1.β-arrestin2 recruitment by β-adrenergic receptor agonists and antagonists.
Yi-Ran WANG ; De-Qin CHENG ; Lan MA ; Xing LIU
Acta Physiologica Sinica 2022;74(6):993-1004
A large number of β-adrenergic receptor (β-AR) agonists and antagonists are widely used in the treatment of cardiovascular diseases and other diseases. Nonetheless, it remains unclear whether these commonly used β-AR drugs can activate downstream β- arrestin-biased signaling pathways. The objective of this study was to investigate β-arrestin2 recruitment effects of β-AR agonists and antagonists that were commonly used in clinical practice. We used TANGO (transcriptional activation following arrestin translocation) assay to detect the β-arrestin2 recruitment by β-AR ligands in HEK293 cell line (HTLA cells) stably transfected with tetracycline transactivator protein (tTA) dependent luciferase reporter and β-arrestin2-TEV fusion gene. Upon activation of β-AR by a β-AR ligand, β-arrestin2 was recruited to the C terminus of the receptor, followed by cleavage of the G protein-coupled receptors (GPCRs) fusion protein at the TEV protease-cleavage site. The cleavage resulted in the release of tTA, which, after being transported to the nucleus, activated transcription of the luciferase reporter gene. The results showed that β-AR non-selective agonists epinephrine, noradrenaline and isoprenaline all promoted β-arrestin2 recruitment at β1-AR and β2-AR. β1-AR selective agonists dobutamine and denopamine both promoted β-arrestin2 recruitment at β1-AR. β2-AR selective agonists procaterol and salbutamol promoted β-arrestin2 recruitment at β2-AR. β-AR non-selective antagonists alprenolol and pindolol promoted β-arrestin2 recruitment at β1-AR. β1-AR selective antagonists celiprolol and bevantolol showed β-arrestin2 recruitment at β1-AR. β2-AR selective antagonists butoxamine showed β-arrestin2 recruitment at β1-AR. These results provide some clues for the potential action of β-AR drugs, and lay a foundation for the screening of β-arrestin-biased β-AR ligands.
Humans
;
beta-Arrestin 2/metabolism*
;
HEK293 Cells
;
Adrenergic beta-Agonists/pharmacology*
;
Isoproterenol/pharmacology*
;
Receptors, Adrenergic, beta-2/metabolism*
;
Norepinephrine/pharmacology*
2.Dexmedetomidine improves alcohol withdrawal symptom via activating α2 adrenergic receptor in rat hippocampus.
Ting ZENG ; Hong-Yan ZHANG ; Xin ZHAO ; Yan LIU ; Yan-Zhong GUAN
Acta Physiologica Sinica 2022;74(4):541-547
The purpose of this study was to investigate the effects of α2 adrenergic receptor agonist dexmedetomidine on withdrawal symptoms in alcohol-dependent rats and the underlying mechanism, so as to provide a scientific basis for the treatment of alcohol withdrawal syndrome (AWS). Adult Sprague-Dawley (SD) male rats were orally administered with 6% aqueous alcohol continuously for 28 d to establish alcohol drinking model, and then stopped drinking to induce AWS. Enzyme-linked immunosorbent assay (ELISA) was used to determine the content of norepinephrine (NE) in the locus coeruleus and hippocampus of rats. Dexmedetomidine (5, 10, and 20 μg/kg) was intraperitoneally injected respectively when the rats showed significant AWS. In some rats, α2 adrenergic receptor antagonist yohimbine was injected into hippocampus in advance. The results showed that, compared with the control group, the 6 h withdrawal group exhibited significantly increased AWS score and amount of repeat drinking. The NE contents in hippocampus and locus coeruleus of the last drinking and the 6 h withdrawal groups were significantly increased compared with those of the control group. Dexmedetomidine intervention significantly decreased AWS score and hippocampus NE content in the 6 h withdrawal group, while yohimbine could reverse these effects of dexmedetomidine. These results suggest that dexmedetomidine might improve the withdrawal symptoms in alcohol-dependent rats via activating α2 adrenergic receptor.
Adrenergic alpha-2 Receptor Agonists/therapeutic use*
;
Alcoholism/drug therapy*
;
Animals
;
Dexmedetomidine/therapeutic use*
;
Hippocampus/metabolism*
;
Male
;
Norepinephrine
;
Rats
;
Rats, Sprague-Dawley
;
Receptors, Adrenergic, alpha-2/metabolism*
;
Substance Withdrawal Syndrome/drug therapy*
;
Yohimbine/pharmacology*
3.Dexmedetomidine Promotes Angiogenesis and Vasculogenic Mimicry in Human Hepatocellular Carcinoma through α 2-AR/HIF-1α/VEGFA Pathway.
