice model were conducted to illustrate the mechanisms of action on anti-CRC effects of actein. Actein could significantly inhibit the human CRC cell lines SW480 and HT-29 proliferation, whereas less antiproliferation effects were found in normal colorectal cell lines HCoEpiC and FHC. Administration of actein resulted in G1 phase cell cycle arrest in both SW480 and HT-29 cells. Moreover, mitochondria-mediated apoptosis was also observed after treatment with actein in SW480 and HT-29 cell lines. Further investigation of mechanisms of action on actein-mediated anti-CRC proliferation effects indicated that the phosphoinositide 3-kinases (PI3K)/Akt pathways were involved. Actein significantly downregulated the phosphorylation of key molecules in PI3K/Akt pathways, including mTOR, glycogen synthesis kinase 3ß (GSK-3ß), as well as FOXO1. In addition, inosine 5'-monophosphate dehydrogenase type II (IMPDH2) was also observed decreasing in both SW480 and HT-29 cell lines after actein treatment, suggesting that actein may inhibit the PI3K/Akt pathways by decreasing IMPDH2. Finally, our SW480 xenograft model verified the anti-CRC effects and the safety of actein in vivo. Our findings suggest actein is worthy of further investigation as a novel drug candidate for the treatment of CRC.


Subject(s)
Colorectal Neoplasms/pathology , Phosphatidylinositol 3-Kinases/drug effects , Proto-Oncogene Proteins c-akt/drug effects , Saponins/pharmacology , Triterpenes/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Down-Regulation/drug effects , Humans , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/drug effects
17.
Clin Exp Dermatol ; 46(8): 1551-1554, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34115902

ABSTRACT

Merkel cell carcinoma (MCC) is a highly aggressive neuroendocrine neoplasm of the skin, which has an exceedingly poor prognosis. The AKT/mammalian target of rapamycin (mTOR) signalling pathway, which plays a pivotal role in the modulation of protein synthesis and cell survival, has been shown to be extremely important for Merkel cell carcinogenesis. In the current study, we found that AKT has important regulatory functions in MCC cells and that inhibition of AKT with the novel ATP-competitive AKT inhibitor, afuresertib, has widespread effects on proliferative pathways. In particular, we found that treatment of MCC cells with afuresertib led to deactivation of mTOR and glycogen synthase kinase 3 pathway proteins while increasing activation of proapoptotic pathways through the upregulation of p16 expression and phosphomodulation of the B-cell lymphoma-2-associated death promoter. Overall, afuresertib treatment led to significant and robust inhibition of MCC cell proliferation, thus raising intriguing questions regarding the potential efficacy of AKT inhibition for the future clinical management of MCC.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Merkel Cell/drug therapy , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Pyrazoles/therapeutic use , Skin Neoplasms/drug therapy , Thiophenes/therapeutic use , Trans-Activators/antagonists & inhibitors , Carcinoma, Merkel Cell/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Genes, p16/drug effects , Glycogen Synthase Kinase 3/drug effects , Glycogen Synthase Kinase 3/metabolism , Humans , Phosphorylation , Proto-Oncogene Proteins c-bcl-2/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction , Skin Neoplasms/pathology , TOR Serine-Threonine Kinases/drug effects , TOR Serine-Threonine Kinases/metabolism , Up-Regulation
18.
J Integr Neurosci ; 20(1): 67-75, 2021 Mar 30.
Article in English | MEDLINE | ID: mdl-33834692

ABSTRACT

Overactivation of the PI3-K/Akt/mTOR signaling pathway and inhibition of autophagy in the brain are involved in Alzheimer's disease. The present paper's goal was to explore the potential mechanisms of geniposide to protect against Alzheimer's disease. We treated the human neuroblastoma SH-SY5Y cell line with Aß1-42 as an Alzheimer's disease in vitro model to explore the potential mechanisms of geniposide to protect against Alzheimer's disease. Further, SH-SY5Y cells damaged by Aß1-42 were treated with geniposide. Akt/mTOR-related proteins and autophagy-associated proteins were measured to reveal the molecular mechanisms by which geniposide protects against Aß1-42-induced toxicity. Results showed that Akt and mTOR's geniposide inhibited phosphorylation induced by Aß1-42, enhanced expression of the LC3II/LC3I ratio, and Atg7 and Beclin1 expression and inhibited expression of p62 induced by Aß1-42. Our results lead us to hypothesize that inhibition of the Akt/mTOR signaling pathway and autophagy enhancement are fundamental molecular mechanisms for geniposide to protect against Aß toxicity.


