Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 138
Filter
1.
FASEB J ; 36(1): e22075, 2022 01.
Article in English | MEDLINE | ID: mdl-34919285

ABSTRACT

Long non-coding RNAs (lncRNAs) regulate neurological damage in cerebral ischemia-reperfusion injury (CIRI). This study aimed to investigate the biological roles of lncRNA CEBPA-AS1 in CIRI. Middle cerebral artery occlusion and ischemia-reperfusion injury (MCAO/IR) rat model and oxygen-glucose deprivation and reoxygenation (OGD/R) cell lines were generated; the expression of CEBPA-AS1 was evaluated by qRT-PCR. The effects of CEBPA-AS1 on cell apoptosis and nerve damage were examined. The downstream microRNA (miRNA) and mRNA of CEBPA-AS1 were predicted and verified. We found that overexpression of CEBPA-AS1 could attenuate MCAO/IR-induced nerve damage and neuronal apoptosis in the rat model. Knockdown of CEBPA-AS1 aggravated cell apoptosis and enhanced the production of LDH and MDA in the OGD/R cells. Upon examining the molecular mechanisms, we found that CEBPA-AS1 stimulated APPL1 expression by combining with miR-340-5p, thereby regulating the APPL1/LKB1/AMPK pathway. In the rescue experiments, CEBPA-AS1 overexpression was found to attenuate OGD/R-induced cell apoptosis and MCAO/IR induced nerve damage, while miR-340-5p reversed these effects of CEBPA-AS1. In conclusion, CEBPA-AS1 could decrease CIRI by sponging miR-340-5, regulating the APPL1/LKB1/AMPK pathway.


Subject(s)
AMP-Activated Protein Kinase Kinases/biosynthesis , AMP-Activated Protein Kinases/biosynthesis , Adaptor Proteins, Signal Transducing/biosynthesis , Cerebrovascular Disorders/metabolism , MicroRNAs/biosynthesis , Nerve Tissue Proteins/biosynthesis , RNA, Long Noncoding/biosynthesis , Reperfusion Injury/metabolism , Signal Transduction , AMP-Activated Protein Kinase Kinases/genetics , AMP-Activated Protein Kinases/genetics , Adaptor Proteins, Signal Transducing/genetics , Animals , Apoptosis , Cerebrovascular Disorders/genetics , Cerebrovascular Disorders/pathology , Disease Models, Animal , Gene Expression Regulation , MicroRNAs/genetics , Nerve Tissue Proteins/genetics , RNA, Long Noncoding/genetics , Rats , Rats, Sprague-Dawley , Reperfusion Injury/genetics , Reperfusion Injury/pathology
2.
Cell Tissue Res ; 380(1): 173-189, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31838605

ABSTRACT

Male reproductive dysfunction represents one of the overlooked consequences of diabetes that still deserve more scientific attention. We designed this study to explore the therapeutic potential of hydroxytyrosol (HT) on diabetic testicular damage and to investigate its relationship with adenosine monophosphate-activated protein kinase (AMPK) expression. In this context, 30 adult male Wistar rats were utilized and subdivided into control, diabetic and HT-treated diabetic groups. Testicular sections were prepared for histopathological examination and immunohistochemical detection of 8-hydroxy-2'-deoxyguanosine, Sertoli cell vimentin, myoid cell α-SMA, androgen receptors and caspase-3. We also assessed oxidative enzymatic and lipid peroxidation biochemical profiles, sperm count, morphology and motility. Real-time PCR of AMPK expression in tissue homogenate was performed. We observed that HT restored testicular histopathological structure and significantly reduced oxidative DNA damage and the apoptotic index. The HT-treated group also exhibited significantly higher Sertoli cell vimentin, myoid cell α-SMA and androgen receptor immune expression than the diabetic group. A rescue of the oxidative enzymatic activity, lipid peroxidation profiles, sperm count, morphology and motility to control levels was also evident in the HT-treated group. Significant upregulation of AMPK mRNA expression in the HT-treated group clarified the role of AMPK as an underlying molecular interface of the ameliorative effects of HT. We concluded that HT exhibited tangible antioxidant and antiapoptotic impacts on the testicular cytomorphological and immunohistochemical effects of experimentally induced diabetes. Furthermore, AMPK has an impactful role in the molecular machinery of these effects.


Subject(s)
AMP-Activated Protein Kinases/biosynthesis , Antioxidants/pharmacology , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Phenylethyl Alcohol/analogs & derivatives , Testicular Diseases/drug therapy , Testicular Diseases/enzymology , 8-Hydroxy-2'-Deoxyguanosine/metabolism , AMP-Activated Protein Kinases/metabolism , Animals , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Experimental/pathology , Male , Phenylethyl Alcohol/pharmacology , Rats , Rats, Wistar , Testicular Diseases/etiology , Testis/drug effects , Testis/enzymology , Testis/pathology
3.
Hum Mol Genet ; 26(2): 305-319, 2017 01 15.
Article in English | MEDLINE | ID: mdl-28065882

ABSTRACT

Protein misfolding caused by inherited mutations leads to loss of protein function and potentially toxic 'gain of function', such as the dominant P23H rhodopsin mutation that causes retinitis pigmentosa (RP). Here, we tested whether the AMPK activator metformin could affect the P23H rhodopsin synthesis and folding. In cell models, metformin treatment improved P23H rhodopsin folding and traffic. In animal models of P23H RP, metformin treatment successfully enhanced P23H traffic to the rod outer segment, but this led to reduced photoreceptor function and increased photoreceptor cell death. The metformin-rescued P23H rhodopsin was still intrinsically unstable and led to increased structural instability of the rod outer segments. These data suggest that improving the traffic of misfolding rhodopsin mutants is unlikely to be a practical therapy, because of their intrinsic instability and long half-life in the outer segment, but also highlights the potential of altering translation through AMPK to improve protein function in other protein misfolding diseases.


