Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 780
1.
J Cancer Res Ther ; 19(6): 1560-1567, 2023 Dec 01.
Article En | MEDLINE | ID: mdl-38156922

OBJECTIVE: This study aimed to evaluate the impact of an adenosine monophosphate-activated protein kinase (AMPK) agonist, metformin (MET), on the antitumor effects of macrophages and to determine the underlying mechanism involved in the process. MATERIALS AND METHODS: M0 macrophages were derived from phorbol-12-myristate-13-acetate-stimulated THP-1 cells. RESULTS: The levels of tumor necrosis factor-alpha (TNF-α) and human leukocyte antigen-DR (HLA-DR) were decreased in macrophages incubated with HCT116 cells, whereas those of arginase-1 (Arg-1), CD163, and CD206 were elevated; these effects were reversed by MET. The transfection of small interfering (si) RNA abrogated the influence of MET on the expression of the M1/M2 macrophage biomarkers. MET significantly suppressed the proliferation and migration abilities of HCT116 cells incubated with M0 macrophages; these actions were reversed by siRNA transfection against AMPK. The hypoxia-inducible factor 1-alpha (HIF-1α), phosphorylated protein kinase B (p-AKT), and phosphorylated mammalian target of rapamycin (p-mTOR) levels were reduced by the introduction of MET and promoted by siRNA transfection against AMPK. In addition, the levels of HIF-1α, p-AKT, and p-mTOR suppressed by MET were markedly increased following the transfection of siRNA against AMPK. CONCLUSION: These findings indicate that MET can repress the progression of colorectal cancer by transforming tumor-associated macrophages to the M1phenotype via inhibition of the HIF-1α and mTOR signaling pathways.


Colorectal Neoplasms , Metformin , Signal Transduction , TOR Serine-Threonine Kinases , Tumor-Associated Macrophages , Metformin/pharmacology , Metformin/therapeutic use , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Colorectal Neoplasms/drug therapy , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Tumor-Associated Macrophages/drug effects , HCT116 Cells , Cell Polarity/drug effects , THP-1 Cells , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Gene Knockdown Techniques
2.
PLoS One ; 18(10): e0292309, 2023.
Article En | MEDLINE | ID: mdl-37788269

The coronavirus disease (COVID-19) pandemic has resulted in more than six million deaths by October 2022. Vaccines and antivirals for severe acute respiratory syndrome coronavirus 2 are now available; however, more effective antiviral drugs are required for effective treatment. Here, we report that a potent AMP-activated protein kinase (AMPK) inhibitor, compound C/dorsomorphin, inhibits the replication of the human coronavirus OC43 strain (HCoV-OC43). We examined HCoV-OC43 replication in control and AMPK-knockout (KO) cells and found that the virus replication decreased in AMPK-KO cells. Next, we examined the effect of the AMPK inhibitor, compound C on coronavirus replication. Compound C treatment efficiently inhibited the replication and decreased the coronavirus-induced cytotoxicity, further inhibiting autophagy. In addition, treatment with compound C in combination with chloroquine synergistically inhibited coronavirus replication. These results suggest that compound C can be considered as a potential drug candidate for COVID-19.


Antiviral Agents , Coronavirus OC43, Human , Humans , AMP-Activated Protein Kinases/antagonists & inhibitors , Antiviral Agents/pharmacology , Coronavirus OC43, Human/drug effects , Pyrazoles/pharmacology , Virus Replication/drug effects
3.
Bioorg Med Chem Lett ; 79: 129059, 2023 01 01.
Article En | MEDLINE | ID: mdl-36402454

Diabetes is a global healthcare problem that affects more than 400 million people worldwide. Treatment for type 1 and 2 diabetes is expected by targeting adenosine monophosphate activated protein kinase, AMPK, a well-known master regulator of glucose. Many pharmaceutical companies have tried to identify AMPK activators but few direct AMPK activators with high potency for the ß2-AMPK isoform, which is important for glucose homeostasis, have been found. In addition, their chemical structure is limited to benzimidazole or indole derivatives bearing an aromatic substituent at the C5 position of the core structure. We describe herein our efforts to identify novel benzimidazole derivatives that directly activate the ß2-AMPK isoform. Our newly designed activator 14d bearing a 1-amino indanyl moiety at the C5 position of the core exhibited high in vitro potency and good pharmacokinetic profiles. A single oral dosing of 14d showed dose-dependent activation of AMPK and blood-glucose-lowering effects was observed in a diabetic animal model. In addition, chronic AMPK activation with 14d led to dose-dependent reduction in HbA1c of the animal model.


