Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 269
1.
Int J Mol Sci ; 22(2)2021 Jan 19.
Article En | MEDLINE | ID: mdl-33478150

It has been increasingly recognized that SNAIL1 and SNAIL2, as major EMT-inducers, might also be involved in drug resistance of cancer cells. We sought to determine a relation between SNAIL1/2, E-cadherin and N-cadherin expression, as well as ovarian cancer cells' resistance to cisplatin and EMT markers' level. Thus, four ovarian cancer cell lines, were used: A2780, A2780cis, SK-OV-3 and OVCAR-3. We assessed the impact of ERK1/2, AKT and STAT3 proteins (chosen by the profiling activity of over 40 signaling proteins) on SNAIL1/2 expression, along with E-cadherin and N-cadherin levels. We showed that expression of SNAIL1 and N-cadherin are the highest in cisplatin-resistant A2780cis and SK-OV-3 cells, while high SNAIL2 and E-cadherin levels were observed in cisplatin-sensitive A2780 cells. The highest E-cadherin level was noticed in OVCAR-3 cells. SNAIL1/2 expression was dependent on ERK1/2 activity in cisplatin-resistant and potentially invasive SK-OV-3 and OVCAR-3 cells. STAT-3 regulates expression of SNAIL1/2 and leads to the so-called "cadherin switch" in cancer cells, independently of their chemoresistance. In conclusion, SNAIL1, but not SNAIL2, seems to be involved in ovarian cancer cells' cisplatin resistance. STAT3 is a universal factor determining the expression of SNAIL1/2 in ovarian cancer cells regardless of their chemoresitance or invasive capabilities.


Drug Resistance, Neoplasm/genetics , Epithelial-Mesenchymal Transition/genetics , Ovarian Neoplasms , Snail Family Transcription Factors/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Ovarian Epithelial/drug therapy , Carcinoma, Ovarian Epithelial/genetics , Carcinoma, Ovarian Epithelial/pathology , Cell Line, Tumor , Cisplatin/therapeutic use , Epithelial-Mesenchymal Transition/drug effects , Female , Gene Expression Regulation, Neoplastic , Humans , MAP Kinase Signaling System/physiology , Oncogene Protein v-akt/physiology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , STAT3 Transcription Factor/physiology , Signal Transduction/physiology , Snail Family Transcription Factors/metabolism
2.
J Neurosci ; 40(45): 8652-8668, 2020 11 04.
Article En | MEDLINE | ID: mdl-33060174

Inhibitory interneurons integrate into developing circuits in specific ratios and distributions. In the neocortex, inhibitory network formation occurs concurrently with the apoptotic elimination of a third of GABAergic interneurons. The cell surface molecules that select interneurons to survive or die are unknown. Here, we report that members of the clustered Protocadherins (cPCDHs) control GABAergic interneuron survival during developmentally-regulated cell death. Conditional deletion of the gene cluster encoding the γ-Protocadherins (Pcdhgs) from developing GABAergic neurons in mice of either sex causes a severe loss of inhibitory populations in multiple brain regions and results in neurologic deficits such as seizures. By focusing on the neocortex and the cerebellar cortex, we demonstrate that reductions of inhibitory interneurons result from elevated apoptosis during the critical postnatal period of programmed cell death (PCD). By contrast, cortical interneuron (cIN) populations are not affected by removal of Pcdhgs from pyramidal neurons or glial cells. Interneuron loss correlates with reduced AKT signaling in Pcdhg mutant interneurons, and is rescued by genetic blockade of the pro-apoptotic factor BAX. Together, these findings identify the PCDHGs as pro-survival transmembrane proteins that select inhibitory interneurons for survival and modulate the extent of PCD. We propose that the PCDHGs contribute to the formation of balanced inhibitory networks by controlling the size of GABAergic interneuron populations in the developing brain.SIGNIFICANCE STATEMENT A pivotal step for establishing appropriate excitatory-inhibitory ratios is adjustment of neuronal populations by cell death. In the mouse neocortex, a third of GABAergic interneurons are eliminated by BAX-dependent apoptosis during the first postnatal week. Interneuron cell death is modulated by neural activity and pro-survival pathways but the cell-surface molecules that select interneurons for survival or death are unknown. We demonstrate that members of the cadherin superfamily, the clustered γ-Protocadherins (PCDHGs), regulate the survival of inhibitory interneurons and the balance of cell death. Deletion of the Pcdhgs in mice causes inhibitory interneuron loss in the cortex and cerebellum, and leads to motor deficits and seizures. Our findings provide a molecular basis for controlling inhibitory interneuron population size during circuit formation.


Cadherins/physiology , Cell Death/physiology , Interneurons/physiology , gamma-Aminobutyric Acid/physiology , Animals , Apoptosis/genetics , Cadherin Related Proteins , Cadherins/genetics , Cerebral Cortex/cytology , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/growth & development , Electroencephalography , Female , Magnetic Resonance Imaging , Male , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Nerve Net/physiology , Nervous System Diseases/etiology , Oncogene Protein v-akt/genetics , Oncogene Protein v-akt/physiology , Seizures/etiology , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/physiology
3.
Int Urol Nephrol ; 52(9): 1801-1808, 2020 Sep.
Article En | MEDLINE | ID: mdl-32661617

PURPOSE: Chronic kidney disease (CKD) has become a global public health problem and accompanied by renal fibrosis. MiR-194, a tumor suppressor gene, has been previously reported to be associated with the pathogenesis of tissue fibrosis. However, the role of miR-194 in the pathogenesis of renal fibrosis remains unknown. METHODS: A renal fibrosis model was constructed by unilateral ureteral obstruction (UUO) in male C57BL/6 mice. HE and MASSON stainings were used for histological analysis. The expression level of miR-194 was detected by RT-qPCR. The protein expression was detected by western blotting. The levels of inflammatory cytokines were detected by ELISA. The relationship between miR-194 and Runx1 was further verified by dual luciferase reporter assay. RESULTS: The results showed that miR-194 level was downregulated in kidney tissue of UUO mice, accompanied by significantly pathological damage and renal fibrosis. MiR-194 mimics significantly reduced pathological damage and alleviated renal fibrosis that caused by UOO, and inhibited the expression levels of α-SMA and collagen I. In addition, miR-194 mimics also reduced the expression level of serum inflammatory factors. Moreover, in vitro analysis indicated that Runx1 was a downstream target gene of miR-194. Furthermore, mechanism analysis indicated that miR-194 reduced mouse renal fibrosis by inhibiting the Runx1/AKT pathway in vivo and in vitro. CONCLUSION: The present findings suggested that miR-194 targets Runx1/Akt pathway to reduce renal fibrosis in UOO-induced mice. This study provides a novel strategy for the prevention and treatment of renal fibrosis.