Subject(s)
AMP-Activated Protein Kinases/genetics , Metformin/administration & dosage , Retinal Degeneration/genetics , Retinitis Pigmentosa/genetics , Rhodopsin/genetics , AMP-Activated Protein Kinases/biosynthesis , Animals , Disease Models, Animal , Humans , Mice , Mutant Proteins/genetics , Photoreceptor Cells/drug effects , Photoreceptor Cells/pathology , Protein Folding/drug effects , Proteostasis Deficiencies/genetics , Proteostasis Deficiencies/pathology , Rats , Retinal Degeneration/drug therapy , Retinal Degeneration/pathology , Retinal Rod Photoreceptor Cells/drug effects , Retinal Rod Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/pathology , Retinitis Pigmentosa/drug therapy , Retinitis Pigmentosa/pathology , Rhodopsin/chemistry , Rod Cell Outer Segment/drug effects , Rod Cell Outer Segment/pathology , Transcriptional Activation/drug effects
4.
J Neuroinflammation ; 16(1): 110, 2019 May 25.
Article in English | MEDLINE | ID: mdl-31128596

ABSTRACT

BACKGROUND: Microglia-mediated neuroinflammation is important in Alzheimer's disease (AD) pathogenesis. Extracellular deposition of ß-amyloid (Aß), a major pathological hallmark of AD, can induce microglia activation. Adiponectin (APN), an adipocyte-derived adipokine, exerts anti-inflammatory effects in the periphery and brain. Chronic APN deficiency leads to cognitive impairment and AD-like pathologies in aged mice. Here, we aim to study the role of APN in regulating microglia-mediated neuroinflammation in AD. METHODS: Inflammatory response of cultured microglia (BV2 cells) to AßO and effects of APN were studied by measuring levels of proinflammatory cytokines (tumor necrosis factor α [TNFα] and interleukin-1ß [IL-1ß]) in cultured medium before and after exposure to AßO, with and without APN pretreatment. Adiponectin receptor 1 (AdipoR1) and receptor 2 (AdipoR2) were targeted by small interference RNA. To study the neuroprotective effect of APN, cultured HT-22 hippocampal cells were treated with conditioned medium of AßO-exposed BV2 cells or were co-cultured with BV2 cells in transwells. The cytotoxicity of HT-22 hippocampal cells was assessed by MTT reduction. We generated APN-deficient AD mice (APN-/-5xFAD) by crossing APN-knockout mice with 5xFAD mice to determine the effects of APN deficiency on microglia-mediated neuroinflammation in AD. RESULTS: AdipoR1 and AdipoR2 were expressed in BV2 cells and microglia of mice. Pretreatment with APN for 2 h suppressed TNFα and IL-1ß release induced by AßO in BV2 cells. Additionally, APN rescued the decrease of AMPK phosphorylation and suppressed nuclear translocation of nuclear factor kappa B (NF-κB) induced by AßO. Compound C, an inhibitor of AMPK, abolished these effects of APN. Knockdown of AdipoR1, but not AdipoR2 in BV2 cells, inhibited the ability of APN to suppress proinflammatory cytokine release induced by AßO. Moreover, pretreatment with APN inhibited the cytotoxicity of HT-22 cells co-cultured with AßO-exposed BV2 cells. Lastly, APN deficiency exacerbated microglia activation in 9-month-old APN-/-5xFAD mice associated with upregulation of TNFα and IL-1ß in the cortex and hippocampus. CONCLUSIONS: Our findings demonstrate that APN inhibits inflammatory response of microglia to AßO via AdipoR1-AMPK-NF-κB signaling, and APN deficiency aggravates microglia activation and neuroinflammation in AD mice. APN may be a novel therapeutic agent for inhibiting neuroinflammation in AD.


Subject(s)
AMP-Activated Protein Kinases/biosynthesis , Adiponectin/pharmacology , Amyloid beta-Peptides/toxicity , Microglia/metabolism , NF-kappa B/biosynthesis , Peptide Fragments/toxicity , Receptors, Adiponectin/biosynthesis , AMP-Activated Protein Kinases/antagonists & inhibitors , Amyloid beta-Peptides/antagonists & inhibitors , Animals , Dose-Response Relationship, Drug , Inflammation/chemically induced , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/drug effects , NF-kappa B/antagonists & inhibitors , Peptide Fragments/antagonists & inhibitors , Receptors, Adiponectin/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/physiology
5.
PLoS Genet ; 12(11): e1006449, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27902686

ABSTRACT

Metformin is used as a first-line therapy for type 2 diabetes (T2D) and prescribed for numerous other diseases. However, its mechanism of action in the liver has yet to be characterized in a systematic manner. To comprehensively identify genes and regulatory elements associated with metformin treatment, we carried out RNA-seq and ChIP-seq (H3K27ac, H3K27me3) on primary human hepatocytes from the same donor treated with vehicle control, metformin or metformin and compound C, an AMP-activated protein kinase (AMPK) inhibitor (allowing to identify AMPK-independent pathways). We identified thousands of metformin responsive AMPK-dependent and AMPK-independent differentially expressed genes and regulatory elements. We functionally validated several elements for metformin-induced promoter and enhancer activity. These include an enhancer in an ataxia telangiectasia mutated (ATM) intron that has SNPs in linkage disequilibrium with a metformin treatment response GWAS lead SNP (rs11212617) that showed increased enhancer activity for the associated haplotype. Expression quantitative trait locus (eQTL) liver analysis and CRISPR activation suggest that this enhancer could be regulating ATM, which has a known role in AMPK activation, and potentially also EXPH5 and DDX10, its neighboring genes. Using ChIP-seq and siRNA knockdown, we further show that activating transcription factor 3 (ATF3), our top metformin upregulated AMPK-dependent gene, could have an important role in gluconeogenesis repression. Our findings provide a genome-wide representation of metformin hepatic response, highlight important sequences that could be associated with interindividual variability in glycemic response to metformin and identify novel T2D treatment candidates.


