1.Dexmedetomidine Attenuates High Glucose-induced HK-2 Epithelial-mesenchymal Transition by Inhibiting AKT and ERK.
Qi Zheng PAN ; Kai LI ; Zhuo Dong YANG ; Ming GAO ; Jia Hong SHI ; Shu Ping REN ; Guo Qing ZHAO
Biomedical and Environmental Sciences 2020;33(5):323-330
Objective:
To explore the protective effects of dexmedetomidine (Dex) against high glucose-induced epithelial-mesenchymal transition in HK-2 cells and relevant mechanisms.
Methods:
HK-2 cells were exposed to either glucose or glucose+Dex for 6 h. The production of ROS, morphology of HK-2 cells, and cell cycle were detected. Moreover, the expression of AKT, p-AKT, ERK, p-ERK, PI3K, E-Cadherin, Claudin-1, and α-SMA were determined and compared between HK-2 cells exposed to glucose and those exposed to both glucose and Dex with or without PI3K/AKT pathway inhibitor LY294002 and ERK pathway inhibitor U0126.
Results:
Compared with HK-2 cells exposed to high level of glucose, the HK-2 cells exposed to both high level of glucose and Dex showed: (1) lower level of ROS production; (2) cell morphology was complete; (3) more cells in G1 phase; (4) lower expression of p-AKT, p-ERK and α-SMA, higher expression of E-Cadherin and Claudin-1. PI3K/AKT inhibitor LY294002 and ERK inhibitor U0126 decreased the expression of p-AKT, p-ERK and α-SMA, and increased the expression of E-Cadherin and Claudin-1.
Conclusion
Dex can attenuate high glucose-induced HK-2 epithelial-mesenchymal transition by inhibiting AKT and ERK.
Adrenergic alpha-2 Receptor Agonists
;
pharmacology
;
Cell Line
;
Dexmedetomidine
;
pharmacology
;
Epithelial-Mesenchymal Transition
;
drug effects
;
Glucose
;
metabolism
;
Humans
;
MAP Kinase Signaling System
;
drug effects
;
Proto-Oncogene Proteins c-akt
;
antagonists & inhibitors
;
Signal Transduction
;
drug effects
2.Effects of estrogen receptor GPR30 agonist G1 on neuronal apoptosis and microglia polarization in traumatic brain injury rats.
Meng-Xian PAN ; Jun-Chun TANG ; Rui LIU ; Yu-Gong FENG ; Qi WAN
Chinese Journal of Traumatology 2018;21(4):224-228
PURPOSETo investigate the effects of estrogen G protein-coupled receptor 30 (GPR30) agonist G1 on hippocampal neuronal apoptosis and microglial polarization in rat traumatic brain injury (TBI).
METHODSMale SD rats were randomly divided into sham group, TBI + vehicle group, TBI + G1 group. Experimental moderate TBI was induced using Feeney's weigh-drop method. G1 (100μg/kg) or vehicle was intravenously injected from femoral vein at 30 min post-injury. Rats were sacrificed at 24 h after injury for detection of neuronal apoptosis and microglia polarization. Neuronal apoptosis was assayed by immunofluorescent staining of active caspase-3. M1 type microglia markers (iNOS and IL-1β) and M2 type markers (Arg1 and IL-4) were examined by immunoblotting or ELISA. Total protein level of Akt and phosphorylated Akt were assayed by immunoblotting.
RESULTSG1 significantly reduced active caspase-3 positive neurons in hippocampus. Meanwhile G1 increased the ratio of Arg1/iNOS. IL-1β production was decreased but IL-4 was increased after G1 treatment. G1 treatment also increased the active form of Akt.
CONCLUSIONSGPR30 agonist G1 inhibited neuronal apoptosis and favored microglia polarization to M2 type.