Core Binding Factor Alpha 2 Subunit/physiology , Kidney/pathology , MicroRNAs/physiology , Oncogene Protein v-akt/physiology , Ureteral Obstruction/etiology , Animals , Fibrosis/etiology , Male , Mice , Mice, Inbred C57BL , Signal Transduction
4.
Cell Immunol ; 315: 56-63, 2017 05.
Article En | MEDLINE | ID: mdl-28400057

Recent studies have indicated that Thymic stromal lymphopoietin (TSLP) plays an important role in the prevention and treatment of asthma. However the role of TSLP in dysfunction of airway epithelial adherens junctions E-cadherin in house dust mite (HDM)-induced asthma has not been addressed. We hypothesized that TSLP contributed to HDM-induced E-cadherin dysfunction in asthmatic BALB/c mice and 16HBE cells. In vivo, a HDM-induced asthma mouse model was set up for 8weeks. Mice inhaled an anti-TSLP monoclonal antibody (mAb) before HDM. The mice treated with the anti-TSLP mAb ameliorated airway inflammation, the decreasing and aberrant distribution of E-cadherin and ß-catenin as well as phosphorylation(p)-AKT induced by HDM. In vitro, HDM increased the expression of TSLP and E-cadherin dysfunction by PI3K/Akt signaling pathway. The exposure of 16HBE to TSLP resulted in redistribution of E-cadherin. These results indicate that TSLP may be an important contributor in E-cadherin dysfunction of HDM-induced asthma. TSLP signaling blocking shows a protective effect in mice and that the PI3K/Akt pathway may play a role in this process.


Antibodies, Monoclonal/therapeutic use , Asthma/immunology , Cadherins/metabolism , Cytokines/physiology , Pyroglyphidae/immunology , Administration, Inhalation , Animals , Antibodies, Monoclonal/administration & dosage , Asthma/therapy , Bronchi/cytology , Bronchial Hyperreactivity/etiology , Bronchial Hyperreactivity/immunology , Bronchial Hyperreactivity/prevention & control , Cell Line , Chromones/pharmacology , Cytokines/antagonists & inhibitors , Cytokines/biosynthesis , Cytokines/immunology , Disease Models, Animal , Epithelial Cells , Humans , Lung/pathology , Mice , Mice, Inbred BALB C , Morpholines/pharmacology , Oncogene Protein v-akt/physiology , Phosphatidylinositol 3-Kinases/physiology , Phosphorylation , Protein Processing, Post-Translational , Random Allocation , Signal Transduction/immunology , Specific Pathogen-Free Organisms , beta Catenin/analysis , Thymic Stromal Lymphopoietin
5.
PLoS One ; 12(2): e0171503, 2017.
Article En | MEDLINE | ID: mdl-28152035

Nik-related kinase (Nrk) is a Ser/Thr kinase and was initially discovered as a molecule that was predominantly detected in skeletal muscles during development. A recent study using Nrk-null mice suggested the importance of Nrk in proper placental development; however, the molecular mechanism remains unknown. In this study, we demonstrated that differentiated trophoblasts from murine embryonic stem cells (ESCs) endogenously expressed Nrk and that Nrk disruption led to the enhanced proliferation of differentiated trophoblasts. This phenomenon may reflect the overproliferation of trophoblasts that has been reported in enlarged placentas of Nrk-null mice. Furthermore, we demonstrated that AKT phosphorylation at Ser473 was upregulated in Nrk-null trophoblasts and that inhibition of AKT phosphorylation cancelled the enhanced proliferation observed in differentiated Nrk-null trophoblasts. These results indicated that the upregulation of AKT phosphorylation was the possible cause of enhanced proliferation observed in Nrk-null trophoblasts. The upregulation of AKT phosphorylation was also confirmed in enlarged Nrk-null placentas in vivo, suggesting that proper regulation of AKT by Nrk was important for normal placental development. In addition, our detailed analysis on phosphorylation status of AKT isoforms in newly established trophoblast stem cells (TSCs) revealed that different levels of upregulation of AKT phosphorylation were occurred in Nrk-null TSCs depending on AKT isoforms. These results further support the importance of Nrk in proper development of trophoblast lineage cells and indicate the possible application of TSCs for the analysis of differently regulated activation mechanisms of AKT isoforms.


Intracellular Signaling Peptides and Proteins/physiology , Oncogene Protein v-akt/physiology , Placentation/physiology , Protein Serine-Threonine Kinases/physiology , Trophoblasts/physiology , Animals , Blotting, Western , Cell Differentiation/physiology , Cell Proliferation/physiology , Embryonic Development/physiology , Embryonic Stem Cells/physiology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Knockout , Oncogene Protein v-akt/metabolism , Phosphorylation , Placenta/physiology , Polymerase Chain Reaction , Pregnancy , Up-Regulation
6.
PLoS One ; 12(1): e0170624, 2017.
Article En | MEDLINE | ID: mdl-28107461

Fatty acid synthase (FASN) is an enzyme responsible for the de novo synthesis of long-chain fatty acids. During oncogenesis, FASN plays a role in growth and survival rather than acting within the energy storage pathways. Here, the function of FASN during early embryonic development was studied using its specific inhibitor, C75. We found that the presence of the inhibitor reduced blastocyst hatching. FASN inhibition decreased Cpt1 expression, leading to a reduction in mitochondria numbers and ATP content. This inhibition of FASN resulted in the down-regulation of the AKT pathway, thereby triggering apoptosis through the activation of the p53 pathway. Activation of the apoptotic pathway also leads to increased accumulation of reactive oxygen species and autophagy. In addition, the FASN inhibitor impaired cell proliferation, a parameter of blastocyst quality for outgrowth. The level of OCT4, an important factor in embryonic development, decreased after treatment with the FASN inhibitor. These results show that FASN exerts an effect on early embryonic development by regulating both fatty acid oxidation and the AKT pathway in pigs.