Tao FANG ; Li LIN ; Zhi Jian YE ; Lian FANG ; Shuai SHI ; Ke Da YU ; Hui Hui MIAO ; Tian Zuo LI
Biomedical and Environmental Sciences 2022;35(10):931-942
OBJECTIVE:
Dexmedetomidine (DEX), the most specific α 2-adrenergic receptor agonist widely used for its sedative and analgesic properties, has been reported to upregulate HIF-1α expression to protect hypoxic and ischemic tissues. However, it is largely unclear whether DEX can also upregulate Hypoxia-inducible factor-1 alpha (HIF-1α) expression and its downstream vascular endothelial growth factor-A (VEGFA) in cancer tissues with oxygen-deficient tumor microenvironment.
METHODS:
We used SMMC-7721 cells, MHCC97-H cells, and a mouse model of orthotopic hepatic carcinoma to explore the effect of DEX on angiogenesis and vasculogenic mimicry (VM) and its mechanism. Under normoxic (20% O 2) and hypoxic (1% O 2) conditions, DEX was used to intervene cells, and yohimbine was used to rescue them.
RESULTS:
The results showed that DEX promoted angiogenesis and VM in human liver cancer cells within a certain dose range, and the addition of yohimbine inhibited this effect. DEX could activate HIF-1α/VEGFA pathway, which was further verified by silencing HIF-1α. Consistently, in vivo results also showed that DEX can up-regulate HIF-1α/VEGFA expression, and enhance the number of VM channels and microvessel density (MVD).
CONCLUSION
We believe that HIF-1α/VEGFA might be an important signaling pathway by which DEX promotes angiogenesis and VM formation in human hepatocellular carcinoma, whereas α 2-adrenergic receptor mediation might be the critical mechanisms.
Animals
;
Humans
;
Mice
;
Adrenergic alpha-2 Receptor Agonists/pharmacology*
;
Carcinoma, Hepatocellular
;
Cardiovascular Physiological Phenomena
;
Dexmedetomidine/pharmacology*
;
Hypoxia
;
Liver Neoplasms/drug therapy*
;
Oxygen
;
Tumor Microenvironment
;
Vascular Endothelial Growth Factor A/genetics*
;
Receptors, Adrenergic, alpha-2/metabolism*
4.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
5.Electroacupuncture Alleviates Motor Symptoms and Up-Regulates Vesicular Glutamatergic Transporter 1 Expression in the Subthalamic Nucleus in a Unilateral 6-Hydroxydopamine-Lesioned Hemi-Parkinsonian Rat Model.
Yanyan WANG ; Yong WANG ; Junhua LIU ; Xiaomin WANG
Neuroscience Bulletin 2018;34(3):476-484
Previous studies have shown that electroacupuncture (EA) promotes recovery of motor function in Parkinson's disease (PD). However the mechanisms are not completely understood. Clinically, the subthalamic nucleus (STN) is a critical target for deep brain stimulation treatment of PD, and vesicular glutamate transporter 1 (VGluT1) plays an important role in the modulation of glutamate in the STN derived from the cortex. In this study, a 6-hydroxydopamine (6-OHDA)-lesioned rat model of PD was treated with 100 Hz EA for 4 weeks. Immunohistochemical analysis of tyrosine hydroxylase (TH) showed that EA treatment had no effect on TH expression in the ipsilateral striatum or substantia nigra pars compacta, though it alleviated several of the parkinsonian motor symptoms. Compared with the hemi-parkinsonian rats without EA treatment, the 100 Hz EA treatment significantly decreased apomorphine-induced rotation and increased the latency in the Rotarod test. Notably, the EA treatment reversed the 6-OHDA-induced down-regulation of VGluT1 in the STN. The results demonstrated that EA alleviated motor symptoms and up-regulated VGluT1 in the ipsilateral STN of hemi-parkinsonian rats, suggesting that up-regulation of VGluT1 in the STN may be related to the effects of EA on parkinsonian motor symptoms via restoration of function in the cortico-STN pathway.