AMP-Activated Protein Kinases , Benzimidazoles , Animals , AMP-Activated Protein Kinases/antagonists & inhibitors , Antinematodal Agents , Benzimidazoles/pharmacology , Benzimidazoles/therapeutic use , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Disease Models, Animal , Glucose , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology
4.
mBio ; 13(4): e0107622, 2022 08 30.
Article En | MEDLINE | ID: mdl-35703434

Chlamydia are obligate intracellular bacteria that reside within a membrane-bound compartment called the chlamydial inclusion inside a eukaryotic host cell. These pathogens have a complex biphasic developmental cycle, which involves conversion between a replicating, but noninfectious, reticulate body (RB) and an infectious elementary body (EB). Small molecule inhibitors have been reported to have deleterious effects on the intracellular Chlamydia infection, but these studies have typically been limited in terms of assays and time points of analysis. We compared published and novel inhibitors and showed that they can differentially alter inclusion size, chlamydial number and infectious EB production, and that these effects can vary over the course of the intracellular infection. Our results provide the justification for analysis with multiple assays performed either at the end of the infection or over a time course. We also show that this approach has the potential to identify the particular step in the developmental cycle that is impacted by the inhibitor. We furthermore propose that the magnitude of inhibitor-induced progeny defects are best quantified and compared by using a new value called maximal progeny production (Progenymax). As a demonstration of the validity of this systematic approach, we applied it to inhibitors of Akt and AMPK, which are host kinases involved in lipid synthesis and cholesterol trafficking pathways. Both inhibitors reduced EB production, but Akt disruption primarily decreased RB-to-EB conversion while AMPK inhibition paradoxically enhanced RB replication. IMPORTANCE Chlamydia is the most reported cause of bacterial, sexually transmitted infection in the United States. This bacterium infects human cells and reproduces within a cytoplasmic inclusion via an unusual developmental cycle involving two specialized chlamydial forms. Small molecule compounds have been reported to negatively affect the inclusion as well as chlamydial replication and infectious progeny production, but we showed that these effects can be discordant and vary over the course of the 48- to 72-hour long intracellular infection. We propose approaches to analyze these nonuniform effects, including measurements at the end of the intracellular infection, and more detailed analysis with multiple assays performed over the course of the developmental cycle. We then applied this approach to investigate and compare the anti-chlamydial effects of two inhibitors that alter host lipid synthesis and cholesterol trafficking.


Chlamydia Infections , Chlamydia , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/metabolism , AMP-Activated Protein Kinases/pharmacology , Bacterial Proteins/metabolism , Chlamydia/metabolism , Chlamydia Infections/drug therapy , Chlamydia trachomatis/metabolism , Humans , Lipids , Proto-Oncogene Proteins c-akt/metabolism
5.
Food Funct ; 13(6): 3234-3246, 2022 Mar 21.
Article En | MEDLINE | ID: mdl-35213678

Background: Abnormal proliferation of vascular smooth muscle cells (VSMCs) in the intimal region is a key event in the development of neointimal hyperplasia. 10-G, a bioactive compound found in ginger, exerted inhibitory effects on the proliferation of several cancer cells. However, the effect and mechanism of 10-G on neointimal hyperplasia are not clear. Purpose: To explore the suppressive effects of 10-G on the proliferation and migration of VSMCs, and investigate the underlying mechanisms. Methods: In vivo, a left common carotid artery ligation mouse model was used to observe the effects of neointimal formation through immunohistochemistry and hematoxylin-eosin staining. In vitro, the cell proliferation and migration of HASMCs and A7r5 cells were detected by MTS assay, EdU staining, wound healing assay, Transwell assay, and western blotting as well. Molecular docking, molecular dynamics simulations and surface plasmon resonance imaging were collectively used to evaluate the interaction of 10-G with AMP-activated protein kinase (AMPK). Compound C and si-AMPK were used to inhibit the expression of AMPK. Results: Treatment with 10-G significantly reduced neointimal hyperplasia in the left common carotid artery ligation mouse model. MST and EdU staining showed that 10-G inhibited the proliferation of VSMC cells A7r5 and HASMC. We also found that 10-G altered the expression of proliferation-related proteins, including CyclinD1, CyclinD2, CyclinD3, and CDK4. Molecular docking revealed that the binding energy between AMPK and 10-G is -7.4 kcal mol-1. Molecular simulations suggested that the binding between 10-G and AMPK is stable. Surface plasmon resonance imaging analysis also showed that 10-G has a strong binding affinity to AMPK (KD = 6.81 × 10-8 M). 10-G promoted AMPKα phosphorylation both in vivo and in vitro. Blocking AMPK by an siRNA or AMPK inhibitor pathway partly abolished the anti-proliferation effects of 10-G on VSMCs. Conclusion: These data showed that 10-G might inhibit neointimal hyperplasia and suppress VSMC proliferation by the activation of AMPK as a natural AMPK agonist.


AMP-Activated Protein Kinases/metabolism , Catechols/pharmacology , Fatty Alcohols/pharmacology , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/drug effects , Neointima/pathology , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/chemistry , Animals , Catechols/chemistry , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Enzyme Activation , Fatty Alcohols/chemistry , Humans , Hyperplasia , Male , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Docking Simulation , Muscle, Smooth, Vascular/drug effects , Phosphorylation , Protein Conformation , Rats , Signal Transduction , Surface Plasmon Resonance , TOR Serine-Threonine Kinases/metabolism
6.
Invest Ophthalmol Vis Sci ; 63(1): 4, 2022 01 03.
Article En | MEDLINE | ID: mdl-34982146