Blastocyst/physiology , Fatty Acid Synthases/antagonists & inhibitors , Oncogene Protein v-akt/physiology , Signal Transduction/physiology , Adenosine Triphosphate/metabolism , Animals , Cell Proliferation/drug effects , Cell Proliferation/physiology , Down-Regulation , Fatty Acid Synthases/metabolism , Fatty Acid Synthases/physiology , Fluorescent Antibody Technique , MicroRNAs/metabolism , Microscopy, Confocal , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Signal Transduction/drug effects , Swine/embryology
7.
PLoS One ; 12(1): e0170620, 2017.
Article En | MEDLINE | ID: mdl-28107526

Rab14 is a member of RAS oncogene family, and its dysfunction has been reported to be involved in various types of human cancer. However, its expression and function were still unclear in gastric cancer. The aim of this study was to investigate the function and mechanism of Rab14 in gastric cancer cell lines. Quantitative real-time PCR (qRT-PCR) was performed in 17 gastric adenocarcinoma tissues and 4 cell lines to detect the expression of Rab14. 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyl-tetrazolium bromide (MTT), colony formation and flow cytometry assays were employed to determine the proliferative ability, cell cycle transition and apoptosis in vitro in BGC-823 or SGC-7901 cells. Western blot was performed to investigate the pathways and mechanism of Rab14 regulation. In this study, we show that Rab14 presents a significant up-regulated expression among the paired tissue samples and cell lines in gastric cancer. When we overexpressed Rab14 in SGC-7901 cells or silenced Rab14 in BGC-823 cells, we found that Rab14 could modify cell growth, cell cycle or apoptosis, which accompanied with an obvious regulation of CCND1, CDK2 and BAX involving in AKT signaling pathway. In conclusion, this study provides a new evidence on that Rab14 functions as a novel tumor oncogene and could be a potential therapeutic target in gastric cancer.


Adenocarcinoma/physiopathology , Cell Proliferation/physiology , Oncogene Protein v-akt/physiology , Oncogenes/physiology , Signal Transduction/physiology , Stomach Neoplasms/physiopathology , rab GTP-Binding Proteins/physiology , Apoptosis/physiology , Blotting, Western , Cell Cycle/physiology , Cell Line, Tumor , Flow Cytometry , Gene Expression Regulation, Neoplastic/physiology , Humans , Real-Time Polymerase Chain Reaction , Up-Regulation
8.
Am J Physiol Heart Circ Physiol ; 312(3): H478-H484, 2017 Mar 01.
Article En | MEDLINE | ID: mdl-28039203

Ischemic preconditioning (IPC), i.e., brief episodes of nonlethal myocardial ischemia-reperfusion (I/R) before sustained ischemia with subsequent reperfusion, reduces infarct size in all species tested so far, including humans. In rodents, the cardioprotective signal transduction causally involves an activation of Akt, ERK1/2, and STAT3. However, there are apparent species differences in the signal transduction between rodents and larger mammals such as pigs, where data on IPC's signal transduction are inconsistent for Akt and ERK1/2. The role of STAT3 has not yet been analyzed. Pigs were subjected to 60 min of left anterior descending coronary artery occlusion and 180 min of reperfusion without or with IPC (2 cycles of 3-min occlusion separated by 2 min of reperfusion 15 min before sustained I/R). Infarct size was analyzed by triphenyl tetrazolium chloride staining, and Akt, ERK1/2, and STAT3 phosphorylation was quantified in myocardial biopsies taken at baseline and early reperfusion. AG490 was used to block the STAT3 signaling pathway. IPC reduced infarct size (%area at risk; mean ± SE, I/R, 45 ± 3 vs. IPC, 18 ± 3, P < 0.05). Akt and ERK1/2 phosphorylation was increased at early reperfusion without and with IPC. In contrast, STAT3 phosphorylation at early reperfusion was only increased with IPC (%baseline; mean ± SE, I/R, 126 ± 29 vs. IPC, 408 ± 147, P < 0.05). AG490 prevented the IPC-related increase of STAT3 phosphorylation at reperfusion (%baseline; mean ± SE, 82 ± 12) and abolished IPC's cardioprotection (%area at risk; mean ± SE, 35 ± 4). In pigs, increased phosphorylation of STAT3 is causally involved, whereas Akt and ERK1/2 seem to play no role in IPC's cardioprotection.NEW & NOTEWORTHY In pig hearts in situ, ischemic preconditioning (IPC) causally involves increased phosphorylation of STAT3, whereas Akt and ERK1/2 play no role for cardioprotection. The cardioprotective signal transduction of IPC is similar to that of ischemic postconditioning and remote IPC in pigs.