Adrenergic Agents
;
toxicity
;
Animals
;
Apomorphine
;
pharmacology
;
Disease Models, Animal
;
Dopamine Agonists
;
pharmacology
;
Electroacupuncture
;
methods
;
Functional Laterality
;
drug effects
;
Male
;
Medial Forebrain Bundle
;
injuries
;
Motor Activity
;
drug effects
;
physiology
;
Neurons
;
drug effects
;
metabolism
;
Oxidopamine
;
toxicity
;
Parkinson Disease, Secondary
;
chemically induced
;
physiopathology
;
therapy
;
Rats
;
Rats, Sprague-Dawley
;
Subthalamic Nucleus
;
drug effects
;
metabolism
;
pathology
;
Tyrosine 3-Monooxygenase
;
metabolism
;
Up-Regulation
;
drug effects
;
physiology
;
Vesicular Glutamate Transport Protein 1
;
metabolism
6.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
7.Isoprenaline Induces Periostin Expression in Gastric Cancer.
Guo Xiao LIU ; Hong Qing XI ; Xiao Yan SUN ; Zhi Jun GENG ; Shao Wei YANG ; Yan Jie LU ; Bo WEI ; Lin CHEN
Yonsei Medical Journal 2016;57(3):557-564
PURPOSE: Periostin mediates critical steps in gastric cancer and is involved in various signaling pathways. However, the roles of periostin in promoting gastric cancer metastasis are not clear. The aim of this study was to investigate the relevance between periostin expression and gastric cancer progression and the role of stress-related hormones in the regulation of cancer development and progression. MATERIALS AND METHODS: Normal, cancerous and metastatic gastric tissues were collected from patients diagnosed with advanced gastric cancer. The in vivo expression of periostin was evaluated by in situ hybridization and immunofluorescent staining. Meanwhile, human gastric adenocarcinoma cell lines MKN-45 and BGC-803 were used to detect the in vitro expression of periostin by using quantitative real-time polymerase chain reaction (PCR) and western blotting. RESULTS: Periostin is expressed in the stroma of the primary gastric tumors and metastases, but not in normal gastric tissue. In addition, we observed that periostin is located mainly in pericryptal fibroblasts, but not in the tumor cells, and strongly correlated to the expression of α-smooth muscle actin (SMA). Furthermore, the distribution patterns of periostin were broader as the clinical staging of tumors progressed. We also identified a role of stress-related signaling in promoting cancer development and progression, and found that isoprenaline upregulated expression levels of periostin in gastric cancer cells. CONCLUSION: These findings suggest that the distribution pattern of periostin was broader as the clinical staging of the tumor progressed and found that isoprenaline upregulated expression levels of periostin in gastric cancer cells.
Adenocarcinoma/*metabolism/pathology
;
Adrenergic beta-Agonists/pharmacology
;
Aged
;
Blotting, Western
;
Cell Adhesion Molecules/drug effects/*metabolism
;
Cell Line, Tumor
;
Fibroblasts/*metabolism
;
Gene Expression Regulation, Neoplastic/*drug effects
;
Humans
;
Isoproterenol/*pharmacology
;
Male
;
Neoplasm Staging
;
RNA, Messenger/genetics/metabolism
;
Real-Time Polymerase Chain Reaction
;
Signal Transduction
;
Stomach/metabolism/pathology
;
Stomach Neoplasms/*metabolism/pathology
;
Up-Regulation
8.Dexmedetomidine alleviates pulmonary edema by upregulating AQP1 and AQP5 expression in rats with acute lung injury induced by lipopolysaccharide.
Yuan-xu JIANG ; Zhong-liang DAI ; Xue-ping ZHANG ; Wei ZHAO ; Qiang HUANG ; Li-kun GAO
Journal of Huazhong University of Science and Technology (Medical Sciences) 2015;35(5):684-688
This study aims to elucidate the mechanisms by which dexmedetomidine alleviates pulmonary edema in rats with acute lung injury induced by lipopolysaccharide (LPS). Male Wistar rats were randomly divided into five groups: normal saline control (NS) group, receiving intravenous 0.9% normal saline (5 mL/kg); LPS group, receiving intravenous LPS (10 mg/kg); small-dose dexmedetomidine (S) group, treated with a small dose of dexmedetomidine (0.5 μg · kg(-1) · h(-1)); medium-dose dexmedetomidine (M) group, treated with a medium dose of dexmedetomidine (2.5 μg · kg(-1) · h(-1)); high-dose dexmedetomidine (H) group, treated with a high dose of dexmedetomidine (5 μg · kg(-1) · h(-1)). The rats were sacrificed 6 h after intravenous injection of LPS or NS, and the lungs were removed for evaluating histological characteristics and determining the lung wet/dry weight ratio (W/D). The levels of tumor necrosis factor-alpha (TNF-α) and interleukin-1β (IL-1β) in the lung tissues were assessed by enzyme- linked immunosorbent assay (ELISA). The mRNA and protein expression levels of aquaporin-1 (AQP1) and aquaporin-5 (AQP5) were detected by RT-PCR, immunohistochemistry, and Western blotting. The lung tissues from the LPS groups were significantly damaged, which were less pronounced in the H group but not in the small-dose dexmedetomidine group or medium-dose dexmedetomidine group. The W/D and the concentrations of TNF-α and IL-1β in the pulmonary tissues were increased in the LPS group as compared with those in NS group, which were reduced in the H group but not in S group or M group (P<0.01). The expression of AQP1 and AQP5 was lower in the LPS group than in the NS group, and significantly increased in the H group but not in the S group or M group (P<0.01). Our findings suggest that dexmedetomidine may alleviate pulmonary edema by increasing the expression of AQP-1 and AQP-5.