Purpose: Netarsudil, a Rho kinase inhibitor with norepinephrine transport inhibitory effect, lowers intraocular pressure, however, its effect on axon damage remains to be elucidated. The aim of the current study was to investigate the effect of netarsudil on TNF-induced axon loss and to examine whether it affects phosphorylated-AMP-activated kinase (p-AMPK) and autophagy in the optic nerve. Methods: Intravitreal administration of TNF or TNF with netarsudil was carried out on rats and quantification of axon number was determined. Electron microscopy determined autophagosome numbers. Localization of p-AMPK expression was examined by immunohistochemistry. The changes in p62, LC3-II, and p-AMPK levels were estimated in the optic nerve by immunoblot analysis. The effect of an AMPK activator A769662 or an AMPK inhibitor dorsomorphin on axon number was evaluated. Results: Morphometric analysis revealed apparent protection by netarsudil against TNF-induced axon degeneration. Netarsudil increased autophagosome numbers inside axons. Netarsudil treatment significantly upregulated optic nerve LC3-II levels in both the TNF-treated eyes and the control eyes. Increased p62 protein level induced by TNF was significantly ameliorated by netarsudil. The netarsudil administration alone lessened p62 levels. Netarsudil significantly upregulated the optic nerve p-AMPK levels. A769662 exhibited obvious axonal protection against TNF-induced damage. A769662 treatment upregulated LC3-II levels and the increment of p62 level induced by TNF was significantly ameliorated by A769662. Immunohistochemical analysis revealed that p-AMPK is present in axons. Netarsudil-mediated axonal protection was significantly suppressed by dorsomorphin administration. Conclusions: Netarsudil upregulated p-AMPK and autophagy. Netarsudil-mediated axonal protection may be associated with upregulated p-AMPK.


AMP-Activated Protein Kinases/metabolism , Autophagy/physiology , Axons/drug effects , Benzoates/pharmacology , Nerve Degeneration/prevention & control , Optic Nerve/drug effects , Tumor Necrosis Factor-alpha/toxicity , beta-Alanine/analogs & derivatives , rho-Associated Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/antagonists & inhibitors , Animals , Axons/enzymology , Axons/pathology , Biphenyl Compounds/pharmacology , Enzyme Inhibitors/pharmacology , Immunohistochemistry , Intravitreal Injections , Male , Microscopy, Electron , Microtubule-Associated Proteins/metabolism , Nerve Degeneration/enzymology , Optic Nerve/ultrastructure , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Pyrones/pharmacology , Rats , Rats, Wistar , Sequestosome-1 Protein/metabolism , Thiophenes/pharmacology , beta-Alanine/pharmacology
7.
Biochem Pharmacol ; 197: 114885, 2022 03.
Article En | MEDLINE | ID: mdl-34968488

As a highly prevalent neuropsychiatric disorder worldwide, the pathophysiology of depression is not yet fully understood and based on multiple factors among which chronic stress is critical. Numerous previous studies have shown the role of central mammalian target of rapamycin complex 1 (mTORC1) signaling in depression. However, so far it remains elusive by which way chronic stress down-regulates the activity of central mTORC1. Liver kinase b1 (LKB1) has been demonstrated to regulate the activity of the mTORC1 signaling cascade by phosphorylating AMP activated protein kinase (AMPK). Here, this study aimed to explore whether LKB1 participates in depression by regulating the downstream AMPK-mTORC1 signaling, and various methods including mouse models of depression, western blotting and immunofluorescence were used together. Our results showed that chronic stress significantly enhanced the expression of both phosphorylated LKB1 and total LKB1 in the medial prefrontal cortex (mPFC) but not the hippocampus. Furthermore, genetic knockdown of LKB1 in the mPFC fully reversed not only the depressive-like behaviors induced by chronic stress in mice but also the effects of chronic stress on the activity of AMPK and the mTORC1 system. Taken together, this study preliminarily suggests that LKB1 in the mPFC could be a feasible target for antidepressants. This study also provides support for the potential use of LKB1 inhibition strategies against the chronic stress-related neuropsychiatric disorders.


AMP-Activated Protein Kinases/metabolism , Dependovirus/metabolism , Depression/metabolism , Prefrontal Cortex/metabolism , Social Defeat , Stress, Psychological/metabolism , AMP-Activated Protein Kinase Kinases/metabolism , AMP-Activated Protein Kinases/antagonists & inhibitors , Animals , Depression/virology , Female , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Inbred C57BL , Prefrontal Cortex/virology , Stress, Psychological/virology
8.
Bioorg Med Chem ; 52: 116522, 2021 12 15.
Article En | MEDLINE | ID: mdl-34837819

Stroke is a major public health problem with an imperative need for a more effective and tolerated therapy. Neuroprotective therapy may be an effective therapeutic intervention for stroke. The morbidity and mortality of stroke-induced secondary brain injury is mainly caused by neuronal apoptosis, which can be executed in a caspase-dependent or apoptosis inducing factor (AIF)-dependent manner. As apoptosis is an energy-dependent process with a relative time delay, abnormal energy metabolism could be a significant and fundamental pathophysiological basis of stroke. To our knowledge, convincible evidences that AMPK inhibition exerts neuroprotection in cerebral ischemia injury via anti-apoptosis remain to be investigated. Accordingly, the aims of this study were to investigate the protective effects of AMPK inhibitor BML-275 on cerebral ischemic/reperfusion (I/R) injury and to elucidate the underlying mechanisms. Cerebral ischemia was induced by transient middle cerebral artery occlusion (tMCAO) in male C57BL/6 mice. The therapeutic effects of BML-275 were evaluated by infarct sizes, neurological scores and the proportion of apoptotic neurons after 24 h of reperfusion. The cell apoptosis markers cyt c and AIF were also evaluated. The results showed that intraperitoneally administration of BML-275 alleviate the cerebral infarction, neurological deficit and neuronal apoptosis induced by MCAO. BML-275 simultaneously induces anti-apoptosis and decreases the expression of cyt c and AIF. This study supports the hypothesis that anti-apoptosis is one of potential neuroprotective strategies for the treatment of stroke.