Ischemic Preconditioning, Myocardial , STAT3 Transcription Factor/physiology , Animals , Body Temperature , Hemodynamics , Janus Kinase 2/antagonists & inhibitors , MAP Kinase Signaling System/drug effects , Male , Myocardial Infarction/pathology , Myocardial Infarction/prevention & control , Oncogene Protein v-akt/drug effects , Oncogene Protein v-akt/physiology , Phosphorylation , STAT3 Transcription Factor/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/physiology , Sus scrofa , Swine , Swine, Miniature , Tyrphostins/pharmacology
9.
J Occup Health ; 59(1): 8-16, 2017 Jan 24.
Article En | MEDLINE | ID: mdl-27885243

OBJECTIVE: To explore the mechanisms of hypospadias induced by in utero exposure to din-butyl phthalate (DBP). METHODS: Timed-pregnant Sprague-Dawley rats were administered 750 mg/kg of DBP by gavage from GD (gestation days) 13 to GD 18, whereas control group received corn oil. Genital tubercles (GTs) and blood samples were collected from male fetuses on GD 19. The serum testosterone concentration, apoptosis activity, autophagosomes and their related proteins (light chain 3 (LC3-I, LC3-II) ), and sequestosomes (SQSTM1/p62) in the GTs were then measured. Protein expression of protein kinase B (Akt), Beclin 1, phosphorylated Akt (p-Akt), p-S6, and phosphorylated mammalian target of rapamycin (p-mTOR) in the GTs were analyzed by Western blotting. RESULTS: The incidence of hypospadias induced by DBP was 43.64% in male fetuses. The GT volume and GT volume/body weight of fetuses were significantly reduced in the hypospadias and the non-hypospadias groups. Apoptotic cell number was significantly decreased in the GTs of the hypospadias group, but unchanged in the non-hyposadias group. The ratio of LC3-II/LC3-I was higher in the GTs from DBP exposed fetuses compared to the control group. The ratio of LC3-II/LC3-I in the GTs was higher in the hypospadias group than in the non-hypospadias group. The number of autophagosomes was increased in the GTs of the hypospadias group. Protein expression of p-S6, p-mTOR, and p-Akt were significantly decreased in the GTs of hypospadiac rats. CONCLUSIONS: DBP-induced hypospadias might be associated with apoptosis and autophagy mediated by the PI3K/Akt/mTOR signaling pathway in the GT.


Autophagy/drug effects , Carrier Proteins , Dibutyl Phthalate/toxicity , Hypospadias/chemically induced , Maternal Exposure/adverse effects , Plasticizers/toxicity , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Female , Fetus/drug effects , Male , Oncogene Protein v-akt/physiology , Penis/drug effects , Penis/embryology , Pregnancy , Rats , Rats, Sprague-Dawley , TOR Serine-Threonine Kinases/physiology , Testosterone/blood
10.
J Neurosci ; 37(5): 1176-1186, 2017 02 01.
Article En | MEDLINE | ID: mdl-28007761

The neuropeptide galanin has been shown to interact with the opioid system. More specifically, galanin counteracts the behavioral effects of the systemic administration of µ-opioid receptor (MOR) agonists. Yet the mechanism responsible for this galanin-opioid interaction has remained elusive. Using biophysical techniques in mammalian transfected cells, we found evidence for selective heteromerization of MOR and the galanin receptor subtype Gal1 (Gal1R). Also in transfected cells, a synthetic peptide selectively disrupted MOR-Gal1R heteromerization as well as specific interactions between MOR and Gal1R ligands: a negative cross talk, by which galanin counteracted MAPK activation induced by the endogenous MOR agonist endomorphin-1, and a cross-antagonism, by which a MOR antagonist counteracted MAPK activation induced by galanin. These specific interactions, which represented biochemical properties of the MOR-Gal1R heteromer, could then be identified in situ in slices of rat ventral tegmental area (VTA) with MAPK activation and two additional cell signaling pathways, AKT and CREB phosphorylation. Furthermore, in vivo microdialysis experiments showed that the disruptive peptide selectively counteracted the ability of galanin to block the dendritic dopamine release in the rat VTA induced by local infusion of endomorphin-1, demonstrating a key role of MOR-Gal1R heteromers localized in the VTA in the direct control of dopamine cell function and their ability to mediate antagonistic interactions between MOR and Gal1R ligands. The results also indicate that MOR-Gal1R heteromers should be viewed as targets for the treatment of opioid use disorders. SIGNIFICANCE STATEMENT: The µ-opioid receptor (MOR) localized in the ventral tegmental area (VTA) plays a key role in the reinforcing and addictive properties of opioids. With parallel in vitro experiments in mammalian transfected cells and in situ and in vivo experiments in rat VTA, we demonstrate that a significant population of these MORs form functional heteromers with the galanin receptor subtype Gal1 (Gal1R), which modulate the activity of the VTA dopaminergic neurons. The MOR-Gal1R heteromer can explain previous results showing antagonistic galanin-opioid interactions and offers a new therapeutic target for the treatment of opioid use disorder.


Receptors, Galanin/metabolism , Receptors, Opioid, mu/metabolism , Ventral Tegmental Area/metabolism , Animals , Cells, Cultured , Cyclic AMP Response Element-Binding Protein , Dopaminergic Neurons/drug effects , Galanin/pharmacology , HEK293 Cells , Humans , Ligands , Mitogen-Activated Protein Kinases/metabolism , Oncogene Protein v-akt/physiology , Phosphorylation , Rats , Receptor Cross-Talk , Receptor, Galanin, Type 1/genetics , Receptor, Galanin, Type 1/metabolism , Receptor, Galanin, Type 2/genetics , Receptor, Galanin, Type 2/metabolism , Receptors, Galanin/genetics , Receptors, Opioid, mu/genetics , Signal Transduction/genetics , Signal Transduction/physiology , Transfection
11.
J Neurosci ; 36(16): 4506-21, 2016 Apr 20.
Article En | MEDLINE | ID: mdl-27098694