Animals ; Apoptosis ; drug effects ; Brain Injuries, Traumatic ; drug therapy ; pathology ; Cell Polarity ; Hippocampus ; drug effects ; Interleukin-1beta ; biosynthesis ; Male ; Microglia ; drug effects ; Neurons ; drug effects ; Proto-Oncogene Proteins c-akt ; metabolism ; Rats ; Rats, Sprague-Dawley ; Receptors, G-Protein-Coupled ; agonists
3.β3-adrenoceptor impacts apoptosis in cultured cardiomyocytes via activation of PI3K/Akt and p38MAPK.
Miao-miao MA ; Xiao-li ZHU ; Li WANG ; Xiao-fang HU ; Zhong WANG ; Jin ZHAO ; Yi-tong MA ; Yi-ning YANG ; Bang-dang CHEN ; Fen LIU
Journal of Huazhong University of Science and Technology (Medical Sciences) 2016;36(1):1-7
β3-adrenoceptor (β3-AR) has been shown to promote myocardial apoptosis. However, the exact physiological role and importance of this receptor in the human myocardium, and its underlying mode of action, have not been fully elucidated. The present study aimed to determine the effects of β3-AR on the promotion of myocardial apoptosis and on norepinephrine (NE) injury. We analyzed NE-induced cardiomyocyte (CM) apoptosis by using a TUNEL and an annexin V/propidium iodide apoptosis assay. Furthermore, we investigated the NE-induced expression of the apoptosis marker genes Akt and p38MAPK, their phosphorylated counterparts p-Akt and p-p38MAPK, caspase-3, Bcl-2, and Bax. In addition, we determined the effect of a 48-h treatment with a β3-AR agonist and antagonist on expression of these marker genes. β3-AR overexpression was found to increase CM apoptosis, accompanied by an increased expression of caspase-3, bax/bcl-2, and p-p38MAPK. In contrast, the β3-blocker reduced apoptosis of CMs and the associated elevated Akt expression. We identified a novel and potent anti-apoptosis mechanism via the PI3K/Akt pathway and a pro-apoptosis pathway mediated by p38MAPK.
Adrenergic Agonists
;
pharmacology
;
Adrenergic Antagonists
;
pharmacology
;
Animals
;
Apoptosis
;
Cells, Cultured
;
Myocytes, Cardiac
;
drug effects
;
metabolism
;
Phosphatidylinositol 3-Kinases
;
metabolism
;
Proto-Oncogene Proteins c-akt
;
metabolism
;
Rats
;
Rats, Sprague-Dawley
;
Receptors, Adrenergic, beta-3
;
genetics
;
metabolism
;
Signal Transduction
;
p38 Mitogen-Activated Protein Kinases
;
metabolism
4.Lysophosphatidic acid induces cell migration through the selective activation of Akt1.
Eun Kyoung KIM ; Sung Ji YUN ; Kee Hun DO ; Min Sung KIM ; Mong CHO ; Dong Soo SUH ; Chi Dae KIM ; Jae Ho KIM ; Morris J BIRNBAUM ; Sun Sik BAE
Experimental & Molecular Medicine 2008;40(4):445-452
Akt plays pivotal roles in many physiological responses including growth, proliferation, survival, metabolism, and migration. In the current studies, we have evaluated the isoform-specific role of akt in lysophosphatidic acid (LPA)-induced cell migration. Ascites from ovarian cancer patients (AOCP) induced mouse embryo fibroblast (MEF) cell migration in a dose-dependent manner. On the other hand, ascites from liver cirrhosis patients (ALCP) did not induce MEF cell migration. AOCP-induced MEF cell migration was completely blocked by pre-treatment of cells with LPA receptor antagonist, Ki16425. Both LPA- and AOCP-induced MEF cell migration was completely attenuated by PI3K inhibitor, LY294002. Furthermore, cells lacking Akt1 displayed defect in LPA-induced cell migration. Re-expression of Akt1 in DKO (Akt1(-/-)Akt2(-/-)) cells restored LPA-induced cell migration, whereas re-expression of Akt2 in DKO cells could not restore the LPA-induced cell migration. Finally, Akt1 was selectively phosphorylated by LPA and AOCP stimulation. These results suggest that LPA is a major factor responsible for AOCP-induced cell migration and signaling specificity of Akt1 may dictate LPA-induced cell migration.