Acute Lung Injury
;
chemically induced
;
drug therapy
;
genetics
;
pathology
;
Adrenergic alpha-2 Receptor Agonists
;
pharmacology
;
Animals
;
Aquaporin 1
;
agonists
;
genetics
;
immunology
;
Aquaporin 5
;
agonists
;
genetics
;
immunology
;
Dexmedetomidine
;
pharmacology
;
Dose-Response Relationship, Drug
;
Drug Administration Schedule
;
Gene Expression Regulation
;
Injections, Intravenous
;
Interleukin-1beta
;
antagonists & inhibitors
;
genetics
;
immunology
;
Lipopolysaccharides
;
Lung
;
drug effects
;
immunology
;
pathology
;
Male
;
Organ Size
;
drug effects
;
Pulmonary Edema
;
chemically induced
;
drug therapy
;
genetics
;
pathology
;
Rats
;
Rats, Wistar
;
Signal Transduction
;
Transcription, Genetic
;
Tumor Necrosis Factor-alpha
;
antagonists & inhibitors
;
genetics
;
immunology
9.Updated roles of adrenergic receptors in prostate cancer.
Heng-Quan LUO ; Xiang-Xing KUANG ; Ben-Yi LI
National Journal of Andrology 2014;20(4):372-376
Adrenergic receptors are members of the G-protein coupled receptor superfamily. Recent studies revealed that these adrenergic receptors are playing an important role in the growth and metastasis of prostate cancer cells. The expression of adrenergic receptors rises significantly in prostate cancer cells and tissues. Agonists of these receptors promote the growth and mobility of prostate cancer cells, while antagonists may suppress their proliferation, trigger their apoptosis, and inhibit their metastasis. Clinically, receptor antagonists can significantly reduce the risk of prostate cancer and improve its prognosis after androgen depravation therapy. This article presents an overview on the roles of adrenergic receptors in prostate cancer.
Adrenergic Agonists
;
pharmacology
;
Adrenergic Antagonists
;
pharmacology
;
Apoptosis
;
Humans
;
Male
;
Prostatic Neoplasms
;
metabolism
;
pathology
;
Receptors, Adrenergic
;
drug effects
;
physiology
10.Effects of anti-glaucoma drugs on resistive index of the medial long posterior ciliary artery using color Doppler imaging in Beagle dogs.
Hojung CHOI ; Youngwon LEE ; Seongchan YEON ; Hyojong LEE ; Heechun LEE
Journal of Veterinary Science 2011;12(1):99-101
Color Doppler imaging (CDI) was carried out to evaluate the effects of anti-glaucoma drugs on ophthalmic circulation using CDI-derived resistive index (RI) values. CDI was performed on nine Beagle dogs, and RI values were calculated for the medial long posterior ciliary artery before and after the administration of anti-glaucoma drugs. A significant increase in RI values was found after topical administration of levobunolol (p < 0.05) or dipivefrin (p < 0.05). Pilocarpine showed no effects on RI values after topical administration. The results suggest that some anti-glaucoma drugs could affect ophthalmic blood flow.
Adrenergic Agonists/pharmacology
;
Animals
;
Ciliary Arteries/*drug effects/*ultra
;
Dogs
;
Epinephrine/analogs & derivatives/therapeutic use
;
Eye/*blood supply
;
Female
;
Glaucoma/*drug therapy
;
Levobunolol/therapeutic use
;
Male
;
Ocular Physiological Phenomena
;
Pilocarpine/therapeutic use
;
Ultra
;
*Vascular Resistance

Result Analysis
Print
Save
E-mail