The signaling pathways that regulate myelination in the PNS remain poorly understood. Phosphatidylinositol-4,5-bisphosphate 3-kinase 1A, activated in Schwann cells by neuregulin and the extracellular matrix, has an essential role in the early events of myelination. Akt/PKB, a key effector of phosphatidylinositol-4,5-bisphosphate 3-kinase 1A, was previously implicated in CNS, but not PNS myelination. Here we demonstrate that Akt plays a crucial role in axon ensheathment and in the regulation of myelin sheath thickness in the PNS. Pharmacological inhibition of Akt in DRG neuron-Schwann cell cocultures dramatically decreased MBP and P0 levels and myelin sheath formation without affecting expression of Krox20/Egr2, a key transcriptional regulator of myelination. Conversely, expression of an activated form of Akt in purified Schwann cells increased expression of myelin proteins, but not Krox20/Egr2, and the levels of activated Rac1. Transgenic mice expressing a membrane-targeted, activated form of Akt under control of the 2',3'-cyclic nucleotide 3'-phosphodiesterase promoter, exhibited thicker PNS and CNS myelin sheaths, and PNS myelin abnormalities, such as tomacula and myelin infoldings/outfoldings, centered around the paranodes and Schmidt Lanterman incisures. These effects were corrected by rapamycin treatmentin vivo Importantly, Akt activity in the transgenic mice did not induce myelination of nonmyelinating Schwann cells in the sympathetic trunk or Remak fibers of the dorsal roots, although, in those structures, they wrapped membranes redundantly around axons. Together, our data indicate that Akt is crucial for PNS myelination driving axonal wrapping by unmyelinated and myelinated Schwann cells and enhancing myelin protein synthesis in myelinating Schwann cells. SIGNIFICANCE STATEMENT: Although the role of the key serine/threonine kinase Akt in promoting CNS myelination has been demonstrated, its role in the PNS has not been established and remains uncertain. This work reveals that Akt controls several key steps of the PNS myelination. First, its activity promotes membrane production and axonal wrapping independent of a transcriptional effect. In myelinated axons, it also enhances myelin thickness through the mTOR pathway. Finally, sustained Akt activation in Schwann cells leads to hypermyelination/dysmyelination, mimicking some features present in neuropathies, such as hereditary neuropathy with liability to pressure palsies or demyelinating forms of Charcot-Marie-Tooth disease. Together, these data demonstrate the role of Akt in regulatory mechanisms underlying axonal wrapping and myelination in the PNS.


Axons/physiology , Myelin Sheath/physiology , Oncogene Protein v-akt/physiology , Sciatic Nerve/physiology , Animals , Axons/ultrastructure , Cells, Cultured , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myelin Sheath/ultrastructure , Peripheral Nerves/physiology , Peripheral Nerves/ultrastructure , Peripheral Nervous System/physiology , Peripheral Nervous System/ultrastructure , Sciatic Nerve/ultrastructure
12.
Alcohol Alcohol ; 51(6): 647-654, 2016 Nov.
Article En | MEDLINE | ID: mdl-26946194

AIMS: Previously we have demonstrated altered microglia P2X4R expression in response to alcohol and pharmacological blockade with a selective P2X4R antagonist can reverse the action, suggesting that P2X4R play a role in mediating alcohol-induced effects on microglia. In the present study, we investigated the underlying signaling mediators, which may play a role in modulating P2X4R expression in microglia cells in response to alcohol. METHODS: Embryonic stem cell-derived microglia (ESdM) cells were used to investigate the potential mechanisms involved in the regulation of P2X4R in response to alcohol. Selective P2X4R antagonist and kinase inhibitors were used to further corroborate the signal transduction pathway through which alcohol modulates P2X4R expression in microglia. RESULTS: Alcohol (100 mM) suppressed phosphorylated AKT and ERK cascades in native ESdM cells. This alcohol-induced suppression was confirmed to be P2X4R-dependent through the use of a selective P2X4R antagonist and knockdown of P2XR4 by siRNA. Alcohol increased transcriptional activity of CREB. P2X4R antagonist blocked alcohol-induced effects on CREB, suggesting a P2X4R-mediated effect. CONCLUSION: These findings provide important clues to the underlying mechanism of purinoceptors in alcohol-induced microglia immune suppression.


Cyclic AMP Response Element-Binding Protein/physiology , Ethanol/pharmacology , Gene Expression Regulation/drug effects , MAP Kinase Signaling System/physiology , Microglia/drug effects , Oncogene Protein v-akt/physiology , Receptors, Purinergic P2X4/metabolism , Blotting, Western , Brain-Derived Neurotrophic Factor/physiology , Gene Expression Regulation/physiology , Gene Knockdown Techniques , Human Embryonic Stem Cells , Humans , Microglia/physiology , Real-Time Polymerase Chain Reaction , Receptors, Purinergic P2X4/physiology , Signal Transduction/drug effects , Signal Transduction/physiology
13.
Zhonghua Nan Ke Xue ; 22(11): 1016-1020, 2016 Nov.
Article Zh | MEDLINE | ID: mdl-29281211

Male infertility is closely associated with spermatogenesis disorders triggered by aberrant gene expression or abnormal signaling pathways in the testis. The mammalian target of rapamycin (mTOR) is a central regulator of cell metabolism, playing an important role in regulating cell proliferation, differentiation, translation, actin polymerization, cycle progression, energy metabolism, autophagy, and other cellular activities. PI3K-Akt and LKB1-AMPK, the two well-defined classic signal transduction pathways, regulate the expressions of mTOR and its downstream p70S6K/4EBP1 through different molecular pathways. Recent studies show that mTOR-p70S6K/4EBP1 signaling participates in the regulation of the proliferation and differentiation of testicular cells and spermatogenesis. This review focuses on the role of PI3K-Akt/LKB1- AMPK-mTOR signaling cascades in testis development and spermatogenesis, providing some new perspectives for the studies of the molecular mechanism underlying male sterility.