AMP-Activated Protein Kinases/antagonists & inhibitors , Apoptosis Inducing Factor/antagonists & inhibitors , Brain Ischemia/drug therapy , Cytochromes c/antagonists & inhibitors , Neuroprotective Agents/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , AMP-Activated Protein Kinases/metabolism , Animals , Apoptosis/drug effects , Apoptosis Inducing Factor/genetics , Apoptosis Inducing Factor/metabolism , Brain Ischemia/metabolism , Brain Ischemia/pathology , Cytochromes c/genetics , Cytochromes c/metabolism , Dose-Response Relationship, Drug , Male , Mice , Mice, Inbred C57BL , Molecular Structure , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/chemistry , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Structure-Activity Relationship
9.
J Immunol Res ; 2021: 4084566, 2021.
Article En | MEDLINE | ID: mdl-34734090

Nonalcoholic fatty liver disease (NAFLD) is closely related to glycolipid metabolism and liver inflammation. And there is no effective drug approved for its clinical therapy. In this study, we focused on mangiferin (Man) and explored its effects and mechanisms on NAFLD treatment based on the regulation of glycolipid metabolism and anti-inflammatory in vivo and in vitro. The results exhibited that Man can significantly attenuate liver injury, insulin resistance, and glucose tolerance in high-fat diet- (HFD-) induced NAFLD mice and significantly reduce fat accumulation and inflammation in hepatic tissue of NAFLD mice. The transcriptome level RNA-seq analysis showed that the significantly different expression genes between the Man treatment group and the HFD-induced NAFLD model group were mainly related to regulation of energy, metabolism, and inflammation in liver tissue. Furthermore, western blots, real-time PCR, and immunohistochemistry experiments confirmed that Man significantly activated the AMPK signal pathway and inhibited NLRP3 inflammasome activation and pyroptosis in NAFLD mice. In in vitro cell experiments, we further confirmed that Man can promote glucose consumption and reduce intracellular triglyceride (TG) accumulation induced by free fatty acids in HepG2 cells and further that it can be blocked by AMPK-specific inhibitors. Western blot results showed that Man upregulated p-AMPKα levels and exhibited a significant AMPK activation effect, which was blocked by compound C. At the same time, Man downregulated the expression of NLRP3 inflammasome-related proteins and inhibited the activation of NLRP3 inflammasome, alleviating cell pyroptosis and inflammation effects. These results indicate that Man anti-NAFLD activity is mediated through its regulation of glucolipid metabolism by AMPK activation and its anti-inflammatory effects by NLRP3 inflammasome inhibition. Our study indicates that Man is a promising prodrug for the therapy of NAFLD patients.


AMP-Activated Protein Kinases/antagonists & inhibitors , Non-alcoholic Fatty Liver Disease/drug therapy , Xanthones/pharmacology , AMP-Activated Protein Kinases/metabolism , Animals , Diet, High-Fat , Disease Models, Animal , Hep G2 Cells , Humans , Inflammasomes/antagonists & inhibitors , Inflammasomes/metabolism , Lipid Metabolism/drug effects , Lipid Metabolism/immunology , Liver/drug effects , Liver/immunology , Liver/pathology , Male , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Non-alcoholic Fatty Liver Disease/immunology , Non-alcoholic Fatty Liver Disease/pathology , Pyroptosis/drug effects , Pyroptosis/immunology , Signal Transduction/drug effects , Signal Transduction/immunology , Xanthones/therapeutic use
10.
Cell Rep ; 37(1): 109785, 2021 10 05.
Article En | MEDLINE | ID: mdl-34610303

A hallmark of type 2 diabetes (T2D) is hepatic resistance to insulin's glucose-lowering effects. The serum- and glucocorticoid-regulated family of protein kinases (SGK) is activated downstream of mechanistic target of rapamycin complex 2 (mTORC2) in response to insulin in parallel to AKT. Surprisingly, despite an identical substrate recognition motif to AKT, which drives insulin sensitivity, pathological accumulation of SGK1 drives insulin resistance. Liver-specific Sgk1-knockout (Sgk1Lko) mice display improved glucose tolerance and insulin sensitivity and are protected from hepatic steatosis when fed a high-fat diet. Sgk1 promotes insulin resistance by inactivating AMP-activated protein kinase (AMPK) via phosphorylation on inhibitory site AMPKαSer485/491. We demonstrate that SGK1 is dominant among SGK family kinases in regulation of insulin sensitivity, as Sgk1, Sgk2, and Sgk3 triple-knockout mice have similar increases in hepatic insulin sensitivity. In aggregate, these data suggest that targeting hepatic SGK1 may have therapeutic potential in T2D.