Subject(s)
AMP-Activated Protein Kinases/biosynthesis , Activating Transcription Factor 3/genetics , Ataxia Telangiectasia Mutated Proteins/biosynthesis , Diabetes Mellitus, Type 2/drug therapy , Liver/metabolism , AMP-Activated Protein Kinases/genetics , Adaptor Proteins, Signal Transducing/genetics , Ataxia Telangiectasia Mutated Proteins/genetics , DEAD-box RNA Helicases/genetics , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Enhancer Elements, Genetic , Gene Knockdown Techniques , Gluconeogenesis/genetics , Haplotypes , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Linkage Disequilibrium , Liver/drug effects , Metformin/adverse effects , Metformin/therapeutic use , Polymorphism, Single Nucleotide
6.
Mol Cell Biochem ; 449(1-2): 219-226, 2018 Dec.
Article in English | MEDLINE | ID: mdl-29675630

ABSTRACT

Autosomal dominant polycystic kidney disease (ADPKD) is a common heritable human disease. Recently, the role of repressed autophagy in ADPKD has drawn increasing attention. Here, we investigate the mechanism underlying the effect of Saikosaponin-d (SSd), a sarcoplasmic/endoplasmic reticulum Ca2+ ATPase pump (SERCA) inhibitor. We show that SSd suppresses proliferation in ADPKD cells by up-regulating autophagy. We found that treatment with SSd results in the accumulation of intracellular calcium, which in turn activates the CaMKKß-AMPK signalling cascade, inhibits mTOR signalling and induces autophagy. Conversely, we also found that treatment with an autophagy inhibitor (3-methyladenine), AMPK inhibitor (Compound C), CaMKKß inhibitor (STO-609) and intracellular calcium chelator (BAPTA/AM) could reduce autophagy puncta formation mediated by SSd. Our results demonstrated that SSd induces autophagy through the CaMKKß-AMPK-mTOR signalling pathway in ADPKD cells, indicating that SSd might be a potential therapy for ADPKD and that SERCA might be a new target for ADPKD treatment.


Subject(s)
AMP-Activated Protein Kinases/biosynthesis , Autophagy/drug effects , Calcium-Calmodulin-Dependent Protein Kinase Kinase/biosynthesis , Cell Proliferation/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Oleanolic Acid/analogs & derivatives , Saponins/pharmacokinetics , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/biosynthesis , AMP-Activated Protein Kinases/genetics , Calcium-Calmodulin-Dependent Protein Kinase Kinase/genetics , Cell Line, Transformed , Humans , Oleanolic Acid/pharmacokinetics , Polycystic Kidney, Autosomal Dominant/drug therapy , Polycystic Kidney, Autosomal Dominant/genetics , Polycystic Kidney, Autosomal Dominant/metabolism , Polycystic Kidney, Autosomal Dominant/pathology , Signal Transduction/genetics , TOR Serine-Threonine Kinases/genetics
7.
J Pharmacol Sci ; 138(3): 161-166, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30316692

ABSTRACT

As one kind of complications of pregnancy, gestational diabetes mellitus (GDM) can influence the health of maternal-child in clinical practice. The C57 BL/KsJdb/+(db/+) mice, genetic GDM model, and C57 BL/KsJ+/+ (wild-type) mice were purchased and classified into three groups: normal pregnancy (C57 BL/KsJ+/+), GDM (C57 BL/KsJdb/+), and GDM plus Mogroside IIIE (20.0 mg/kg) group. GDM symptoms (maternal body weight, serum glucose, and insulin levels), glucose and insulin tolerance, and reproductive outcome (body weight at birth and litter size of offspring) were investigated. The inflammatory factors such as IL-1ß, IL-6, and TNF-α in the serum and the pancreas were detected by ELISA and qRT-PCR, while the expression of pAMPK, AMPK, pHDAC4, HDAC4, and G6Pase in the livers were analyzed by Western Blot. Mogroside IIIE greatly improved glucose metabolism, insulin tolerance, and reproductive outcome of the GDM mice. Moreover, Mogroside IIIE treatment significantly decreased inflammatory factors expression and relieved GDM symptoms through enhanced AMPK activation, inhibited HDAC4 expression, and reduced production of G6Pase. The alleviation of GDM by Mogroside IIIE was mediated by elevated AMPK activation, which in turn inhibited HDAC4 phosphorylation, and eventually down-regulated G6Pase expression and activity.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Diabetes, Gestational/drug therapy , Glucosides/pharmacology , Glucosides/therapeutic use , Signal Transduction/drug effects , Triterpenes/pharmacology , Triterpenes/therapeutic use , AMP-Activated Protein Kinases/biosynthesis , Animals , Blood Glucose/drug effects , Female , Glucose-6-Phosphatase/biosynthesis , Histone Deacetylases/biosynthesis , Inflammation Mediators/blood , Inflammation Mediators/metabolism , Insulin/blood , Litter Size/drug effects , Liver/metabolism , Mice , Mice, Transgenic , Pancreas/metabolism , Phosphorylation/drug effects , Pregnancy
8.
Exp Cell Res ; 354(1): 18-24, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28300563

ABSTRACT

Impaired insulin-stimulated glucose uptake in skeletal muscle serves a critical role in the development of insulin resistance (IR), whereas the precise mechanism of the process remains unknown. Recently, the evolutionarily conserved, stress-inducible protein Sestrin2 (Sesn2) has been proposed to play a protective role against obesity-induced IR and diabetes. Activation of Sesn2 may activate AMP-activated protein kinase (AMPK) accompanied by suppression of mammalian target of rapamycin (mTOR), which may ultimately lead to autophagy induction. In view of the potential protective effects of autophagy on the physiological and the pathological regulatory processes via the regulation of energy homeostasis and metabolism, we investigated the effects of Sesn2 on the components of the insulin signaling pathway and insulin-stimulated glucose uptake in palmitate-induced insulin-resistant C2C12 myotubes. We showed that Sesn2 effectively restored the impaired insulin signaling. Moreover, autophagic activity decreased in response to palmitate, whereas Sesn2 significantly reversed the palmitate-suppressed autophagic signaling in this context. Our findings further revealed that Sesn2-induced autophagy contributed to restore the impaired insulin signaling through the activation of AMPK signal. Even in the presence of palmitate, Sesn2 up-regulation maintained insulin sensitivity and glucose metabolism via AMPK-dependent autophagic activation.