1-Phosphatidylinositol 3-Kinase/physiology
;
Adult
;
Aged
;
Animals
;
Ascites/pathology
;
Cell Movement/*drug effects
;
Cells, Cultured
;
Embryo, Mammalian
;
Enzyme Activation/drug effects
;
Female
;
Humans
;
Liver Cirrhosis/pathology
;
Lysophospholipids/isolation & purification/*pharmacology
;
Mice
;
Middle Aged
;
Ovarian Neoplasms/pathology
;
Pregnancy
;
Proto-Oncogene Proteins c-akt/*agonists/*metabolism
;
Substrate Specificity
5.P2Y purinergic receptor activated PI-3K/Akt signaling pathway in regulation of growth and invasion of prostatic cancer.
Yu-xiang WANG ; Yong-hong SHI ; Li-hua GONG ; Yan LI ; Wan-jie HENG ; Jiang-feng YOU ; Hao-hao ZHONG ; Wei-gang FANG
Chinese Journal of Pathology 2007;36(10):681-686
OBJECTIVETo investigate P2Y purinergic receptor activated PI-3K/Akt signaling pathway in the regulation of growth and invasion of prostate cancer in vitro.
METHODSWestern blot was used to detect phosphorylation of Akt (a downstream target molecule of PI-3K) by P2Y receptor agonist in 1E8 cells (a highly metastatic subclone derived from PC-3 prostatic cancer cell line). Cell counts, flow cytometry, Matrigel invasion assay, wound healing assay and gelatin zymography were used to detect changes of biological behaviors of 1E8 cells after P2Y receptor activation.
RESULTSAMP-PNP, one non-hydrolysis ATP analogue and P2Y receptor agonist, induced significant phosphorylation of Akt in a time- and dose-dependent manner in IE8 cells. LY294002, a specific inhibitor of PI-3K, effectively blocked Akt phosphorylation induced by AMP-PNP. Continuous exposure to AMP-PNP induced significant growth inhibition of 1E8 cells (inhibition rate at 50.2% at the 8th day), and this inhibition was mainly due to an arrest at S phase of the cell cycle (the S phase fraction of AMP-PNP treated cells was 22.3% higher than that of the control). Application of LY294002 did not reverse the growth inhibition effect of AMP-PNP. Matrigel invasion assay showed that AMP-PNP stimulation increased invasive ability of 1E8 cells, and this effect was effectively blocked by LY294002. No significant changes in the activation of MMP-2 and MMP-9 were detected by gelatin zymography, although wound healing assay showed 21.2% increase in cell migration after AMP-PNP treatment.
CONCLUSIONSPI-3K/Akt signaling pathway participates in P2Y receptor-stimulated prostate cancer invasion by enhancing cell motility, rather than up-regulating MMP-2 and MMP-9 activities. PI-3K signaling pathway plays an important role in prostate cancer proliferation, but is not involved in P2Y receptor mediated growth inhibition.
Adenylyl Imidodiphosphate ; pharmacology ; Animals ; Apoptosis ; drug effects ; Cell Line, Tumor ; Cell Movement ; drug effects ; Cell Proliferation ; drug effects ; Chromones ; pharmacology ; Humans ; Male ; Matrix Metalloproteinase 2 ; metabolism ; Matrix Metalloproteinase 9 ; metabolism ; Mice ; Mice, Nude ; Morpholines ; pharmacology ; Neoplasm Invasiveness ; Phosphatidylinositol 3-Kinases ; antagonists & inhibitors ; metabolism ; Phosphorylation ; Prostatic Neoplasms ; metabolism ; pathology ; Proto-Oncogene Proteins c-akt ; metabolism ; Purinergic P2 Receptor Agonists ; S Phase ; drug effects ; Signal Transduction ; drug effects