AMP-Activated Protein Kinases/metabolism , Immediate-Early Proteins/metabolism , Liver/metabolism , Protein Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinases/antagonists & inhibitors , Animals , Diabetes Mellitus, Type 2/pathology , Diet, High-Fat , Forkhead Box Protein O1/metabolism , Glucose/metabolism , Immediate-Early Proteins/deficiency , Immediate-Early Proteins/genetics , Insulin/metabolism , Insulin Resistance , Mechanistic Target of Rapamycin Complex 2/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Guide, Kinetoplastida/metabolism , Signal Transduction
11.
FASEB J ; 35(10): e21932, 2021 10.
Article En | MEDLINE | ID: mdl-34549830

Myocardial fibrosis is a characteristic of various cardiomyopathies, and myocardial fibroblasts play a central role in this process. Gelsolin (GSN) is an actin severing and capping protein that regulates actin assembly and may be involved in fibroblast activation. While the role of GSN in mechanical stress-mediated cardiac fibrosis has been explored, its role in myocardial fibrosis in the absence of mechanical stress is not defined. In this study, we investigated the role of GSN in myocardial fibrosis induced by Angiotensin II (Ang II), a profibrotic hormone that is elevated in cardiovascular disease. We utilized mice lacking GSN (Gsn-/- ) and cultured primary adult cardiac fibroblasts (cFB). In vivo, Ang II infusion in mice resulted in significantly less severe myocardial fibrosis in Gsn-/- compared with Gsn+/+ mice, along with diminished activation of the TGFß1-Smad2/3 pathway, and reduced expression of cardiac extracellular matrix proteins (collagen, fibronectin, periostin). Moreover, Gsn-deficient hearts exhibited suppressed activity of the AMPK pathway and its downstream effectors, mTOR and P70S6Kinase, which could contribute to the suppressed TGFß1 activity. In vitro, the Ang II-induced activation of cFBs was reduced in Gsn-deficient fibroblasts evident from decreased expression of αSMA and periostin, diminished actin filament turnover; which also exhibited reduced activity of the AMPK-mTOR pathway, and P70S6K phosphorylation. AMPK inhibition compensated for the loss of GSN, restored the levels of G-actin in Gsn-/- cFBs and promoted activation to myofibroblasts by increasing αSMA and periostin levels. This study reveals a novel role for GSN in mediating myocardial fibrosis by regulating the AMPK-mTOR-P70S6K pathway in cFB activation independent from mechanical stress-induced factors.


Angiotensin II/pharmacology , Fibroblasts/drug effects , Fibrosis/pathology , Gelsolin/metabolism , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/chemistry , AMP-Activated Protein Kinases/metabolism , Actins/metabolism , Animals , Fibroblasts/metabolism , Fibroblasts/pathology , Fibrosis/metabolism , Gelsolin/deficiency , Gelsolin/genetics , Homeostasis , Male , Mice , Myocardium/metabolism , Myocardium/pathology , Myofibroblasts/drug effects , Myofibroblasts/pathology , Phosphorylation , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Transforming Growth Factor beta1/metabolism
12.
EMBO J ; 40(21): e108028, 2021 11 02.
Article En | MEDLINE | ID: mdl-34472622

Adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) is an important cellular metabolite-sensing enzyme that can directly sense changes not only in ATP but also in metabolites associated with carbohydrates and fatty acids. However, less is known about whether and how AMPK senses variations in cellular amino acids. Here, we show that cysteine deficiency significantly triggers calcium/calmodulin-dependent protein kinase kinase 2 (CaMKK2)-mediated activation of AMPK. In addition, we found that CaMKK2 directly associates with cysteinyl-tRNA synthetase (CARS), which then binds to AMPKγ2 under cysteine deficiency to activate AMPK. Interestingly, we discovered that cysteine inhibits the binding of CARS to AMPKγ2, and thus, under cysteine deficiency conditions wherein the inhibitory effect of cysteine is abrogated, CARS mediates the binding of AMPK to CaMKK2, resulting in the phosphorylation and activation of AMPK by CaMKK2. Importantly, we demonstrate that blocking AMPK activation leads to cell death under cysteine-deficient conditions. In summary, our study is the first to show that CARS senses the absence of cysteine and activates AMPK through the cysteine-CARS-CaMKK2-AMPKγ2 axis, a novel adaptation strategy for cell survival under nutrient deprivation conditions.


AMP-Activated Protein Kinases/genetics , Adaptation, Physiological/genetics , Amino Acyl-tRNA Synthetases/genetics , Calcium-Calmodulin-Dependent Protein Kinase Kinase/genetics , Cysteine/deficiency , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/metabolism , Acetyl-CoA Carboxylase/genetics , Acetyl-CoA Carboxylase/metabolism , Adenosine Triphosphate/metabolism , Amino Acyl-tRNA Synthetases/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Kinase/metabolism , Cell Line, Tumor , Cell Survival/genetics , Epithelial Cells/cytology , Epithelial Cells/metabolism , Fatty Acids/metabolism , Gene Expression Regulation , HEK293 Cells , Humans , Protein Binding , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Regulatory-Associated Protein of mTOR/genetics , Regulatory-Associated Protein of mTOR/metabolism , Signal Transduction
13.
Cell Biochem Funct ; 39(8): 991-997, 2021 Dec.
Article En | MEDLINE | ID: mdl-34472641