Subject(s)
AMP-Activated Protein Kinases/genetics , Insulin Resistance/genetics , Nuclear Proteins/genetics , Obesity/genetics , AMP-Activated Protein Kinases/biosynthesis , Animals , Autophagy/genetics , Gene Expression Regulation , Glucose/metabolism , Humans , Insulin/metabolism , Mice , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Nuclear Proteins/metabolism , Obesity/metabolism , Obesity/pathology , Palmitates/metabolism , Peroxidases , Phosphorylation , TOR Serine-Threonine Kinases/biosynthesis , TOR Serine-Threonine Kinases/genetics
9.
J Biol Chem ; 291(35): 18536-46, 2016 08 26.
Article in English | MEDLINE | ID: mdl-27405764

ABSTRACT

Although numerous biological functions of the activating transcription factor 4 (ATF4) have been identified, a direct effect of ATF4 on alcoholic liver steatosis has not been described previously. The aim of our current study is to investigate the role of ATF4 in alcoholic liver steatosis and elucidate the underlying mechanisms. Here, we showed that the expression of ATF4 is induced by ethanol in hepatocytes in vitro and in vivo, and liver-specific ATF4 knock-out mice are resistant to ethanol-induced liver steatosis, associated with stimulated hepatic AMP-activated protein kinase (AMPK) activity. Furthermore, adenovirus-mediated AMPK knockdown significantly reversed the suppressive effects of ATF4 deficiency on ethanol-induced liver steatosis in mice. In addition, ethanol-fed ATF4 knock-out mice exhibit AMPK-dependent inhibition of fatty acid synthase and stimulation of carnitine palmitoyltransferase 1 (CPT1) in the liver. Moreover, hepatic Tribbles homolog 3 (TRB3) expression was stimulated by ethanol in an ATF4-dependent manner, and adenovirus-mediated TRB3 knockdown blocked ATF4-dependent ethanol-induced AMPK inhibition and triglyceride accumulation in AML-12 cells. Finally, TRB3 directly interacted with AMPK to suppress its phosphorylation. Taken together, these results identify the ATF4-TRB3-AMPK axis as a novel pathway responsible for ethanol-induced liver steatosis.


Subject(s)
Activating Transcription Factor 4/metabolism , Fatty Liver, Alcoholic/metabolism , Hepatocytes/metabolism , Liver/metabolism , Signal Transduction , Triglycerides/biosynthesis , AMP-Activated Protein Kinases/biosynthesis , AMP-Activated Protein Kinases/genetics , Activating Transcription Factor 4/genetics , Animals , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/genetics , Cell Line , Ethanol/adverse effects , Ethanol/pharmacology , Fatty Liver, Alcoholic/genetics , Fatty Liver, Alcoholic/pathology , Hepatocytes/pathology , Liver/pathology , Male , Mice , Mice, Knockout , Organ Specificity/genetics , Triglycerides/genetics
10.
J Biol Chem ; 291(20): 10625-34, 2016 May 13.
Article in English | MEDLINE | ID: mdl-27013659

ABSTRACT

In a microarray study, we found that hepatic miR-291b-3p was significantly increased in leptin-receptor-deficient type 2 mice (db/db), a mouse model of diabetes. The function of miR-291b-3p is unknown. The potential role of miR-291b-3p in regulating hepatic lipid metabolism was explored in this study. High-fat diet (HFD)- and chow-fed mice were injected with an adenovirus expressing a miR-291b-3p inhibitor and a miR-291b-3p mimic through the tail vein. Hepatic lipids and lipogenic gene expression were analyzed. Additionally, gain- and loss-of-function studies were performed in vitro to identify direct targets of miR-291b-3p. MiR-291b-3p expression and the protein levels of sterol regulatory element-binding protein 1 (SREBP1) and fatty acid synthase (FAS) were increased in the steatotic liver of db/db mice and HFD-fed mice versus their respective controls. Inhibition of hepatic miR-291b-3p expression prevented increases in hepatic lipogenesis and steatosis in HFD-fed mice. The opposite was observed when miR-291b-3p was overexpressed in the livers of chow-fed C57BL/6J wild-type mice. In vitro studies revealed that silencing of miR-291b-3p in NCTC1469 hepatic cells ameliorated oleic acid/palmitic acid mixture-induced elevation of cellular triglycerides. Importantly, we identified AMP-activated protein kinase (AMPK)-α1 as a direct target of miR-291b-3p. Using metformin, an activator of AMPK, we showed that AMPK activation-induced inhibition of hepatic lipid accumulation was accompanied by reduced expression of miR-291b-3p in the liver. Liver miR-291b-3p promoted hepatic lipogenesis and lipid accumulation in mice. AMPKα1 is a direct target of miR-291b-3p. In conclusion, our findings indicate that miR-291b-3p promotes hepatic lipogenesis by suppressing AMPKα1 expression and activity, indicating the therapeutic potential of miR-291b-3p inhibitors in fatty liver disease.