Signal Transduction , Spermatogenesis , Testis/embryology , Adaptor Proteins, Signal Transducing/physiology , Adenylate Kinase/physiology , Animals , Autophagy , Cell Cycle Proteins , Cell Proliferation , Humans , Male , Oncogene Protein v-akt/physiology , Phosphatidylinositol 3-Kinases/physiology , Phosphoproteins/physiology , Ribosomal Protein S6 Kinases, 70-kDa/physiology , TOR Serine-Threonine Kinases/physiology
14.
J Physiol Sci ; 66(3): 229-39, 2016 May.
Article En | MEDLINE | ID: mdl-26541157

Sulfur dioxide (SO2) is naturally synthesized by glutamate-oxaloacetate transaminase (GOT) from L-cysteine in mammalian cells. We found that SO2 may have a protective effect on acute lung injury (ALI) induced by limb ischemia/reperfusion (I/R) in rats. The PI3K/Akt, p38MAPK, and JAK2/STAT3 pathways are crucial in cell signaling transduction. The present study aims to verify the role of SO2 on limb I/R-induced ALI, and investigate whether PI3K/Akt, p38MAPK, and JAK2/STAT3 pathways were involved, as well as the relationship among the three pathways; we used specific inhibitors (LY294002, SB03580, and Stattic) to block them, respectively. The experimental methods of Western, ELISA, TUNEL, etc., were used to test the results. In the I/R group, the parameters of lung injury (MDA, MPO, TUNEL, cytokines) increased significantly, but the administration of Na2SO3/NaHSO3 attenuated the damage in the lung. The Western results showed that the rat's lung exist expression of P-STAT3, P-AKT, and P-p38 proteins. After I/R, P-STAT3, P-Akt, and P-p38 proteins expression all increased. After using Na2SO3/NaHSO3, P-Akt, and P-p38 proteins expression increased, but P-STAT3 protein expression decreased. We also found a strange phenomenon; compared to the I/R + SO2 group, the administration of stattic, P-p38 protein expression showed no change, but P-Akt protein expression increased (p < 0.05). In conclusion, SO2 has a protective effect on rats with limb I/R-induced ALI. The JAK2/STAT3, PI3K/Akt, and p38MAPK pathways are likely all involved in the process, and the JAK2/STAT3 pathway may have an impact on the P13K/Akt pathway.


Acute Lung Injury/etiology , Extremities/blood supply , Ischemia/complications , Janus Kinase 2/physiology , MAP Kinase Signaling System/physiology , Oncogene Protein v-akt/physiology , Phosphatidylinositol 3-Kinases/physiology , Reperfusion Injury/complications , STAT3 Transcription Factor/physiology , Signal Transduction/physiology , Sulfur Dioxide/metabolism , Acute Lung Injury/physiopathology , Animals , Interleukins/physiology , Ischemia/physiopathology , Male , Rats , Rats, Sprague-Dawley , Reperfusion Injury/physiopathology
15.
PLoS One ; 10(11): e0141082, 2015.
Article En | MEDLINE | ID: mdl-26539823

BACKGROUND: GPBAR1 is a bile acids activated receptor expressed in entero-hepatic tissues. In the liver expression of GPBAR1 is restricted to sinusoidal and Kuppfer cells. In the systemic circulation vasodilation caused by GPBAR1 agonists is abrogated by inhibition of cystathione-γ-liase (CSE), an enzyme essential to the generation of hydrogen sulfide (H2S), a vasodilatory agent. Portal BAR501 is a semisynthetic bile acid derivative endowed with a potent and selective agonistic activity toward GPBAR1. METHODS: Cirrhosis was induced in mice by carbon tetrachloride (CCL4) administration for 9 weeks. Liver endothelial dysfunction was induced by feeding wild type and Gpbar1-/- mice with methionine for 4 weeks. In both models, mice were administered BAR501, 15 mg/kg/day. RESULTS: By transactivation assay we demonstrate that BAR501 is a selective GPBAR1 agonist devoid of any FXR agonistic activity. In naïve rats, BAR501 effectively reduced hepatic perfusion pressure and counteracted the vasoconstriction activity of norepinephrine. In the CCl4 model, 9 weeks treatment with BAR501 effectively protected against development of endothelial dysfunction by increasing liver CSE expression and activity and by reducing endothelin (ET)-1 gene expression. In mice feed methionine, treatment with BAR501 attenuated endothelial dysfunction and caused a GPBAR1-dependent regulation of CSE. Using human liver sinusoidal cells, we found that modulation of CSE expression/activity is mediated by both genomic (recruitment of CREB to CRE in the CSE promoter) and non-genomic effects, involving a Akt-dependent phosporylation of CSE and endothelial nitric oxide (NO) synthase (eNOS). BAR501, phosphorylates FOXO1 and inhibits ET-1 transcription in liver sinusoidal cells. CONCLUSIONS: BAR501, a UDCA-like GPBAR1 agonist, rescues from endothelial dysfunction in rodent models of portal hypertension by exerting genomic and non-genomic effects on CSE, eNOS and ET-1 in liver sinusoidal cells.


Endothelin-1/physiology , Endothelium, Vascular/drug effects , Hydrogen Sulfide/metabolism , Hypertension, Portal/physiopathology , Receptors, G-Protein-Coupled/agonists , Animals , Bile Acids and Salts/metabolism , Cholestanols/pharmacology , Endothelium, Vascular/physiopathology , Forkhead Box Protein O1 , Forkhead Transcription Factors/physiology , Humans , Hypertension, Portal/drug therapy , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Oncogene Protein v-akt/physiology , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction , Receptors, G-Protein-Coupled/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , Ursodeoxycholic Acid/analogs & derivatives
16.
Hepatology ; 62(6): 1791-803, 2015 Dec.
Article En | MEDLINE | ID: mdl-26257239

UNLABELLED: Sorafenib is a specific adenosine triphosphate-competitive RAF inhibitor used as a first-line treatment of advanced hepatocellular carcinoma (HCC). However, the responses are variable, reflecting heterogeneity of the disease, while the resistance mechanism remains poorly understood. Here, we report that sorafenib treatment can exacerbate disease progression in both patient-derived xenografts and cell line-derived xenografts and that the therapeutic effect of the drug inversely covaries to the ratio of epithelial cell adhesion molecule-positive cells, which may be tumor initiating cells in HCC. The TSC2-AKT cascade mediates this sorafenib resistance. In response to sorafenib treatment, formation of the TSC1/2 complex is enhanced, causing increased phosphorylation of AKT, which contributes to up-regulation of "stemness"-related genes in epithelial cell adhesion molecule-positive cells and enhancement of tumorigenicity. The expression of TSC2 negatively correlated with prognosis in clinical sorafenib therapy. Furthermore, all-trans retinoic acid decreased AKT activity, reduced the epithelial cell adhesion molecule-positive cell population enriched by sorafenib, and potentiated the therapeutic effect of sorafenib in the patient-derived xenograft model. CONCLUSION: Our findings suggest that a subtype of HCC is not suitable for sorafenib therapy; this resistance to sorafenib can be predicted by the status of TSC2, and agents inducing differentiation of tumor initiating cells (e.g., all-trans retinoic acid) should improve the prognosis of this subtype of HCC.