Breast cancer is associated with a high rate of recurrence, resistance therapy and mortality worldwide. We aimed at investigating the inhibitory effects of Sulindac and vitamin D3 (VD) on MCF-7 human breast cancer cells. MCF-7 cells were cultured with different concentrations of Sulindac and VD over a period of 24, 48 and 72 hours for cell viability and IC50 experiments. Hochst staining was used to evaluate apoptosis, whereas quantitative PCR (qPCR) was performed to measure mRNA levels of BCL-2 and BAX genes. Immunofluorescence staining was used to monitor intracellular ß-catenin expression. The protein levels of AKT, AMPK and P65 were measured by western blotting. The result showed that cell viability decreased in treated cells dose/time dependently (P < .05). Hochst staining showed an increase in fragmented nuclei in treated cells. The expression of BCL-2 and BAX genes decreased and increased in treated cells, respectively (P < .05). Immunofluorescence staining indicated that the expression of ß-catenin significantly reduced in treated cells. The AKT-1/p-Akt-1 and AMPK/p-AMPK ratio increased in treated cells (P < .05), but the P65/p-P65 ratio did not change significantly (P > .05). Our results indicated that the combination of Sulindac and VD has a growth-inhibiting effect on MCF-7 cells through AMPK/Akt/ß-catenin axis.


AMP-Activated Protein Kinases/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Cholecalciferol/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Sulindac/pharmacology , beta Catenin/antagonists & inhibitors , AMP-Activated Protein Kinases/metabolism , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Cholecalciferol/chemistry , Drug Screening Assays, Antitumor , Female , Humans , MCF-7 Cells , Proto-Oncogene Proteins c-akt/metabolism , Sulindac/chemistry , Tumor Cells, Cultured , beta Catenin/metabolism
14.
Biochem Pharmacol ; 192: 114730, 2021 10.
Article En | MEDLINE | ID: mdl-34400125

Activation of hepatic stellate cells (HSCs) is a central event in the pathogenesis of liver fibrosis and is often accompanied by the disappearance of lipid droplets (LDs). Although interference with LD metabolism can effectively reverse the activation of HSCs, there is currently no effective therapy for liver fibrosis. Our previous evidence indicates that long non-coding RNA (lncRNA)-H19 plays an essential role in LD metabolism of HSC. In this study, we investigated the potential molecular mechanism of dihydroartemisinin (DHA) inhibits LD metabolism and liver fibrosis by regulating H19-AMPK pathway. We found that DHA restores LDs content in activated HSCs via reducing the transcription of H19 driven by hypoxia inducible factor 1 subunit alpha (HIF1α) and inhibiting the lipid oxidation signal mediated by AMP-activated protein kinase (AMPK) phosphorylation. In vivo experiments, we have proved that DHA reduced the deposition of extracellular matrix (ECM) and reduce the level of liver fibrosis in CCl4-induced liver fibrosis of mice. In summary, our results emphasize the importance of H19 in liver fibrosis and the potential of DHA to regulate H19 to treat liver fibrosis, providing a new direction for the prevention and treatment of liver fibrosis.


AMP-Activated Protein Kinases/antagonists & inhibitors , Artemisinins/therapeutic use , Hepatic Stellate Cells/drug effects , Lipid Droplets/drug effects , Lipid Metabolism/drug effects , RNA, Long Noncoding/antagonists & inhibitors , AMP-Activated Protein Kinases/metabolism , Animals , Artemisinins/pharmacology , Cell Line , Hepatic Stellate Cells/metabolism , Humans , Lipid Droplets/metabolism , Lipid Metabolism/physiology , Liver Cirrhosis/drug therapy , Liver Cirrhosis/metabolism , Male , Mice , Mice, Inbred ICR , RNA, Long Noncoding/metabolism
15.
Biochem J ; 478(15): 2977-2997, 2021 08 13.
Article En | MEDLINE | ID: mdl-34259310

SBI-0206965, originally identified as an inhibitor of the autophagy initiator kinase ULK1, has recently been reported as a more potent and selective AMP-activated protein kinase (AMPK) inhibitor relative to the widely used, but promiscuous inhibitor Compound C/Dorsomorphin. Here, we studied the effects of SBI-0206965 on AMPK signalling and metabolic readouts in multiple cell types, including hepatocytes, skeletal muscle cells and adipocytes. We observed SBI-0206965 dose dependently attenuated AMPK activator (991)-stimulated ACC phosphorylation and inhibition of lipogenesis in hepatocytes. SBI-0206965 (≥25 µM) modestly inhibited AMPK signalling in C2C12 myotubes, but also inhibited insulin signalling, insulin-mediated/AMPK-independent glucose uptake, and AICA-riboside uptake. We performed an extended screen of SBI-0206965 against a panel of 140 human protein kinases in vitro, which showed SBI-0206965 inhibits several kinases, including members of AMPK-related kinases (NUAK1, MARK3/4), equally or more potently than AMPK or ULK1. This screen, together with molecular modelling, revealed that most SBI-0206965-sensitive kinases contain a large gatekeeper residue with a preference for methionine at this position. We observed that mutation of the gatekeeper methionine to a smaller side chain amino acid (threonine) rendered AMPK and ULK1 resistant to SBI-0206965 inhibition. These results demonstrate that although SBI-0206965 has utility for delineating AMPK or ULK1 signalling and cellular functions, the compound potently inhibits several other kinases and critical cellular functions such as glucose and nucleoside uptake. Our study demonstrates a role for the gatekeeper residue as a determinant of the inhibitor sensitivity and inhibitor-resistant mutant forms could be exploited as potential controls to probe specific cellular effects of SBI-0206965.