Subject(s)
AMP-Activated Protein Kinases/biosynthesis , Dietary Fats/adverse effects , Fatty Liver/metabolism , Lipogenesis/drug effects , Liver/metabolism , MicroRNAs/biosynthesis , AMP-Activated Protein Kinases/genetics , Animals , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Dietary Fats/pharmacology , Enzyme Activation/drug effects , Enzyme Activation/genetics , Fatty Acid Synthase, Type I/genetics , Fatty Acid Synthase, Type I/metabolism , Fatty Liver/chemically induced , Fatty Liver/genetics , Fatty Liver/pathology , Lipogenesis/genetics , Liver/pathology , Metformin/pharmacology , Mice , MicroRNAs/genetics , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism
11.
Biochim Biophys Acta Mol Basis Dis ; 1863(6): 1479-1491, 2017 06.
Article in English | MEDLINE | ID: mdl-28433710

ABSTRACT

Islet amyloid polypeptide (IAPP) exerts its biological effects by participating in the regulation of glucose metabolism and cell apoptosis. The main goal of the present study was to investigate the expression of IAPP in degenerated intervertebral disc tissue and IAPP's modulation of extracellular matrix (ECM) catabolic and anabolic genes in human AF cells. We found that the expression of IAPP, the calcitonin receptor, and receptor activity modifying protein decreased considerably in AF cells during the progression of intervertebral disc degeneration (IDD). Meanwhile, transfection with pLV-siIAPP decreased the expression of IAPP and its receptors and reduced glucose uptake and the expression of aggrecan, Col2A1, and BG. Down-regulation of IAPP also induced a significant increase in reactive oxygen species generation in AF cells, along with a decrease in matrix metalloproteinases and an increase in the concentration of cellular Ca2+, ultimately leading to death. Further analysis revealed that siIAPP intervention promoted the release of cytochrome c from mitochondria, resulting in the activation of Caspase-3 and Caspase-9. In contrast, significantly decreased expression of Caspase-3 and Caspase-9 was observed in AF cells transfected with pLV-IAPP. The concentrations of Fas and FasL proteins were significantly decreased in AF cells transfected with PLV-IAPP, while activation of the Fas/FasL system and cell death were induced by siIAPP intervention. Mechanistically, AMPK/Akt-mTOR signaling pathways were involved. In conclusion, down-regulation of IAPP expression induces the death of human AF cells via mitochondrial and death receptor pathways, potentially offering a novel therapeutic target for the treatment of IDD.


Subject(s)
Annulus Fibrosus/metabolism , Down-Regulation , Fas Ligand Protein/biosynthesis , Intervertebral Disc Degeneration/metabolism , Islet Amyloid Polypeptide/biosynthesis , Mitochondrial Proteins/biosynthesis , fas Receptor/biosynthesis , AMP-Activated Protein Kinases/biosynthesis , AMP-Activated Protein Kinases/genetics , Adolescent , Adult , Annulus Fibrosus/pathology , Caspase 3/biosynthesis , Caspase 3/genetics , Caspase 9/biosynthesis , Caspase 9/genetics , Cell Death , Fas Ligand Protein/genetics , Female , Humans , Intervertebral Disc Degeneration/genetics , Intervertebral Disc Degeneration/pathology , Islet Amyloid Polypeptide/genetics , Male , Middle Aged , Mitochondrial Proteins/genetics , Proto-Oncogene Proteins c-akt/biosynthesis , Proto-Oncogene Proteins c-akt/genetics , TOR Serine-Threonine Kinases/biosynthesis , TOR Serine-Threonine Kinases/genetics , fas Receptor/genetics
12.
Tumour Biol ; 39(6): 1010428317709676, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28639909

ABSTRACT

NRAGE has been reported to be overexpressed in cancer cells, especially in lung cancer cells. To determine the role of NRAGE in non-small-cell lung cancer cells, we investigated the effects of NRAGE on autophagy in non-small-cell lung cancer cells. Human A549 and H1299 cells were transfected with NRAGE-specific small-interfering RNA. The Cell Counting Kit-8 and plate clone assay showed that downregulation of NRAGE could induce the proliferation in A549 and H1299 cells. In addition, our data suggested that downregulation of NRAGE enhances autophagosome formation by immunofluorescence. We found that knockdown of NRAGE induced autophagy, together with downregulation of P62 and upregulation of LC3-II protein. Furthermore, to elucidate the mechanism of NRAGE in suppressing autophagy, the protein expressions of AMPK, Ulk1, and Atg13 were assessed. Collectively, these results demonstrate the effective anti-autophagic of NRAGE in non-small-cell lung cancer cells through AMPK/Ulk1/Atg13 autophagy signaling pathways. Therefore, NRAGE could be used as a potential therapeutic target for lung cancer.


Subject(s)
AMP-Activated Protein Kinases/genetics , Adaptor Proteins, Signal Transducing/genetics , Antigens, Neoplasm/genetics , Autophagy-Related Protein-1 Homolog/genetics , Autophagy-Related Proteins/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Intracellular Signaling Peptides and Proteins/genetics , Neoplasm Proteins/genetics , A549 Cells , AMP-Activated Protein Kinases/biosynthesis , Adaptor Proteins, Signal Transducing/biosynthesis , Autophagy/genetics , Autophagy-Related Protein-1 Homolog/biosynthesis , Autophagy-Related Proteins/biosynthesis , Carcinoma, Non-Small-Cell Lung/pathology , Gene Expression Regulation, Neoplastic , Humans , Intracellular Signaling Peptides and Proteins/biosynthesis , Neoplasm Proteins/antagonists & inhibitors , Phosphorylation , RNA, Small Interfering , Signal Transduction
13.
Tumour Biol ; 37(4): 4803-11, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26520441