Antigens, Neoplasm/drug effects , Antineoplastic Agents/adverse effects , Carcinoma, Hepatocellular/chemically induced , Cell Adhesion Molecules/drug effects , Liver Neoplasms/chemically induced , Neoplastic Stem Cells/drug effects , Niacinamide/analogs & derivatives , Oncogene Protein v-akt/physiology , Phenylurea Compounds/adverse effects , Tumor Suppressor Proteins/physiology , Animals , Carcinoma, Hepatocellular/classification , Disease Progression , Epithelial Cell Adhesion Molecule , Humans , Liver Neoplasms/classification , Mice , Niacinamide/adverse effects , Sorafenib , Tuberous Sclerosis Complex 2 Protein
17.
Arch Pharm Res ; 38(12): 2153-62, 2015 Dec.
Article En | MEDLINE | ID: mdl-26119076

It has been reported that alkaloids derived from Coptis chinensis exert anti-adipogenic activity on 3T3-L1 adipocytes by downregulating peroxisome proliferation-activity receptor-γ (PPAR-γ) and CCAAT/enhancer binding protein-α (C/EBP-α). However, the signaling-based mechanism of the inhibitory role of epiberberine in the early stages of 3T3-L1 adipocyte differentiation is uncharacterized. Here, we show that epiberberine had inhibitory effects on adipocyte differentiation and significantly decreased lipid accumulation by downregulating an adipocyte-specific transcription factor, sterol regulatory element-binding protein-1 (SREBP-1). Furthermore, we observed that epiberberine markedly suppressed the differentiation-mediated phosphorylation of components of both the Raf/mitogen-activated protein kinase 1 (MEK1)/extracellular signal-regulated protein kinase 1/2 (ERK1/2) and AMP-activated protein kinase-α1 (AMPKα)/Akt pathways. In addition, gene expression of fatty acid synthase (FAS) was significantly inhibited by treatment with epiberberine during adipogenesis. These results indicate that the anti-adipogenic mechanism of epiberberine is associated with inhibition of phosphorylation of Raf/MEK1/ERK1/2 and AMPKα/Akt, followed by downregulation of the major transcription factors of adipogenesis, such as PPAR-γ, C/EBP-α, and SREBP-1, and FAS. Taken together, this study suggests that the anti-adipogenic effect of epiberberine is mediated by downregulation of the Raf/MEK1/ERK1/2 and AMPKα/Akt pathways during 3T3-L1 adipocyte differentiation. Moreover, the anti-adipogenic effects of epiberberine were not accompanied by modulation of ß-catenin.


AMP-Activated Protein Kinases/physiology , Adipogenesis/physiology , Berberine/analogs & derivatives , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinases/physiology , Oncogene Protein v-akt/physiology , raf Kinases/physiology , 3T3-L1 Cells , AMP-Activated Protein Kinases/antagonists & inhibitors , Adipogenesis/drug effects , Animals , Anti-Obesity Agents/pharmacology , Berberine/pharmacology , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Drugs, Chinese Herbal/pharmacology , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 1/physiology , MAP Kinase Kinase 2/antagonists & inhibitors , MAP Kinase Kinase 2/physiology , MAP Kinase Signaling System/drug effects , Mice , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Oncogene Protein v-akt/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/physiology , raf Kinases/antagonists & inhibitors
18.
J Clin Endocrinol Metab ; 100(8): E1046-55, 2015 Aug.
Article En | MEDLINE | ID: mdl-26066673

CONTEXT: IGF-2 is highly expressed in the granulosa cells of human dominant ovarian follicles; however, little is known about the regulation of the IGF-2 gene or the interaction of IGF-2 and FSH during follicle development. OBJECTIVE: To examine the mechanisms involved in the regulation of the IGF-2 gene by FSH and the interplay between FSH and IGF-2 during granulosa cell differentiation. Design, Setting, Patients, and Interventions: Cumulus granulosa cells were separated from cumulus-oocyte complexes isolated from the follicular aspirates of in vitro fertilization patients and cultured for in vitro studies. MAIN OUTCOME: Protein and mRNA levels of IGF-2 and CYP19A1 (aromatase) were quantified using Western blot and quantitative real-time PCR. IGF-2 promoter-specific activation was determined by the amplification of alternative exons by PCR. Cell proliferation was assessed after treatment with FSH and/or IGF-2. RESULTS: FSH significantly enhanced IGF-2 expression after 8 hours of treatment and at low doses (1 ng/mL). Reciprocally, IGF-2 synergized with FSH to increase cell proliferation and the expression of CYP19A1. When IGF-2 activity was blocked, FSH was no longer able to stimulate CYP19A1 expression. Determination of IGF-2 promoter usage in human cumulus cells showed that the IGF-2 gene is driven by promoters P3 and P4. However, FSH exclusively increased P3 promoter-derived transcripts. Moreover, the FSH-induced stimulation of P3-driven IGF-2 transcripts was blocked by cotreatment with inhibitors of AKT or IGF-1 receptor (IGF-1R). The inhibitory effect of the IGF-1R inhibitor on FSH-induced IGF-2 mRNA accumulation was reversed by overexpression of a constitutively active AKT construct. CONCLUSIONS: FSH is a potent enhancer of IGF-2 expression in human granulosa cells. In return, IGF-2 activation of the IGF-1R and AKT is required for FSH to stimulate CYP19A1 expression and proliferation of granulosa cells. These findings suggest a positive loop interaction between FSH and IGF-2 that is critical for human granulosa cell proliferation and differentiation.