AMP-Activated Protein Kinases/antagonists & inhibitors , Autophagy-Related Protein-1 Homolog/antagonists & inhibitors , Benzamides/pharmacology , Pyrimidines/pharmacology , Recombinant Proteins/metabolism , 3T3-L1 Cells , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Autophagy-Related Protein-1 Homolog/genetics , Autophagy-Related Protein-1 Homolog/metabolism , Benzamides/metabolism , Cell Line , Cell Line, Tumor , Cells, Cultured , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Molecular Docking Simulation , Mutation, Missense , Protein Binding/drug effects , Protein Multimerization , Pyrimidines/metabolism , Rats, Sprague-Dawley , Recombinant Proteins/chemistry , Recombinant Proteins/genetics
16.
Cell Commun Signal ; 19(1): 68, 2021 06 30.
Article En | MEDLINE | ID: mdl-34193159

BACKGROUND: The oncogenic PIM kinases and the tumor-suppressive LKB1 kinase have both been implicated in the regulation of cell growth and metabolism, albeit in opposite directions. Here we investigated whether these kinases interact with each other to influence AMPK activation and tumorigenic growth of prostate and breast cancer cells. METHODS: We first determined how PIM and LKB1 kinases affect AMPK phosphorylation levels. We then used in vitro kinase assays to demonstrate that LKB1 is phosphorylated by PIM kinases, and site-directed mutagenesis to identify the PIM target sites in LKB1. The cellular functions of PIM and LKB1 kinases were evaluated using either pan-PIM inhibitors or CRISPR/Cas9 genomic editing, with which all three PIM family members and/or LKB1 were knocked out from PC3 prostate and MCF7 breast cancer cell lines. In addition to cell proliferation assays, we examined the effects of PIM and/or LKB1 loss on tumor growth using the chick embryo chorioallantoic membrane (CAM) xenograft model. RESULTS: We provide both genetic and pharmacological evidence to demonstrate that inhibition of PIM expression or activity increases phosphorylation of AMPK at Thr172 in both PC3 and MCF7 cells, but not in their derivatives lacking LKB1. This is explained by our observation that all three PIM family kinases can phosphorylate LKB1 at Ser334. Wild-type LKB1, but not its phosphodeficient derivative, can restore PIM inhibitor-induced AMPK phosphorylation in LKB1 knock-out cells. In the CAM model, loss of LKB1 enhances tumorigenicity of PC3 xenografts, while cells lacking both LKB1 and PIMs exhibit slower proliferation rates and form smaller tumors. CONCLUSION: PIM kinases are novel negative regulators of LKB1 that affect AMPK activity in an LKB1-dependent fashion. The impairment of cell proliferation and tumor growth in cells lacking both LKB1 and PIMs indicates that these kinases possess a shared signaling role in the context of cancer. These data also suggest that PIM inhibitors may be a rational therapeutic option for LKB1-deficient tumors. Video Abstract.


AMP-Activated Protein Kinase Kinases/metabolism , AMP-Activated Protein Kinases/metabolism , Carcinogenesis/metabolism , Carcinogenesis/pathology , Proto-Oncogene Proteins c-pim-1/metabolism , AMP-Activated Protein Kinases/antagonists & inhibitors , Cell Line, Tumor , Cell Proliferation , Enzyme Activation , Humans , Phosphorylation , Protein Binding , Substrate Specificity
17.
Nat Commun ; 12(1): 4246, 2021 07 12.
Article En | MEDLINE | ID: mdl-34253728

Modeling human diseases as networks simplify complex multi-cellular processes, helps understand patterns in noisy data that humans cannot find, and thereby improves precision in prediction. Using Inflammatory Bowel Disease (IBD) as an example, here we outline an unbiased AI-assisted approach for target identification and validation. A network was built in which clusters of genes are connected by directed edges that highlight asymmetric Boolean relationships. Using machine-learning, a path of continuum states was pinpointed, which most effectively predicted disease outcome. This path was enriched in gene-clusters that maintain the integrity of the gut epithelial barrier. We exploit this insight to prioritize one target, choose appropriate pre-clinical murine models for target validation and design patient-derived organoid models. Potential for treatment efficacy is confirmed in patient-derived organoids using multivariate analyses. This AI-assisted approach identifies a first-in-class gut barrier-protective agent in IBD and predicted Phase-III success of candidate agents.


Artificial Intelligence , Inflammatory Bowel Diseases/therapy , Intestinal Mucosa/pathology , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/metabolism , Animals , Cohort Studies , Colitis/genetics , Dextran Sulfate , Disease Models, Animal , Gene Expression Regulation , Humans , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/pathology , Likelihood Functions , Machine Learning , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Targeted Therapy , Multigene Family , Organoids/pathology , Reproducibility of Results , Treatment Outcome
18.
Neurochem Res ; 46(11): 2948-2957, 2021 Nov.
Article En | MEDLINE | ID: mdl-34268656

Our previous studies revealed that the expression of stanniocalcin-1 (STC1) in astrocytes increased under hypoxic conditions. However, the role of STC1 in hypoxic astrocytes is not well understood. In this work, we first showed the increased expression of STC1 in astrocyte cell line and astrocytes in the brain tissues of mice after exposure to hypoxia. Then, we found that knockdown of STC1 inhibited cell viability and increased apoptosis. These effects were mediated by decreasing the levels of SIRT3, UCP2, and glycolytic genes and increasing the levels of ROS. Further studies suggested that STC1 silencing promoted oxidative stress and suppressed glycolysis by downregulating AMPKα1. Moreover, HIF-1α knockdown in hypoxic astrocytes led to decreased expression of STC1 and AMPKα1, indicating that the expression of STC1 was regulated by HIF-1α. In conclusion, our study showed that HIF-1α-induced STC1 could protect astrocytes from hypoxic damage by regulating glycolysis and redox homeostasis in an AMPKα1-dependent manner.