ABSTRACT

Activation of AMP-activated protein kinase (AMPK) suppressed mammalian target of rapamycin (mTOR) pathway, resulting in impaired cancer cell proliferation. Two cohorts (50 and 1072 cases) of patients with resected gastric adenocarcinoma were enrolled in the study. Immunohistochemical staining for p-AMPKa, p-ACC, p-mTOR, p-S6, and p-4EBP1 was performed on the 50-patient cohort. Tissue microarray blocks containing samples from 1072 patients of Chinese ethnicity were used for the immunohistochemical detection of p-AMPKa and p-S6 levels. p-AMPK and p-ACC were frequently inactivated in both cohorts of gastric cancer samples, while p-mTOR, p-S6, and p-4EBP1 were frequently activated in the small cohort of gastric cancer. However, only levels of p-AMPKa and p-S6 were associated with the overall survival of gastric cancer patients. In the larger 1072-patient cohort, downregulation of p-AMPKa and upregulation of p-S6 were associated with tumor progression and were independent predictors of survival after resection of primary gastric cancer. Therefore, reciprocal expression of p-AMPKa and p-S6 may be promising prognostic biomarkers in patients with gastric cancer.


Subject(s)
AMP-Activated Protein Kinases/biosynthesis , Biomarkers, Tumor/biosynthesis , Ribosomal Protein S6 Kinases, 70-kDa/biosynthesis , Stomach Neoplasms/genetics , AMP-Activated Protein Kinases/genetics , Acetyl-CoA Carboxylase/biosynthesis , Acetyl-CoA Carboxylase/genetics , Adaptor Proteins, Signal Transducing/biosynthesis , Adaptor Proteins, Signal Transducing/genetics , Adult , Aged , Biomarkers, Tumor/genetics , Cell Cycle Proteins , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic , Humans , Middle Aged , Neoplasm Proteins/biosynthesis , Phosphoproteins/biosynthesis , Phosphoproteins/genetics , Phosphorylation , Prognosis , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Signal Transduction , Stomach Neoplasms/pathology , TOR Serine-Threonine Kinases/biosynthesis
14.
Tumour Biol ; 37(4): 4295-304, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26493996

ABSTRACT

Hepatocellular carcinoma (HCC) is a crucial health issue worldwide. High glucose-regulated protein 94 (GRP94) expression has been observed in different types of cancer, suggesting a link between tumor progression and GRP94 expression. However, the mechanisms underlying the role of GRP94 in HCC progression remain unclear. We used specific small hairpin RNA (shRNA) to manipulate GRP94 expression in HCC cells. Tissue arrays, MTT assays, xCELLigence assays, and in vivo xenograft model were performed to identify clinicopathological correlations and to analyze cell growth. We found that high GRP94 expression reflected a poor response and a lower survival rate. In vitro and in vivo studies showed that silencing GRP94 suppressed cancer progression. Mechanistically, GRP94 knockdown reduced AKT, phospho-AKT, and eNOS levels but did not influence the AMPK pathway. Our results demonstrated that GRP94 is a key molecule in HCC progression that modulates the AKT pathway and eNOS levels. Our findings suggest that GRP94 may be a new prognostic and therapeutic target for HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Membrane Glycoproteins/genetics , Nitric Oxide Synthase Type III/biosynthesis , Proto-Oncogene Proteins c-akt/biosynthesis , AMP-Activated Protein Kinases/biosynthesis , AMP-Activated Protein Kinases/genetics , Animals , Carcinogenesis/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Disease Progression , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/pathology , Membrane Glycoproteins/antagonists & inhibitors , Mice , Nitric Oxide Synthase Type III/genetics , Proto-Oncogene Proteins c-akt/genetics , Xenograft Model Antitumor Assays
15.
Int J Med Sci ; 13(4): 260-70, 2016.
Article in English | MEDLINE | ID: mdl-27076782

ABSTRACT

The protein deacetylase sirtuin 1 (SIRT1) and activate peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) pathway drives the muscular fiber-type switching, and can directly regulate the biophysiological functions of skeletal muscle. To investigate whether 12-week swimming exercise training modulates the SIRT1/PGC-1α pathway associated proteins expression in rats of different age. Male 3-month-old (3M), 12-month-old (12M) and 18-month-old (18M) Sprague-Dawley rats were used and assigned to sedentary control (C) or 12-week swimming exercise training (E) and divided into six groups: 3MC (n = 8), 12MC (n = 6), 18MC (n = 8), 3ME (n = 8), 12ME (n = 5) and 18ME (n = 6). Body weight, muscle weight, epididymal fat mass and muscle morphology were performed at the end of the experiment. The protein levels of SIRT1, PGC-1α, AMPK and FOXO3a in the gastrocnemius and soleus muscles were examined. The SIRT1, PGC-1α and AMPK levels in the gastrocnemius and soleus muscles were up-regulated in the three exercise training groups than three control groups. The FOXO3a level in the 12ME group significantly increased in the gastrocnemius muscles than 12MC group, but significantly decreased in the soleus muscles. In 3-, 12- and 18-month-old rats with and without exercise, there was a significant main effect of exercise on PGC-1α, AMPK and FOXO3a in the gastrocnemius muscles, and SIRT1, PGC-1α and AMPK in the soleus muscles. Our result suggests that swimming training can regulate the SIRT1/PGC-1α, AMPK and FOXO3a proteins expression of the soleus muscles in aged rats.