Follicle Stimulating Hormone/pharmacology , Granulosa Cells/drug effects , Insulin-Like Growth Factor II/genetics , Oncogene Protein v-akt/physiology , Aromatase/genetics , Aromatase/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cells, Cultured , Cumulus Cells/drug effects , Cumulus Cells/metabolism , Female , Gene Expression Regulation/drug effects , Granulosa Cells/metabolism , Humans , Insulin-Like Growth Factor II/metabolism , RNA, Messenger/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics
19.
Am J Surg Pathol ; 39(7): 902-11, 2015 Jul.
Article En | MEDLINE | ID: mdl-25970684

B-cell receptor (BCR) signaling is crucial for the survival of normal and neoplastic B cells, and inhibitors targeting BCR signaling pathways have shown promising therapeutic outcomes for patients with B-cell lymphomas. In the current study, we analyzed de novo diffuse large B-cell lymphoma without BCR expression (DLBCL, BCR) in 25 cases to determine the BCR/phosphatidylinositol-3-kinase/AKT (BCR/PI3K/AKT) signaling status, clinicopathologic features, and underlying causes leading to the loss of BCR. On the basis of clinical features, 15 (60%) DLBCL, BCR patients were classified into the low-risk group, and 18 (86%) experienced complete remission. Morphologically and immunophenotypically, DLBCL, BCR demonstrated centroblastic cytology (21/25, 84%) and germinal center B-cell-like cell origin (18/25, 72%). Other components in BCR complexity remained intact, on the basis of immunohistochemical findings. Epstein-Barr virus infection, deficiency in B-lineage transcription factors (PAX5, Oct-2, and Bob.1), and oncogene rearrangement did not seem to be associated with BCR loss. The activated form of signaling proteins (pSYK and pAKT) involved in the BCR/PI3K/AKT pathway were expressed at low levels in DLBCL, BCR tissue. In vitro validation revealed that in DLBCL, BCR cell lines, the BCR/PI3K/AKT pathway did not respond to BCR stimulation or inhibition. Our findings suggest that DLBCL, BCR was characterized by a silent BCR/PI3K/AKT pathway, germinal center phenotype, and low risk and may not be a candidate for BCR-targeted therapies.


Germinal Center , Lymphoma, Large B-Cell, Diffuse/classification , Lymphoma, Large B-Cell, Diffuse/pathology , Oncogene Protein v-akt/physiology , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-bcr/physiology , Signal Transduction , Adult , Aged , Aged, 80 and over , Female , Gene Expression Regulation, Neoplastic , Humans , Lymphoma, Large B-Cell, Diffuse/genetics , Male , Middle Aged , Phenotype , Risk Assessment , Young Adult
20.
Drug Des Devel Ther ; 9: 2463-73, 2015.
Article En | MEDLINE | ID: mdl-25995618

BACKGROUND: CCN1 (also called Cyr 61) is an extracellular matrix signaling molecule that has been implicated in neovascularization through its interactions with several endothelial integrin receptors. The roles of vascular endothelial growth factor (VEGF) in angiogenesis are well described. The aim of this study was to investigate the signal transduction mechanism of CCN1-PI3K/Akt-VEGF in retinopathy of prematurity (ROP), and the effects of CCN1 knockdown on ROP. METHODS: The oxygen-induced retinopathy (OIR) model was established in C57BL/6J mice exposed to a high concentration of oxygen. Retinas were obtained from the normoxia, OIR, OIR control (treated with scramble siRNA) and OIR treated (with CCN1 siRNA) groups. Retinal neovascularization (RNV) was qualitatively analyzed with ADPase staining and quantitatively analyzed by counting neovascular endothelial cell nuclei at postnatal day 17 when RNV reached a peak. mRNA level and protein expression of CCN1, p-Akt, and VEGF were measured by real-time PCR and Western blotting, and located with immunohistochemistry. RESULTS: CCN1 depletion resulted in less neovascularization clock hour scores in the number of preretinal neovascular cells compared with the OIR treated group (1.28±0.83 versus 4.80±0.82; and 7.12±2.50 versus 23.25±2.35, respectively, both P<0.05). Furthermore, CCN1, p-Akt and VEGF mRNA, and protein were significantly expressed in the retina of the OIR and OIR control groups. Intravitreal injection of CCN1 siRNA significantly reduced PI3K/Akt-VEGF pathway expression of the OIR mouse model (all P<0.05). CCN1 siRNA significantly enhanced the avascular area and avascular diameter of OIR model (P<0.05). CCN1 siRNA decreased the levels of IL-1ß, IL-6, and TNF-α significantly compared to the OIR group (P<0.05). CONCLUSION: These results suggest that CCN1 plays an important role in RNV via the PI3K/Akt-VEGF signaling pathway. CCN1 may be a potential target for the prevention and treatment of ROP.


Cysteine-Rich Protein 61/genetics , Retinal Neovascularization/drug therapy , Retinopathy of Prematurity/drug therapy , Signal Transduction/drug effects , Animals , Animals, Newborn , Apyrase/metabolism , Cysteine-Rich Protein 61/drug effects , Cytokines/metabolism , Endothelial Cells/drug effects , Female , Gene Knockdown Techniques , Male , Mice , Mice, Inbred C57BL , Oncogene Protein v-akt/physiology , Oxygen/toxicity , Phosphatidylinositol 3-Kinases/physiology , RNA, Small Interfering/pharmacology , Retinopathy of Prematurity/chemically induced , Vascular Endothelial Growth Factor A/physiology
...