AMP-Activated Protein Kinases/metabolism , Astrocytes/metabolism , Cell Hypoxia/physiology , Cytoprotection/physiology , Glycoproteins/biosynthesis , Hypoxia/metabolism , AMP-Activated Protein Kinases/antagonists & inhibitors , Animals , Astrocytes/pathology , Cell Survival/physiology , Cells, Cultured , Gene Knockdown Techniques/methods , Humans , Hypoxia/prevention & control , Male , Mice , Mice, Inbred C57BL
19.
Immunopharmacol Immunotoxicol ; 43(5): 519-526, 2021 Oct.
Article En | MEDLINE | ID: mdl-34308732

BACKGROUND: Cytokines can induce a chronic inflammatory response in the periodontium, leading to periodontitis. Quercetin, a naturally occuring flavonoid, has been shown to inhibit periodontitis, but how it works is poorly understood. In this study, we assessed the impact of quercetin on lipopolysaccharide (LPS)-induced inflammatory damage in oral mucosal keratinocytes (hOMK107) and explored its underlying mechanism. METHODS: The viability and apoptosis of hOMK107 cells were measured after exposure to LPS, followed or not by quercetin. The production of IL-1ß, IL-6, IL-8, TNF-ɑ, iNOS, and COX-2 was quantified by enzyme-linked immunosorbent assay (ELISA), while levels of Akt, AMPK, and mTOR and their phosphorylation were detected semi-quantitatively by western blotting. RESULTS: Quercetin significantly improved cell viability and apoptosis by reversing LPS-induced upregulation of Bax and downregulation of Bcl-2 in hOMK107 cells. Quercetin decreased the production of IL-1ß, IL-6, IL-8, TNF-ɑ, iNOS, and COX-2, as well as signal transduction via the Akt/AMPK/mTOR pathway. Inhibitors of Akt, AMPK, and mTOR strengthened the anti-apoptotic effects of quercetin, while agonists of Akt, AMPK, or mTOR or Akt overexpression weakened the anti-apoptotic effects. CONCLUSION: These results indicate that quercetin may have a potential protective effect against the chronic inflammation-related periodontitis via suppressing Akt/AMPK/mTOR pathway.


AMP-Activated Protein Kinases/antagonists & inhibitors , Keratinocytes/drug effects , Mouth Mucosa/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Quercetin/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/metabolism , Antioxidants/pharmacology , Cell Line , Cell Survival/drug effects , Cell Survival/physiology , Cytoprotection/drug effects , Cytoprotection/physiology , Dose-Response Relationship, Drug , Humans , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Keratinocytes/metabolism , Lipopolysaccharides/toxicity , Mouth Mucosa/cytology , Mouth Mucosa/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , TOR Serine-Threonine Kinases/metabolism
20.
Mol Med Rep ; 24(4)2021 10.
Article En | MEDLINE | ID: mdl-34328196

Inflammation and oxidative stress have indispensable roles in the development of acute lung injury (ALI). MicroRNA (miRNA/miR)­351­5p was initially identified as a myogenesis­associated miRNA; however, its role in lipopolysaccharide (LPS)­induced ALI remains unclear. The aim of the present study was to investigate the role and potential mechanisms of miR­351­5p in ALI. ALI was induced through a single intratracheal injection of LPS for 12 h, and miR­351­5p agomir, antagomir or their corresponding negative controls were injected into the tail vein before LPS stimulation. Compound C, 2',5'­dideoxyadenosine and H89 were used to inhibit AMP­activated protein kinase (AMPK), adenylate cyclase and protein kinase A (PKA), respectively. miR­351­5p levels in the lungs were significantly increased in response to LPS injection. miR­351­5p antagomir alleviated, while miR­351­5p agomir aggravated LPS­induced oxidative stress and inflammation in the lungs. The present results also demonstrated that miR­351­5p antagomir attenuated LPS­induced ALI via activating AMPK, and that the cAMP/PKA axis was required for the activation of AMPK by the miR­351­5p antagomir. In conclusion, the present study indicated that miR­351­5p aggravated LPS­induced ALI via inhibiting AMPK, suggesting that targeting miR­351­5p may help to develop efficient therapeutic approaches for treating ALI.


AMP-Activated Protein Kinases/antagonists & inhibitors , Acute Lung Injury/genetics , Acute Lung Injury/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Acute Lung Injury/chemically induced , Animals , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Disease Models, Animal , Inflammation/genetics , Lipopolysaccharides/toxicity , Male , Mice, Inbred C57BL , MicroRNAs/agonists , MicroRNAs/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Oxidative Stress/genetics
...