Subject(s)
AMP-Activated Protein Kinases/biosynthesis , Aging/metabolism , Forkhead Box Protein O3/biosynthesis , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/biosynthesis , Sirtuin 1/biosynthesis , AMP-Activated Protein Kinases/metabolism , Aging/genetics , Aging/physiology , Animals , Forkhead Box Protein O3/metabolism , Gene Expression Regulation , Humans , Muscle, Skeletal/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Physical Conditioning, Animal , Rats , Rats, Sprague-Dawley , Sirtuin 1/metabolism , Swimming
16.
Molecules ; 21(3): 374, 2016 Mar 18.
Article in English | MEDLINE | ID: mdl-26999101

ABSTRACT

Non-small-cell lung cancer (NSCLC) dominates over 85% of all lung cancer cases. Epidermal growth factor receptor (EGFR) activating mutation is a common situation in NSCLC. In the clinic, molecular-targeting with Gefitinib as a tyrosine kinase inhibitor (TKI) for EGFR downstream signaling is initially effective. However, drug resistance frequently happens due to additional mutation on EGFR, such as substitution from threonine to methionine at amino acid position 790 (T790M). In this study, we screened a traditional Chinese medicine (TCM) compound library consisting of 800 single compounds in TKI-resistance NSCLC H1975 cells, which contains substitutions from leucine to arginine at amino acid 858 (L858R) and T790M mutation on EGFR. Attractively, among these compounds there are 24 compounds CC50 of which was less than 2.5 µM were identified. We have further investigated the mechanism of the most effective one, Digitoxin. It showed a significantly cytotoxic effect in H1975 cells by causing G2 phase arrest, also remarkably activated 5' adenosine monophosphate-activated protein kinase (AMPK). Moreover, we first proved that Digitoxin suppressed microtubule formation through decreasing α-tubulin. Therefore, it confirmed that Digitoxin effectively depressed the growth of TKI-resistance NSCLC H1975 cells by inhibiting microtubule polymerization and inducing cell cycle arrest.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Cardiac Glycosides/administration & dosage , Digitoxin/administration & dosage , Drug Resistance, Neoplasm/genetics , Microtubules/drug effects , AMP-Activated Protein Kinases/biosynthesis , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/genetics , Cardiac Glycosides/chemistry , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Digitoxin/chemistry , Drug Resistance, Neoplasm/drug effects , ErbB Receptors/biosynthesis , ErbB Receptors/genetics , Gefitinib , Gene Expression Regulation, Neoplastic , Humans , Medicine, Chinese Traditional , Microtubules/pathology , Mutation , Quinazolines/therapeutic use , Tubulin/biosynthesis , Tubulin/genetics
17.
Aviakosm Ekolog Med ; 50(1): 28-34, 2016.
Article in Russian | MEDLINE | ID: mdl-27344854

ABSTRACT

The signaling processes initiating proteolytic events in m. soleus of humans during short-term exposure in the non-weight bearing conditions were analyzed. Dry immersion (DI) was used to induce weight deprivation over 3 days. Western blotting was used to define the IRS-1 content, total and phosphorylated neuronal NO-synthase (nNOS), AMP-activated protein kinase (AMPK) that control the anabolic and catabolic pathways, and concentrations of cytoskeletal protein desmin and Ca²âº-activated protease calpin. Already on day-3 of DI calpain-dependent proteolysis manifests itself by reductions in both the total content and level of nNOS phosphorilation. Moreover, AMPK phosphorilation was decreased drastically.


Subject(s)
AMP-Activated Protein Kinases/biosynthesis , Muscle, Skeletal/metabolism , Nitric Oxide Synthase Type I/biosynthesis , Proteolysis , Calpain/biosynthesis , Desmin/biosynthesis , Humans , Immersion , Insulin Receptor Substrate Proteins/biosynthesis , Metabolism/genetics , Muscle, Skeletal/physiology
18.
J Mol Cell Cardiol ; 86: 62-74, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26196303

ABSTRACT

BACKGROUND AND PURPOSE: This study aims to investigate whether and how pharmacological activation of AMP-activated protein kinase (AMPK) improves endothelial function by suppressing mitochondrial ROS-associated endoplasmic reticulum stress (ER stress) in the endothelium. Experimental approach Palmitate stimulation induced mitochondrial fission and ER stress-associated endothelial dysfunction. The effects of AMPK activators salicylate and AICA riboside (AICAR) on mitochondrial ROS production, Drp1 phosphorylation, mitochondrial fission, ER stress, thioredoxin-interacting protein (TXNIP)/NLRP3 inflammasome activation, inflammation, cell apoptosis and endothelium-dependent vasodilation were observed. Key results "Silencing" of TXNIP by RNA interference inhibited NLRP3 inflammasome activation in response to ER stress, indicating that TXNIP was a key link between ER stress and NLRP3 inflammasome activation. AMPK activators salicylate and AICAR prevented ROS-induced mitochondrial fission by enhancing dynamin-related protein 1 (Drp1) phosphorylation (Ser 637) and thereby attenuated IRE-1α and PERK phosphorylation, but their actions were blocked by knockdown of AMPK. Salicylate and AICAR reduced TXNIP induction and inhibited NLRP3 inflammasome activation by reducing NLRP3 and caspase-1 expression, leading to a reduction in IL-1ß secretion. As a result, salicylate and AICAR inhibited inflammation and reduced cell apoptosis. Meanwhile, salicylate and AICAR enhanced eNOS phosphorylation and restored the loss of endothelium-dependent vasodilation in the rat aorta. Immunohistochemistry staining showed that AMPK activation inhibited ER stress and NLRP3 inflammasome activation in the vascular endothelium. CONCLUSION AND IMPLICATIONS: Pharmacological activation of AMPK regulated mitochondrial morphology and ameliorated endothelial dysfunction by suppression of mitochondrial ROS-associated ER stress and subsequent TXNIP/NLRP3 inflammasome activation. These findings suggested that regulation of Drp1 phosphorylation by AMPK activation contributed to suppression of ER stress and thus presented a potential therapeutic strategy for AMPK activation in the regulation of endothelium homeostasis.


Subject(s)
AMP-Activated Protein Kinases/biosynthesis , Carrier Proteins/biosynthesis , Dynamins/biosynthesis , Endoplasmic Reticulum Stress/drug effects , Inflammation/genetics , AMP-Activated Protein Kinases/genetics , Aminoimidazole Carboxamide/administration & dosage , Aminoimidazole Carboxamide/analogs & derivatives , Animals , Carrier Proteins/genetics , Caspase 1/biosynthesis , Cell Cycle Proteins , Dynamins/genetics , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Gene Expression Regulation/drug effects , Humans , Inflammation/drug therapy , Inflammation/pathology , Interleukin-1beta/metabolism , Mitochondrial Dynamics/drug effects , Rats , Ribonucleotides/administration & dosage , Salicylates/administration & dosage , Vasodilation/drug effects
19.
J Biol Chem ; 289(37): 25890-906, 2014 Sep 12.
Article in English | MEDLINE | ID: mdl-25008320

ABSTRACT

Myocardial ischemia and cardioprotection by ischemic pre-conditioning induce signal networks aimed at survival or cell death if the ischemic period is prolonged. These pathways are mediated by protein post-translational modifications that are hypothesized to cross-talk with and regulate each other. Phosphopeptides and lysine-acetylated peptides were quantified in isolated rat hearts subjected to ischemia or ischemic pre-conditioning, with and without splitomicin inhibition of lysine deacetylation. We show lysine acetylation (acetyl-Lys)-dependent activation of AMP-activated protein kinase, AKT, and PKA kinases during ischemia. Phosphorylation and acetyl-Lys sites mapped onto tertiary structures were proximal in >50% of proteins investigated, yet they were mutually exclusive in 50 ischemic pre-conditioning- and/or ischemia-associated peptides containing the KXXS basophilic protein kinase consensus motif. Modifications in this motif were modeled in the C terminus of muscle-type creatine kinase. Acetyl-Lys increased proximal dephosphorylation by 10-fold. Structural analysis of modified muscle-type creatine kinase peptide variants by two-dimensional NMR revealed stabilization via a lysine-phosphate salt bridge, which was disrupted by acetyl-Lys resulting in backbone flexibility and increased phosphatase accessibility.


Subject(s)
Lysine/metabolism , Myocardial Ischemia/genetics , Myocardial Ischemia/metabolism , Protein Processing, Post-Translational/genetics , AMP-Activated Protein Kinases/biosynthesis , Acetylation/drug effects , Amino Acid Motifs , Animals , Cardiotonic Agents/administration & dosage , Ischemic Preconditioning , Myocardial Ischemia/pathology , Naphthalenes/administration & dosage , Phosphorylation , Phosphotransferases/genetics , Phosphotransferases/metabolism , Proto-Oncogene Proteins c-akt/biosynthesis , Pyrones/administration & dosage , Rats , Signal Transduction/drug effects , Signal Transduction/genetics
20.
J Cell Physiol ; 230(1): 199-209, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24958127

ABSTRACT

Ghrelin is a physiological-active peptide with growth hormone-releasing activity, orexigenic activity, etc. In addition, the recent study has also suggested that ghrelin possesses the pathophysiological abilities related with type 2 diabetes. However, the ghrelin-direct-effects implicated in type 2 diabetes on peripheral tissues have been still unclear, whereas its actions on the central nervous system (CNS) appear to induce the development of diabetes. Thus, to assess its peripheral effects correlated with diabetes, we investigated the regulatory mechanisms about adipokines, which play a central role in inducing peripheral insulin resistance, secreted from mature 3T3-L1 adipocytes stimulated with ghrelin in vitro . The stimulation with 50 nmol/L ghrelin for 24 h resulted in the significant 1.9-fold increase on vascular endothelial growth factor-120 (VEGF(120)) releases (p < 0.01) and the 1.7-fold on monocyte chemoattractant protein-1 (MCP-1) (p < 0.01) from 3T3-L1 adipocytes, respectively, while ghrelin failed to enhance tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß), IL-6, IL-10 and adiponectin secretions. In addition, Akt phosphorylation on Ser473 and c-Jun NH2 -terminal protein kinase (JNK) phosphorylation on Thr183/Tyr185 were markedly enhanced 1.4-fold (p < 0.01) and 1.6-fold (p < 0.01) in the ghrelin-stimulated adipocytes, respectively. Furthermore, the treatment with LY294002 (50 µmol/L) and Wortmannin (10nmol/L), inhibitors of phosphatidylinositol 3-kinase (PI3K), significantly decreased the amplified VEGF(120) secretion by 29% (p < 0.01) and 28% (p < 0.01) relative to the cells stimulated by ghrelin alone, respectively, whereas these inhibitors had no effects on increased MCP-1 release. On the other hand, JNK inhibitor SP600125 (10 µmol/L) clearly reduced the increased MCP-1, but not VEGF(120), release by 35% relative to the only ghrelin-stimulated cells (p < 0.01). In conclusion, ghrelin can enhance the secretions of proinflammatory adipokines, VEGF(120) and MCP-1, but fails to affect IL-10 and adiponectin which are considered to be anti-inflammatory adipokines. Moreover, this augmented VEGF(120) release is invited through the activation of PI3K pathways and the MCP-1 is through JNK pathways. Consequently, our results strongly suggest that ghrelin can induce the development of diabetes via its direct-action in peripheral tissues as well as via in CNS.


Subject(s)
Adipocytes/drug effects , Chemokine CCL2/metabolism , Diabetes Mellitus, Type 2/metabolism , Ghrelin/pharmacology , Vascular Endothelial Growth Factor A/metabolism , 3T3 Cells , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/biosynthesis , Adipocytes/metabolism , Adiponectin/biosynthesis , Androstadienes/pharmacology , Animals , Anthracenes/pharmacology , Cell Line , Chromones/pharmacology , Endoplasmic Reticulum Stress/drug effects , Interleukin-10/biosynthesis , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Morpholines/pharmacology , Oxidative Stress/drug effects , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Wortmannin
SELECTION OF CITATIONS
SEARCH DETAIL