Subject(s)
Alzheimer Disease/prevention & control , Amyloid beta-Peptides/toxicity , Autophagy/drug effects , Iridoids/pharmacology , Peptide Fragments/toxicity , Proto-Oncogene Proteins c-akt/drug effects , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/drug effects , Alzheimer Disease/chemically induced , Cell Line, Tumor , Humans
19.
Pak J Pharm Sci ; 34(3): 891-898, 2021 May.
Article in English | MEDLINE | ID: mdl-34602411

ABSTRACT

This study investigates the effects of ß-carboline alkaloids from Peganum harmala on FAK/PI3K/AKT/mTOR pathway in gastric cancer cell line SGC-7901 and tumor-bearing mice. Western blot, immunohistochemistry and RT-PCR were performed to detect protein and mRNA expressions of BCL-2, Bax, FAK, PI3K, AKT and mTOR. Mice model of gastric tumor was established with SGC-7901 cells. TUNEL assay was used to detect apoptosis. HE staining was used to observe morphological changes. In vitro, the protein and mRNA expressions of FAK, PI3K, AKT and mTOR in ß-carboline alkaloids groups were significantly lower than those in control and fluorouracil groups (P<0.05). BCL-2 decreased while Bax increased. In vivo, the tumor weights of ß-carboline alkaloids and fluorouracil groups were significantly lower than those of control group (P<0.05). FAK, PI3K, AKT and mTOR proteins in tumor tissues of ß-carboline alkaloids and fluorouracil groups were significantly lower than control group (P<0.05). Additionally, ß-carboline alkaloids treatment in vivo caused obvious cell necrosis and apoptosis. Conclusively, ß-carboline alkaloids can reduce FAK, PI3K, AKT and mTOR expressions at both protein and mRNA levels in SGC-7901 cells and tumor tissues formed by SGC-7901 cells. They may be targets of ß-carboline in FAK/PI3K/AKT/mTOR pathway.


Subject(s)
Alkaloids/pharmacology , Carbolines/pharmacology , Carcinoma/metabolism , Focal Adhesion Kinase 1/drug effects , Peganum , Phosphatidylinositol 3-Kinases/drug effects , Proto-Oncogene Proteins c-akt/drug effects , Signal Transduction/drug effects , Stomach Neoplasms/metabolism , TOR Serine-Threonine Kinases/drug effects , Animals , Carcinoma/genetics , Cell Line, Tumor , Focal Adhesion Kinase 1/genetics , Focal Adhesion Kinase 1/metabolism , Humans , Mice , Neoplasm Transplantation , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Stomach Neoplasms/genetics , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
20.
Am J Physiol Endocrinol Metab ; 319(1): E110-E116, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32421368

ABSTRACT

Statins lower cholesterol and risk of cardiovascular disease. Statins can increase blood glucose and risk of new-onset diabetes. It is unclear why statins can have opposing effects on lipids versus glucose. Statins have cholesterol-independent pleiotropic effects that influence both insulin and glucose control. Statin lowering of isoprenoids required for protein prenylation promotes pancreatic ß-cell dysfunction and adipose tissue insulin resistance. Protein prenylation influences immune function and statin-mediated adipose tissue insulin resistance involves the NLR family pyrin domain-containing 3 (NLRP3) inflammasome and IL-1ß. However, the intracellular cues that statins engage to activate the NLRP3 inflammasome and those responsible for IL-1ß-mediated insulin resistance in adipose tissue have not been identified. We hypothesized that stress kinases or components of the insulin signaling pathway mediated statin-induced insulin resistance. We tested the associations of p38, ERK, JNK, phosphatase, and tensin homolog (PTEN), and mTOR in statin-exposed adipose tissue from WT and IL-1ß-/- mice. We found that statins increased phosphorylation of p38 in WT and IL-1ß-/- mice. Statin activation of p38 upstream of IL-1ß led to priming of this NLRP3 inflammasome effector in macrophages. We found that mTORC1 inhibition with low doses of rapamycin (2 or 20 nM) lowered macrophage priming of IL-1ß mRNA and secretion of IL-1ß caused by multiple statins. Rapamycin (20 nM) or the rapalog everolimus (20 nM) prevented atorvastatin-induced lowering of insulin-mediated phosphorylation of Akt in mouse adipose tissue. These results position p38 and mTOR as mediators of statin-induced insulin resistance in adipose tissue and highlight rapalogs as candidates to mitigate the insulin resistance and glycemic side effects of statins.


Subject(s)
Atorvastatin/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Inflammasomes/drug effects , Insulin Resistance , Insulin/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/drug effects , TOR Serine-Threonine Kinases/drug effects , p38 Mitogen-Activated Protein Kinases/drug effects , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Inflammasomes/metabolism , Interleukin-1beta/genetics , MAP Kinase Kinase 4/drug effects , MAP Kinase Kinase 4/metabolism , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 1/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/drug effects , Mitogen-Activated Protein Kinase 3/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , PTEN Phosphohydrolase/drug effects , PTEN Phosphohydrolase/metabolism , TOR Serine-Threonine Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL