1051
|
Jia YM, Zhou H, Tai T, Gu TT, Ji JZ, Mi QY, Huang BB, Li YF, Zhu T, Xie HG. Enhanced responsiveness of platelets to vicagrel in IL-10-deficient mice through STAT3-dependent up-regulation of the hydrolase arylacetamide deacetylase in the intestine. Br J Pharmacol 2019; 176:1717-1727. [PMID: 30825385 DOI: 10.1111/bph.14646] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 01/10/2019] [Accepted: 02/05/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND AND PURPOSE Vicagrel is a novel promising antiplatelet drug designed for overcoming clopidogrel resistance. There is limited evidence indicating that exogenous IL-10 suppresses CYP3A4 activity in healthy subjects and that IL-10 knockout (KO) mice exhibit increased clopidogrel bioactivation compared with wild-type (WT) mice. In this study, we sought to determine whether IL-10 could play an important role in the metabolism of and platelet response to vicagrel in mice. EXPERIMENTAL APPROACH IL-10 KO and WT mice were administered vicagrel, then their plasma H4 (active metabolite of vicagrel) concentrations were determined by LC-MS/MS, and inhibition of ADP-induced whole-blood platelet aggregation by vicagrel was assessed with an aggregometer. The mRNA and protein levels of several relevant genes between IL-10 KO and WT mice were measured by qRT-PCR and Western blots, respectively. Intestinal Aadac protein levels were measured in IL-10 WT mice injected i.p. with vehicle control, Stattic, or BAY 11-7082. KEY RESULTS Compared with WT mice, IL-10 KO mice exhibited significantly increased plasma levels of H4 and enhanced platelet responses to vicagrel, as well as significantly higher mRNA and protein levels of arylacetamide deacetylase (Aadac) in the intestine. In WT mice, STAT3, not NF-κB, mediated Aadac expression in the intestine. CONCLUSIONS AND IMPLICATIONS IL-10 suppresses metabolic activation of vicagrel through down-regulation of Aadac in mouse intestine in a STAT3-dependent manner and, consequently, attenuates platelet responses to vicagrel, suggesting that the antiplatelet effect of vicagrel may be modulated by changes in plasma IL-10 levels in relevant clinical settings.
Collapse
Affiliation(s)
- Yu-Meng Jia
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.,Department of Pharmacology, College of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Huan Zhou
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.,Department of Pharmacology, College of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Ting Tai
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Tong-Tong Gu
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Jin-Zi Ji
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Qiong-Yu Mi
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Bei-Bei Huang
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yi-Fei Li
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Ting Zhu
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.,Department of Pharmacology, College of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Hong-Guang Xie
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.,Department of Pharmacology, College of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China.,Department of Clinical Pharmacy, Nanjing Medical University School of Pharmacy, Nanjing, China
| |
Collapse
|
1052
|
Clark JW, Drummond SPA, Hoyer D, Jacobson LH. Sex differences in mouse models of fear inhibition: Fear extinction, safety learning, and fear-safety discrimination. Br J Pharmacol 2019; 176:4149-4158. [PMID: 30710446 DOI: 10.1111/bph.14600] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 11/26/2018] [Accepted: 01/04/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND AND PURPOSE Women are overrepresented in post-traumatic stress disorder (PTSD), a mental disorder characterised by ineffective inhibition of fear. The use of male animals dominates preclinical studies, which may contribute to a lack of understanding as to why this disparity exists. Thus, the current study explores sex differences in three mouse models of fear inhibition. EXPERIMENTAL APPROACH All experiments tested male and female C57Bl/6J mice. Experiment 1 employed two fear conditioning protocols, in which tones were paired with footshocks of differing intensity (moderate or intense). Fear recall and extinction were tested subsequently. In Experiment 2, safety learning was investigated. Tones were explicitly unpaired with footshocks during safety conditioning. Recall of safety learning was tested 24 hr later. Experiment 3 assessed a model of fear-safety discrimination. Cued stimuli were paired or never paired with footshocks during fear and safety conditioning, respectively. Discrimination between stimuli was assessed 24 hr later. KEY RESULTS In fear extinction, males, compared to females, responded with greater fear in sessions most proximal to conditioning but subsequently showed a more rapid fear extinction over time. Sex differences were not observed during safety learning. During fear-safety discrimination, both males and females discriminated between stimuli; however, males revealed a greater level of freezing to stimuli. CONCLUSION AND IMPLICATIONS The current study provides evidence that sex differences influence fear but not safety-based behaviour in C57Bl/6J mice. These findings indicate that processing of fear, but not safety, may play a greater role in sex differences observed for PTSD. LINKED ARTICLES This article is part of a themed section on The Importance of Sex Differences in Pharmacology Research. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.21/issuetoc.
Collapse
Affiliation(s)
- Jacob W Clark
- Monash Institute of Cognitive and Clinical Neuroscience, Monash University, Notting Hill, VIC, Australia.,Department of Pharmacology and Therapeutics, School of Biomedical Sciences, University of Melbourne, Parkville, VIC, Australia.,The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - Sean P A Drummond
- Monash Institute of Cognitive and Clinical Neuroscience, Monash University, Notting Hill, VIC, Australia
| | - Daniel Hoyer
- Department of Pharmacology and Therapeutics, School of Biomedical Sciences, University of Melbourne, Parkville, VIC, Australia.,The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia.,Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Laura H Jacobson
- Department of Pharmacology and Therapeutics, School of Biomedical Sciences, University of Melbourne, Parkville, VIC, Australia.,The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
1053
|
Okada M, Fukuyama K, Kawano Y, Shiroyama T, Ueda Y. Memantine protects thalamocortical hyper-glutamatergic transmission induced by NMDA receptor antagonism via activation of system xc<sup/>. Pharmacol Res Perspect 2019; 7:e00457. [PMID: 30631447 PMCID: PMC6323135 DOI: 10.1002/prp2.457] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 12/14/2018] [Indexed: 12/26/2022] Open
Abstract
Deficiencies in N-methyl-d-aspartate (NMDA)/glutamate receptor (NMDAR) signaling have been considered central to the cognitive impairments of schizophrenia; however, an NMDAR antagonist memantine (MEM) improves cognitive impairments of Alzheimer's disease and schizophrenia. These mechanisms of paradoxical clinical effects of NMDAR antagonists remain unclear. To explore the mechanisms by which MK801 and MEM affect thalamocortical transmission, we determined interactions between local administrations of MK801, MEM, system xc- (Sxc), and metabotropic glutamate receptors (mGluRs) on extracellular glutamate and GABA levels in the mediodorsal thalamic nucleus (MDTN) and medial prefrontal cortex (mPFC) using dual-probe microdialysis with ultra-high-pressure liquid chromatography. Effects of MK801 and MEM on Sxc activity were also determined using primary cultured astrocytes. Sxc activity was enhanced by MEM, but was unaffected by MK801. MK801 enhanced thalamocortical glutamatergic transmission by GABAergic disinhibition in the MDTN. In the MDTN and the mPFC, MEM weakly increased glutamate release by activating Sxc, whereas MEM inhibited thalamocortical glutamatergic transmission. Paradoxical effects of MEM were induced following secondary activation of inhibitory II-mGluR and III-mGluR by exporting glutamate from astroglial Sxc. The present results suggest that the effects of therapeutically relevant concentrations of MEM on thalamocortical glutamatergic transmission are predominantly caused by activation of Sxc rather than inhibition of NMDAR. These demonstrations suggest that the combination between reduced NMDAR and activated Sxc contribute to the neuroprotective effects of MEM. Furthermore, activation of Sxc may compensate for the cognitive impairments that are induced by hyperactivation of thalamocortical glutamatergic transmission following activation of Sxc/II-mGluR in the MDTN and Sxc/II-mGluR/III-mGluR in the mPFC.
Collapse
Affiliation(s)
- Motohiro Okada
- Department of NeuropsychiatryDivision of NeuroscienceGraduate School of MedicineMie UniversityTsuMieJapan
| | - Kouji Fukuyama
- Department of NeuropsychiatryDivision of NeuroscienceGraduate School of MedicineMie UniversityTsuMieJapan
| | - Yasuhiro Kawano
- Department of NeuropsychiatryDivision of NeuroscienceGraduate School of MedicineMie UniversityTsuMieJapan
| | - Takashi Shiroyama
- Department of NeuropsychiatryDivision of NeuroscienceGraduate School of MedicineMie UniversityTsuMieJapan
| | - Yuto Ueda
- Department of NeuropsychiatryDivision of NeuroscienceGraduate School of MedicineMie UniversityTsuMieJapan
| |
Collapse
|
1054
|
Nam SY, Kim HY, Min JY, Kim HM, Jeong HJ. An osteoclastogenesis system, the RANKL/RANK signalling pathway, contributes to aggravated allergic inflammation. Br J Pharmacol 2019; 176:1664-1679. [PMID: 30737962 DOI: 10.1111/bph.14615] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 12/21/2018] [Accepted: 01/08/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND AND PURPOSE As an osteoclast differentiation factor, receptor activator of NF-κB ligand (RANKL) is produced by various immune cells and may be involved in the pathogenesis of osteoporosis and inflammation. Although RANKL is expressed in most immune cells and tissues, it is not clear how this might affect allergic inflammation. EXPERIMENTAL APPROACH The roles of RANKL in allergic rhinitis (AR) were analysed in an ovalbumin (OVA)-induced animal model, human subjects, and a human mast cell line (HMC-1). Small interfering RNA experiments were performed in an OVA-induced AR model. KEY RESULTS RANKL and RANKL receptor (RANK) were up-regulated in serum or nasal mucosal tissues of AR patients and AR mice. RANKL and RANK were colocalised in mast cells of nasal mucosa tissue. Depletion of RANKL by RANKL siRNA ameliorated AR symptoms and reduced AR-related biomarkers, including thymic stromal lymphopoietin (TSLP), IgE, histamine, and inflammatory cell infiltration, whereas recombinant RANKL increased AR responses and TSLP levels. In addition, functional deficiency of TSLP decreased AR responses induced by RANKL. In human mast cells, interaction of RANKL with RANK increased production of TSLP and inflammatory cytokines. Production of TSLP by RANKL stimulation was mediated through activation of the PI3K, MAPK, caspase-1, and NF-κB pathways. Furthermore, dexamethasone alleviated RANKL-induced inflammatory reactions in AR models. CONCLUSION AND IMPLICATIONS Collectively, these data suggest that RANKL may induce development of AR through up-regulation of TSLP.
Collapse
Affiliation(s)
- Sun-Young Nam
- Department of Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul, Korea
| | - Hee-Yun Kim
- Department of Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul, Korea
| | - Jin-Young Min
- Department of Otolaryngology-Head and Neck Surgery, Kyung Hee University, School of Medicine, Seoul, Korea
| | - Hyung-Min Kim
- Department of Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul, Korea
| | - Hyun-Ja Jeong
- Division of Food and Pharmaceutical Engineering, Biochip Research Center and Inflammatory Diseases Research Center, Hoseo University, Asan, Korea
| |
Collapse
|
1055
|
Zhang F, Wan H, Yang X, He J, Lu C, Yang S, Tuo B, Dong H. Molecular mechanisms of caffeine-mediated intestinal epithelial ion transports. Br J Pharmacol 2019; 176:1700-1716. [PMID: 30808064 DOI: 10.1111/bph.14640] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 01/10/2019] [Accepted: 01/31/2019] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND AND PURPOSE As little is known about the effect of caffeine, one of the most widely consumed substances worldwide, on intestinal function, we aimed to study its action on intestinal anion secretion and the underlying molecular mechanisms. EXPERIMENTAL APPROACH Anion secretion and channel expression were examined in mouse duodenal epithelium by Ussing chambers and immunocytochemistry. Ca2+ imaging was also performed in intestinal epithelial cells (IECs). KEY RESULTS Caffeine (10 mM) markedly increased mouse duodenal short-circuit current (Isc ), which was attenuated by a removal of either Cl- or HCO3 - , Ca2+ -free serosal solutions and selective blockers of store-operated Ca2+ channels (SOC/Ca2+ release-activated Ca2+ channels), and knockdown of Orai1 channels on the serosal side of duodenal tissues. Caffeine induced SOC entry in IEC, which was inhibited by ruthenium red and selective blockers of SOC. Caffeine-stimulated duodenal Isc was inhibited by the endoplasmic reticulum Ca2+ chelator (N,N,N',N'-tetrakis(2-pyridylmethyl)ethylenediamine), selective blockers (ruthenium red and dantrolene) of ryanodine receptors (RyR), and of Ca2+ -activated Cl- channels (niflumic acid and T16A). There was synergism between cAMP and Ca2+ signalling, in which cAMP/PKA promoted caffeine/Ca2+ -mediated anion secretion. Expression of STIM1 and Orai1 was detected in mouse duodenal mucosa and human IECs. The Orai1 proteins were primarily co-located with the basolateral marker Na+ , K+ -ATPase. CONCLUSIONS AND IMPLICATIONS Caffeine stimulated intestinal anion secretion mainly through the RyR/Orai1/Ca2+ signalling pathway. There is synergism between cAMP/PKA and caffeine/Ca2+ -mediated anion secretion. Our findings suggest that a caffeine-mediated RyR/Orai1/Ca2+ pathway could provide novel potential drug targets to control intestinal anion secretion.
Collapse
Affiliation(s)
- Fenglian Zhang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Hanxing Wan
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Xin Yang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Jialin He
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Cheng Lu
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Shiming Yang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Biguang Tuo
- Department of Gastroenterology, Affiliated Hospital, Zunyi Medical College, and Digestive Disease Institute of Guizhou Province, Zunyi, China
| | - Hui Dong
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China.,Department of Medicine, School of Medicine, University of California, San Diego, California, USA
| |
Collapse
|
1056
|
Alvarez‐Collazo J, López‐Requena A, Galán L, Talavera A, Alvarez JL, Talavera K. The citrus flavanone hesperetin preferentially inhibits slow-inactivating currents of a long QT syndrome type 3 syndrome Na + channel mutation. Br J Pharmacol 2019; 176:1090-1105. [PMID: 30650182 PMCID: PMC6451064 DOI: 10.1111/bph.14577] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 12/12/2018] [Accepted: 12/21/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND AND PURPOSE The citrus flavanone hesperetin has been proposed for the treatment of several human pathologies, but its cardiovascular actions remain largely unexplored. Here, we evaluated the effect of hesperetin on cardiac electrical and contractile activities, on aortic contraction, on the wild-type voltage-gated NaV 1.5 channel, and on a channel mutant (R1623Q) associated with lethal ventricular arrhythmias in the long QT syndrome type 3 (LQT3). EXPERIMENTAL APPROACH We used cardiac surface ECG and contraction force recordings to evaluate the effects of hesperetin in rat isolated hearts and aortic rings. Whole-cell patch clamp was used to record NaV 1.5 currents (INa ) in rat ventricular cardiomyocytes and in HEK293T cells expressing hNaV 1.5 wild-type or mutant channels. KEY RESULTS Hesperetin increased the QRS interval and heart rate and decreased the corrected QT interval and the cardiac and aortic contraction forces at concentrations equal or higher than 30 μmol·L-1 . Hesperetin blocked rat and human NaV 1.5 channels with an effective inhibitory concentration of ≈100 μmol·L-1 . This inhibition was enhanced at depolarized holding potentials and higher stimulation frequency and was reduced by the disruption of the binding site for local anaesthetics. Hesperetin increased the rate of inactivation and preferentially inhibited INa during the slow inactivation phase, these effects being more pronounced in the R1623Q mutant. CONCLUSIONS AND IMPLICATIONS Hesperetin preferentially inhibits the slow inactivation phase of INa , more markedly in the mutant R1623Q. Hesperetin could be used as a template to develop drugs against lethal cardiac arrhythmias in LQT3.
Collapse
Affiliation(s)
- Julio Alvarez‐Collazo
- Laboratory of Ion Channel Research, Department of Cellular and Molecular MedicineVIB‐KU Leuven Center for Brain & Disease ResearchLeuvenBelgium
| | - Alejandro López‐Requena
- Laboratory of Ion Channel Research, Department of Cellular and Molecular MedicineVIB‐KU Leuven Center for Brain & Disease ResearchLeuvenBelgium
| | - Loipa Galán
- Laboratory of ElectrophysiologyInstitute of Cardiology and Cardiovascular SurgeryHavanaCuba
| | - Ariel Talavera
- Laboratory of Microscopy, Center for Microscopy and Molecular ImagingUniversité Libre de BruxellesGosseliesBelgium
| | - Julio L. Alvarez
- Laboratory of ElectrophysiologyInstitute of Cardiology and Cardiovascular SurgeryHavanaCuba
| | - Karel Talavera
- Laboratory of Ion Channel Research, Department of Cellular and Molecular MedicineVIB‐KU Leuven Center for Brain & Disease ResearchLeuvenBelgium
| |
Collapse
|
1057
|
Mao Z, Han X, Chen D, Xu Y, Xu L, Yin L, Sun H, Qi Y, Fang L, Liu K, Peng J. Potent effects of dioscin against hepatocellular carcinoma through regulating TP53-induced glycolysis and apoptosis regulator (TIGAR)-mediated apoptosis, autophagy, and DNA damage. Br J Pharmacol 2019; 176:919-937. [PMID: 30710454 PMCID: PMC6433650 DOI: 10.1111/bph.14594] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 12/05/2018] [Accepted: 12/18/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND AND PURPOSE Dioscin shows potent effects against cancers. We aimed to elucidate its pharmacological effects and mechanisms of action on hepatocellular carcinoma (HCC) in vivo and in vitro. EXPERIMENTAL APPROACH Effects of dioscin were investigated in SMMC7721 and HepG2 cells, diethylnitrosamine-induced primary liver cancer in rats, and cell xenografts in nude mice. Isobaric tags for relative and absolution quantitation (iTRAQ)-based proteomics was used to find dioscin's targets and investigate its mechanism. KEY RESULTS In SMMC7721 and HepG2 cells dioscin markedly inhibited cell proliferation and migration, induced apoptosis, autophagy, and DNA damage. It inhibited DEN-induced primary liver cancer in rats, markedly changed body weights and restored levels of α fetoprotein, alanine transaminase, aspartate transaminase, γ-glutamyltransferase, alkaline phosphatase, and Ki67. It also inhibited growth of xenografts in mice. In SMMC7721 cells, 191 differentially expressed proteins were found after dioscin, based on iTRAQ-based assay. TP53-inducible glycolysis and apoptosis regulator (TIGAR) was identified as being significantly down-regulated by dioscin. Dioscin induced cell apoptosis, autophagy, and DNA damage via increasing expression levels of p53, cleaved PARP, Bax, cleaved caspase-3/9, Beclin-1, and LC3 and suppressing those of Bcl-2, p-Akt, p-mammalian target of rapamycin (mTOR), CDK5, p-ataxia telangiectasia-mutated gene (ATM). The transfection of TIGAR siRNA into SMMC7721 cells and xenografts in nude mice further confirmed that the potent activity of dioscin against HCC is evoked by adjusting TIGAR-mediated inhibition of p53, Akt/mTOR, and CDK5/ATM pathways. CONCLUSIONS AND IMPLICATIONS The data suggest that dioscin has potential as a therapeutic, and TIGAR as a drug target for treating HCC.
Collapse
Affiliation(s)
- Zhang Mao
- College of PharmacyDalian Medical UniversityDalianChina
| | - Xu Han
- College of PharmacyDalian Medical UniversityDalianChina
| | - Dahong Chen
- College of PharmacyDalian Medical UniversityDalianChina
| | - Youwei Xu
- College of PharmacyDalian Medical UniversityDalianChina
| | - Lina Xu
- College of PharmacyDalian Medical UniversityDalianChina
| | - Lianhong Yin
- College of PharmacyDalian Medical UniversityDalianChina
| | - Huijun Sun
- College of PharmacyDalian Medical UniversityDalianChina
| | - Yan Qi
- College of PharmacyDalian Medical UniversityDalianChina
| | - Lingling Fang
- College of PharmacyDalian Medical UniversityDalianChina
| | - Kexin Liu
- College of PharmacyDalian Medical UniversityDalianChina
| | - Jinyong Peng
- College of PharmacyDalian Medical UniversityDalianChina
- Key Laboratory for Basic and Applied Research on Pharmacodynamic Substances of Traditional Chinese Medicine of Liaoning ProvinceDalian Medical UniversityDalianChina
- National‐Local Joint Engineering Research Center for Drug Development (R&D) of Neurodegenerative DiseasesDalian Medical UniversityDalianChina
| |
Collapse
|
1058
|
Gu M, Zhao P, Zhang S, Fan S, Yang L, Tong Q, Ji G, Huang C. Betulinic acid alleviates endoplasmic reticulum stress-mediated nonalcoholic fatty liver disease through activation of farnesoid X receptors in mice. Br J Pharmacol 2019; 176:847-863. [PMID: 30635917 PMCID: PMC6433649 DOI: 10.1111/bph.14570] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 12/04/2018] [Accepted: 12/09/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND PURPOSE The molecular mechanism for the pathogenesis of nonalcoholic fatty liver disease (NAFLD) remains elusive. Both farnesoid X receptor (FXR) signalling and endoplasmic reticulum (ER) stress contribute to the progression of NAFLD; however, it is not clear whether the actions of these two pathways are dependent on each other. Moreover, the pharmacological benefits and mechanism of betulinic acid (BA) in controlling metabolic syndrome and NAFLD are largely unknown. EXPERIMENTAL APPROACH A reporter assay and a time-resolved FRET assay were used to identify BA as an agonist of the FXR. NAFLD was induced by a methionine and choline-deficient L-amino acid diet (MCD) and high-fat diet (HFD). The pharmacological effects of BA (100 mg·kg-1 ·day-1 ) and potential interactions between hepatic FXR activation and ER stress pathways were evaluated by FXR silencing, Western blot and RT-PCR analyses using control and FXR-/- mice. KEY RESULTS Activation of the FXR inhibited intracellular PERK/EIF2α/ATF4 and CHOP signalling, thereby alleviating hepatic ER stress, whereas FXR silencing resulted in an opposite effect. Furthermore, we identified BA as an FXR agonist that effectively attenuated the progression of NAFLD and metabolic disorders in both HFD- and MCD diet-fed mice and restored the hepatocellular ER homeostasis by stimulating the FXR signalling pathway and blocking PERK/EIF2α signalling. In contrast, the effects of BA were attenuated in FXR-/- mice. CONCLUSIONS AND IMPLICATIONS Our data demonstrate that pharmacological activation of the FXR by BA reduces hepatocellular ER stress and attenuates NAFLD in an animal model of hepatic steatosis.
Collapse
Affiliation(s)
- Ming Gu
- Institute of Digestive Disease, Longhua HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
- School of PharmacyShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Ping Zhao
- School of PharmacyShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Shiying Zhang
- School of PharmacyShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Shengjie Fan
- School of PharmacyShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Li Yang
- Research Center for Traditional Chinese Medicine of Complexity SystemsShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Qingchun Tong
- Brown Foundation Institute of Molecular Medicine and Program in Neuroscience, Graduate School of Biological SciencesUniversity of Texas McGovern Medical SchoolHoustonTexasUSA
| | - Guang Ji
- Institute of Digestive Disease, Longhua HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Cheng Huang
- School of PharmacyShanghai University of Traditional Chinese MedicineShanghaiChina
| |
Collapse
|
1059
|
Li J, Tan YJ, Wang MZ, Sun Y, Li GY, Wang QL, Yao JC, Yue J, Liu Z, Zhang GM, Ren YS. Loganetin protects against rhabdomyolysis-induced acute kidney injury by modulating the toll-like receptor 4 signalling pathway. Br J Pharmacol 2019; 176:1106-1121. [PMID: 30706443 DOI: 10.1111/bph.14595] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 12/10/2018] [Accepted: 01/01/2019] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND AND PURPOSE Acute kidney injury (AKI) is a rapid renal dysfunctional disease, for which no effective drugs or therapies are available to improve prognosis. Loganetin is a natural product with unknown bioactivities. Here, we identified a new protective effect and mechanism of Loganetin in a mouse model of AKI induced by rhabdomyolysis. EXPERIMENTAL APPROACH AKI was induced using glycerol by i.m. injection in mice models. Thirty minutes and 24 and 48 hr after injection of glycerol, the mice received 2 and 18 mg·kg-1 of Loganetin i.p. respectively. Then mice blood and kidney were collected for various biochemical and histopathological studies. Mechanistic studies on modulation of AKI by Loganetin were performed using HK-2 cells and Toll-like receptor 4 (TLR4) knockout mice. KEY RESULTS In the Loganetin treated group, kidney damage and mortality rate were declined, and blood urea nitrogen and serum creatinine were much lower. Loganetin prevented damage to the tubular structures induced by glycerol and decreased apoptotic cells at the corticomedullary junction. In HK-2 cells, Loganetin could inhibit NF-κB pathway and pro-apoptotic genes expression. However, TLR4 was silenced by a specific shRNA, and the inhibitory effect of Loganetin in HK-2 cells vanished. Loganetin also down-regulated the expression of inflammation factors by suppressing TLR4 activity. CONCLUSION AND IMPLICATIONS All the results suggested that TLR4 plays a critical role in AKI development, and Loganetin ameliorates AKI by inhibiting TLR4 activity and blocking the JNK/p38 pathway, which provides a new strategy for AKI treatment.
Collapse
Affiliation(s)
- Jie Li
- Shandong New Time Pharmaceutical Co., Ltd., Lunan Pharmaceutical Group Co., Ltd., Linyi, China.,Center for New Drug Safety Evaluation of Lunan Pharmaceutical, Lunan Pharmaceutical Group Co., Ltd., Linyi, China.,State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co., Ltd., Linyi, China
| | - Yu-Jun Tan
- Shandong New Time Pharmaceutical Co., Ltd., Lunan Pharmaceutical Group Co., Ltd., Linyi, China.,Center for New Drug Safety Evaluation of Lunan Pharmaceutical, Lunan Pharmaceutical Group Co., Ltd., Linyi, China.,State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co., Ltd., Linyi, China
| | - Ming-Zhi Wang
- Shandong New Time Pharmaceutical Co., Ltd., Lunan Pharmaceutical Group Co., Ltd., Linyi, China.,Center for New Drug Safety Evaluation of Lunan Pharmaceutical, Lunan Pharmaceutical Group Co., Ltd., Linyi, China.,State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co., Ltd., Linyi, China
| | - Ying Sun
- Shandong New Time Pharmaceutical Co., Ltd., Lunan Pharmaceutical Group Co., Ltd., Linyi, China.,Center for New Drug Safety Evaluation of Lunan Pharmaceutical, Lunan Pharmaceutical Group Co., Ltd., Linyi, China.,State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co., Ltd., Linyi, China
| | - Guang-Yan Li
- Shandong New Time Pharmaceutical Co., Ltd., Lunan Pharmaceutical Group Co., Ltd., Linyi, China.,State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co., Ltd., Linyi, China
| | - Qi-Long Wang
- Shandong New Time Pharmaceutical Co., Ltd., Lunan Pharmaceutical Group Co., Ltd., Linyi, China.,Center for New Drug Safety Evaluation of Lunan Pharmaceutical, Lunan Pharmaceutical Group Co., Ltd., Linyi, China.,State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co., Ltd., Linyi, China
| | - Jing-Chun Yao
- Shandong New Time Pharmaceutical Co., Ltd., Lunan Pharmaceutical Group Co., Ltd., Linyi, China.,Center for New Drug Safety Evaluation of Lunan Pharmaceutical, Lunan Pharmaceutical Group Co., Ltd., Linyi, China.,State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co., Ltd., Linyi, China
| | - Jiang Yue
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, China
| | - Zhong Liu
- Shandong New Time Pharmaceutical Co., Ltd., Lunan Pharmaceutical Group Co., Ltd., Linyi, China.,State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co., Ltd., Linyi, China.,National Engineering and Technology Research Center of Chirality Pharmaceutical, Lunan Pharmaceutical Group Co., Ltd., Linyi, China
| | - Gui-Min Zhang
- Shandong New Time Pharmaceutical Co., Ltd., Lunan Pharmaceutical Group Co., Ltd., Linyi, China.,State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co., Ltd., Linyi, China
| | - Yu-Shan Ren
- Shandong New Time Pharmaceutical Co., Ltd., Lunan Pharmaceutical Group Co., Ltd., Linyi, China.,State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co., Ltd., Linyi, China
| |
Collapse
|
1060
|
Parlakpinar H, Ozhan O, Ermis N, Vardi N, Cigremis Y, Tanriverdi LH, Colak C, Acet A. Acute and Subacute Effects of Low Versus High Doses of Standardized Panax ginseng Extract on the Heart: An Experimental Study. Cardiovasc Toxicol 2019; 19:306-320. [DOI: 10.1007/s12012-019-09512-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
1061
|
Antonazzo M, Gutierrez-Ceballos A, Bustinza I, Ugedo L, Morera-Herreras T. Cannabinoids differentially modulate cortical information transmission through the sensorimotor or medial prefrontal basal ganglia circuits. Br J Pharmacol 2019; 176:1156-1169. [PMID: 30735570 PMCID: PMC6451076 DOI: 10.1111/bph.14613] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 12/10/2018] [Accepted: 01/02/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND PURPOSE In the sensorimotor (SM) and medial prefrontal (mPF) basal ganglia (BG) circuits, the cortical information is transferred to the substantia nigra pars reticulata (SNr) through the hyperdirect trans-subthalamic pathway and through the direct and indirect trans-striatal pathways. The cannabinoid CB1 receptor, which is highly expressed in both BG circuits, may participate in the regulation of motor and motivational behaviours. Here, we investigated the modulation of cortico-nigral information transmission through the BG circuits by cannabinoids. EXPERIMENTAL APPROACH We used single-unit recordings of SNr neurons along with simultaneous electrical stimulation of motor or mPF cortex in anaesthetized rats. KEY RESULTS Cortical stimulation elicited a triphasic response in the SNr neurons from both SM and mPF-BG circuits, which consisted of an early excitation (hyperdirect transmission pathway), an inhibition (direct transmission pathway), and a late excitation (indirect transmission pathway). In the SM circuit, after Δ9 -tetrahydrocannabinol or WIN 55,212-2 administration, the inhibition and the late excitation were decreased or completely lost, whereas the early excitation response remained unaltered. However, cannabinoid administration dramatically decreased all the responses in the mPF circuit. The CB1 receptor antagonist AM251 (2 mg·kg-1 , i.v.) did not modify the triphasic response, but blocked the effects induced by cannabinoid agonists. CONCLUSIONS AND IMPLICATIONS CB1 receptor activation modulates the SM information transmission through the trans-striatal pathways and profoundly decreases the cortico-BG transmission through the mPF circuit. These results may be relevant for elucidating the involvement of the cannabinoid system in motor performance and in decision making or goal-directed behaviour.
Collapse
Affiliation(s)
- Mario Antonazzo
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain.,Neurodegenerative Diseases Group, Biocruces Health Research Institute, Barakaldo, Spain
| | - Amaia Gutierrez-Ceballos
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain.,Neurodegenerative Diseases Group, Biocruces Health Research Institute, Barakaldo, Spain
| | - Irati Bustinza
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain.,Neurodegenerative Diseases Group, Biocruces Health Research Institute, Barakaldo, Spain
| | - Luisa Ugedo
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain.,Neurodegenerative Diseases Group, Biocruces Health Research Institute, Barakaldo, Spain
| | - Teresa Morera-Herreras
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain.,Neurodegenerative Diseases Group, Biocruces Health Research Institute, Barakaldo, Spain
| |
Collapse
|
1062
|
Park SJ, Im DS. Blockage of sphingosine-1-phosphate receptor 2 attenuates allergic asthma in mice. Br J Pharmacol 2019; 176:938-949. [PMID: 30706444 DOI: 10.1111/bph.14597] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 11/19/2018] [Accepted: 12/10/2018] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND AND PURPOSE Sphingosine-1-phosphate 2 (S1P2 ) receptors have been implicated in degranulation of mast cells. However, functions of S1P2 receptors have not been investigated in an in vivo model of allergic asthma. EXPERIMENTAL APPROACH Using an ovalbumin (OVA)-induced asthma model, the function of S1P2 receptors was evaluated in S1P2 -deficient mice or in mice treated with JTE-013, a selective S1P2 antagonist. Bone marrow-derived dendritic cells (BMDCs) were used to investigate the roles of S1P2 receptors in dendritic cell maturation and migration. KEY RESULTS Eosinophil accumulation and elevated Th2 cytokine levels in bronchoalveolar lavage fluid and inflamed lung tissues were strongly inhibited by administration of JTE-013 before OVA sensitization, before OVA challenge, and before both events. In S1P2 -deficient mice, allergic responses were significantly lower than in wild-type mice. LPS- and OVA-induced maturation of BMDCs was significantly blunted in dendritic cells from S1P2 -deficient mice and by treatment with JTE-013. Migrations of immature and mature BMDCs were also dependent on S1P2 receptors. It was found that OVA-challenged mice into which in vitro OVA primed BMDCs from S1P2 -deficient mice were adoptively transferred, had less severe asthma responses than OVA-challenged mice into which OVA-primed BMDCs from wild-type mice were adoptively transferred. CONCLUSIONS AND IMPLICATIONS Pro-allergic functions of S1P2 receptors were elucidated in a murine asthma model. S1P2 receptors were involved not only in maturation and migration of dendritic cells in the sensitization phase but also in mast cell degranulation in the challenge phase. These results suggest S1P2 receptor as a therapeutic target for allergic asthma.
Collapse
Affiliation(s)
- Soo-Jin Park
- Molecular Inflammation Research Center for Aging Intervention (MRCA) and College of Pharmacy, Pusan National University, Busan, Korea
| | - Dong-Soon Im
- Molecular Inflammation Research Center for Aging Intervention (MRCA) and College of Pharmacy, Pusan National University, Busan, Korea
| |
Collapse
|
1063
|
Picard E, Carvalho FA, Agosti F, Bourinet E, Ardid D, Eschalier A, Daulhac L, Mallet C. Inhibition of Ca v 3.2 calcium channels: A new target for colonic hypersensitivity associated with low-grade inflammation. Br J Pharmacol 2019; 176:950-963. [PMID: 30714145 DOI: 10.1111/bph.14608] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 12/13/2018] [Accepted: 01/02/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND PURPOSE Abdominal pain associated with low-grade inflammation is frequently encountered in irritable bowel syndrome (IBS) and inflammatory bowel disease (IBD) during remission. Current treatments are not very effective and new therapeutic approaches are needed. The role of CaV 3.2 channels, which are important in other chronic pain contexts, was investigated in a murine model of colonic hypersensitivity (CHS) associated with low-grade inflammation. EXPERIMENTAL APPROACH Low doses of dextran sulfate sodium (DSS; 0.5%) were chronically administered to C57BL/6j mice in drinking water. Their inflammatory state was assessed by systemic and local measures of IL-6, myeloperoxidase, and lipocalin-2 using elisa. Colonic sensitivity was evaluated by the visceromotor responses to colorectal distension. Functional involvement of CaV 3.2 channels was assessed with different pharmacological (TTA-A2, ABT-639, and ethosuximide) and genetic tools. KEY RESULTS DSS induced low-grade inflammation associated with CHS in mice. Genetic or pharmacological inhibition of CaV 3.2 channels reduced CHS. Cav3.2 channel deletion in primary nociceptive neurons in dorsal root ganglia (CaV 3.2Nav1.8 KO mice) suppressed CHS. Spinal, but not systemic, administration of ABT-639, a peripherally acting T-type channel blocker, reduced CHS. ABT-639 given intrathecally to CaV 3.2Nav1.8 KO mice had no effect, demonstrating involvement of CaV 3.2 channels located presynaptically in afferent fibre terminals. Finally, ethosuximide, which is a T-type channel blocker used clinically, reduced CHS. CONCLUSIONS AND IMPLICATIONS These results suggest that ethosuximide represents a promising drug reposition strategy and that inhibition of CaV 3.2 channels is an attractive therapeutic approach for relieving CHS in IBS or IBD.
Collapse
Affiliation(s)
- Elodie Picard
- Université Clermont Auvergne, INSERM, CHU, NEURO-DOL Basics & Clinical Pharmacology of Pain, F-63000 Clermont-Ferrand, France.,ANALGESIA Institute, Université Clermont Auvergne, F-63000 Clermont-Ferrand, France
| | - Frederic Antonio Carvalho
- Université Clermont Auvergne, INSERM, CHU, NEURO-DOL Basics & Clinical Pharmacology of Pain, F-63000 Clermont-Ferrand, France.,ANALGESIA Institute, Université Clermont Auvergne, F-63000 Clermont-Ferrand, France
| | - Francina Agosti
- CNRS, INSERM, LABEX ICST, IGF, Université Montpellier, Montpellier, France
| | - Emmanuel Bourinet
- CNRS, INSERM, LABEX ICST, IGF, Université Montpellier, Montpellier, France
| | - Denis Ardid
- Université Clermont Auvergne, INSERM, CHU, NEURO-DOL Basics & Clinical Pharmacology of Pain, F-63000 Clermont-Ferrand, France.,ANALGESIA Institute, Université Clermont Auvergne, F-63000 Clermont-Ferrand, France
| | - Alain Eschalier
- Université Clermont Auvergne, INSERM, CHU, NEURO-DOL Basics & Clinical Pharmacology of Pain, F-63000 Clermont-Ferrand, France.,ANALGESIA Institute, Université Clermont Auvergne, F-63000 Clermont-Ferrand, France
| | - Laurence Daulhac
- Université Clermont Auvergne, INSERM, CHU, NEURO-DOL Basics & Clinical Pharmacology of Pain, F-63000 Clermont-Ferrand, France.,ANALGESIA Institute, Université Clermont Auvergne, F-63000 Clermont-Ferrand, France
| | - Christophe Mallet
- Université Clermont Auvergne, INSERM, CHU, NEURO-DOL Basics & Clinical Pharmacology of Pain, F-63000 Clermont-Ferrand, France.,ANALGESIA Institute, Université Clermont Auvergne, F-63000 Clermont-Ferrand, France
| |
Collapse
|
1064
|
Cooper SL, Soave M, Jörg M, Scammells PJ, Woolard J, Hill SJ. Probe dependence of allosteric enhancers on the binding affinity of adenosine A 1 -receptor agonists at rat and human A 1 -receptors measured using NanoBRET. Br J Pharmacol 2019; 176:864-878. [PMID: 30644086 PMCID: PMC6433648 DOI: 10.1111/bph.14575] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 11/02/2018] [Accepted: 12/04/2018] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND AND PURPOSE Adenosine is a local mediator that regulates a number of physiological and pathological processes via activation of adenosine A1 -receptors. The activity of adenosine can be regulated at the level of its target receptor via drugs that bind to an allosteric site on the A1 -receptor. Here, we have investigated the species and probe dependence of two allosteric modulators on the binding characteristics of fluorescent and nonfluorescent A1 -receptor agonists. EXPERIMENTAL APPROACH A Nano-luciferase (Nluc) BRET (NanoBRET) methodology was used. This used N-terminal Nluc-tagged A1 -receptors expressed in HEK293T cells in conjunction with both fluorescent A1 -receptor agonists (adenosine and NECA analogues) and a fluorescent antagonist CA200645. KEY RESULTS PD 81,723 and VCP171 elicited positive allosteric effects on the binding affinity of orthosteric agonists at both the rat and human A1 -receptors that showed clear probe dependence. Thus, the allosteric effect on the highly selective partial agonist capadenoson was much less marked than for the full agonists NECA, adenosine, and CCPA in both species. VCP171 and, to a lesser extent, PD 81,723, also increased the specific binding of three fluorescent A1 -receptor agonists in a species-dependent manner that involved increases in Bmax and pKD . CONCLUSIONS AND IMPLICATIONS These results demonstrate the power of the NanoBRET ligand-binding approach to study the effect of allosteric ligands on the binding of fluorescent agonists to the adenosine A1 -receptor in intact living cells. Furthermore, our studies suggest that VCP171 and PD 81,723 may switch a proportion of A1 -receptors to an active agonist conformation (R*).
Collapse
Affiliation(s)
- Samantha L Cooper
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, UK.,Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, The Midlands, UK
| | - Mark Soave
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, UK.,Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, The Midlands, UK
| | - Manuela Jörg
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Peter J Scammells
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Jeanette Woolard
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, UK.,Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, The Midlands, UK
| | - Stephen J Hill
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, UK.,Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, The Midlands, UK
| |
Collapse
|
1065
|
Abdollahi Govar A, Törő G, Szaniszlo P, Pavlidou A, Bibli SI, Thanki K, Resto VA, Chao C, Hellmich MR, Szabo C, Papapetropoulos A, Módis K. 3-Mercaptopyruvate sulfurtransferase supports endothelial cell angiogenesis and bioenergetics. Br J Pharmacol 2019; 177:866-883. [PMID: 30644090 DOI: 10.1111/bph.14574] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 11/26/2018] [Accepted: 12/11/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND AND PURPOSE During angiogenesis, quiescent endothelial cells (ECs) are activated by various stimuli to form new blood vessels from pre-existing ones in physiological and pathological conditions. Many research groups have shown that hydrogen sulfide (H2 S), the newest member of the gasotransmitter family, acts as a proangiogenic factor. To date, very little is known about the regulatory role of 3-mercaptopyruvate sulfurtransferase (3-MST), an important H2 S-producing enzyme in ECs. The aim of our study was to explore the potential role of 3-MST in human EC bioenergetics, metabolism, and angiogenesis. EXPERIMENTAL APPROACH To assess in vitro angiogenic responses, we used EA.hy926 human vascular ECs subjected to shRNA-mediated 3-MST attenuation and pharmacological inhibition of proliferation, migration, and tube-like network formation. To evaluate bioenergetic parameters, cell respiration, glycolysis, glucose uptake, and mitochondrial/glycolytic ATP production were measured. Finally, global metabolomic profiling was performed to determine the level of 669 metabolic compounds. KEY RESULTS 3-MST-attenuated ECs subjected to shRNA or pharmacological inhibition of 3-MST significantly reduced EC proliferation, migration, and tube-like network formation. 3-MST silencing also suppressed VEGF-induced EC migration. From bioenergetic and metabolic standpoints, 3-MST attenuation decreased mitochondrial respiration and mitochondrial ATP production, increased glucose uptake, and perturbed the entire EC metabolome. CONCLUSION AND IMPLICATIONS 3-MST regulates bioenergetics and morphological angiogenic functions in human ECs. The data presented in the current report support the view that 3-MST pathway may be a potential candidate for therapeutic modulation of angiogenesis. LINKED ARTICLES This article is part of a themed section on Hydrogen Sulfide in Biology & Medicine. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.4/issuetoc.
Collapse
Affiliation(s)
| | - Gábor Törő
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Peter Szaniszlo
- Department of Otolaryngology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Athanasia Pavlidou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Sofia-Iris Bibli
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt, Germany
| | - Ketan Thanki
- Department of Surgery, University of Texas Medical Branch, Galveston, Texas, USA
| | - Vicente A Resto
- Department of Otolaryngology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Celia Chao
- Department of Surgery, University of Texas Medical Branch, Galveston, Texas, USA
| | - Mark R Hellmich
- Department of Surgery, University of Texas Medical Branch, Galveston, Texas, USA
| | - Csaba Szabo
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, USA.,Chair of Pharmacology, Department of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Andreas Papapetropoulos
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece.,Clinical, Experimental Surgery and Translational Research Center, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Katalin Módis
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas, USA.,Department of Surgery, University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
1066
|
Cristiano C, Volpicelli F, Lippiello P, Buono B, Raucci F, Piccolo M, Iqbal AJ, Irace C, Miniaci MC, Perrone Capano C, Calignano A, Mascolo N, Maione F. Neutralization of IL-17 rescues amyloid-β-induced neuroinflammation and memory impairment. Br J Pharmacol 2019; 176:3544-3557. [PMID: 30673121 DOI: 10.1111/bph.14586] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 12/19/2018] [Accepted: 01/07/2019] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND AND PURPOSE Alzheimer's disease (AD) is a common neurodegenerative disease characterized by a neuroinflammatory state, and to date, there is no cure and its treatment represents a large unmet clinical need. The involvement of Th17 cells in the pathogenesis of AD-related neuroinflammation has been reported in several studies. However, the role of the cytokine, IL-17 has not been well addressed. Herein, we investigate the effects of IL-17 neutralizing antibody (IL-17Ab) injected by i.c.v. or intranasal (IN) routes on amyloid-β (Aβ)-induced neuroinflammation and memory impairment in mice. EXPERIMENTAL APPROACH Aβ1-42 was injected into cerebral ventricles of adult CD1 mice. These mice received IL-17Ab via i.c.v. either at 1 h prior to Aβ1-42 injection or IN 5 and 12 days after Aβ1-42 injection. After 7 and 14 days of Aβ1-42 administration, we evaluated olfactory, spatial and working memory and performed biochemical analyses on whole brain and specific brain areas. KEY RESULTS Pretreatment with IL-17Ab, given, i.c.v., markedly reduced Aβ1-42 -induced neurodegeneration, improved memory function, and prevented the increase of pro-inflammatory mediators in a dose-dependent manner at 7 and 14 days. Similarly, the double IN administration of IL-17Ab after Aβ1-42 injection reduced neurodegeneration, memory decline, and the levels of proinflammatory mediators and cytokines. CONCLUSION AND IMPLICATIONS These findings suggest that the IL-17Ab reduced neuroinflammation and behavioural symptoms induced by Aβ. The efficacy of IL-17Ab IN administration in reducing Aβ1-42 neurodegeneration points to a possible future therapeutic approach in patients with AD. LINKED ARTICLES This article is part of a themed section on Therapeutics for Dementia and Alzheimer's Disease: New Directions for Precision Medicine. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.18/issuetoc.
Collapse
Affiliation(s)
- Claudia Cristiano
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Floriana Volpicelli
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy.,Institute of Genetics and Biophysics "Adriano Buzzati Traverso," Developmental Biology and Genetics division, CNR, Naples, Italy
| | - Pellegrino Lippiello
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Benedetta Buono
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Federica Raucci
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Marialuisa Piccolo
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Asif Jilani Iqbal
- Institute of Cardiovascular Sciences (ICVS), College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Carlo Irace
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Maria Concetta Miniaci
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Carla Perrone Capano
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy.,Institute of Genetics and Biophysics "Adriano Buzzati Traverso," Developmental Biology and Genetics division, CNR, Naples, Italy
| | - Antonio Calignano
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Nicola Mascolo
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Francesco Maione
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| |
Collapse
|
1067
|
Abstract
The controversial plan for scientific research publications to be published in compliant Open Access Journals or on compliant Open Access Platforms is discussed. The article has been co-published with permission in European Heart Journal and British Journal of Pharmacology. The articles are identical except for minor stylistic and spelling differences in keeping with each journal's style. Either citation can be used when citing this article.
Collapse
Affiliation(s)
- Tomasz J. Guzik
- Cardiovascular Research Editorial Office, BHF Centre for Cardiovascular ResearchUniversity of GlasgowGlasgowUK
- Department of MedicineJagiellonian University Collegium MedicumKrakowPoland
| | - Amrita Ahluwalia
- British Journal of Pharmacology Editorial OfficeThe Schild Plot, BPS OfficesLondonUK
- The William Harvey Research Institute, Barts and The London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| |
Collapse
|
1068
|
Hoyer D, Wang MW, Yang D. Welcome editorial. MEDICINE IN DRUG DISCOVERY 2019. [DOI: 10.1016/j.medidd.2019.100005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
1069
|
Pugsley M, Hayes E, Saint D, Walker M. The antiarrhythmic actions of bisaramil and penticainide result from mixed cardiac ion channel blockade. Biomed Pharmacother 2019; 111:427-435. [DOI: 10.1016/j.biopha.2018.12.068] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 12/04/2018] [Accepted: 12/14/2018] [Indexed: 01/27/2023] Open
|
1070
|
Okada M, Fukuyama K, Kawano Y, Shiroyama T, Suzuki D, Ueda Y. Effects of acute and sub-chronic administrations of guanfacine on catecholaminergic transmissions in the orbitofrontal cortex. Neuropharmacology 2019; 156:107547. [PMID: 30802458 DOI: 10.1016/j.neuropharm.2019.02.029] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 02/15/2019] [Accepted: 02/21/2019] [Indexed: 12/20/2022]
Abstract
The selective α2A adrenoceptor agonist guanfacine reduces hyperactivity and improves cognitive impairment in patients with attention-deficit/hyperactivity disorder (ADHD). The major mechanisms of guanfacine have been considered to involve activation of postsynaptic α2A adrenoceptor in frontal pyramidal neurons. However, the effects of chronic guanfacine administration on catecholaminergic transmissions associated with the orbitofrontal cortex (OFC) remain unclear. To explore the mechanisms of action of guanfacine on catecholaminergic transmission, the effects of its acute local or sub-chronic systemic administration on catecholamine release within pathways from locus coeruleus (LC) to OFC and reticular thalamic nucleus (RTN), from RTN to mediodorsal thalamic nucleus (MDTN), and from MDTN to OFC were determined using multi-probe microdialysis with ultra-high performance liquid chromatography. Acute OFC local administration of guanfacine did not affect catecholamine release in OFC. Acute LC local and sub-chronic systemic administrations of guanfacine reduced norepinephrine release in LC, OFC and RTN, and also reduced GABA release in MDTN, whereas AMPA-induced (perfusion with AMPA into NDTN) releases of l-glutamate, norepinephrine and dopamine in OFC were enhanced by sub-chronic systemic guanfacine administration. This study identified that catecholaminergic transmission is composed of three pathways: direct noradrenergic and co-releasing catecholaminergic LC-OFC pathways and intermediate LC-OFC (LC-RTN-MDTN-OFC) pathway. We demonstrated the dual actions of guanfacine on catecholaminergic transmission: attenuation of direct noradrenergic LC-OFC transmission at the resting stage and enhancement of direct co-releasing catecholaminergic LC-OFC transmission via GABAergic disinhibition in the intermediate LC-OFC pathway. These dual actions of guanfacine probably contribute to clinical actions of guanfacine against ADHD and its comorbid symptoms. This article is part of the Special Issue entitled 'Current status of the neurobiology of aggression and impulsivity'.
Collapse
Affiliation(s)
- Motohiro Okada
- Department of Neuropsychiatry, Division of Neuroscience, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan.
| | - Kouji Fukuyama
- Department of Neuropsychiatry, Division of Neuroscience, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| | - Yasuhiro Kawano
- Department of Neuropsychiatry, Division of Neuroscience, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| | - Takashi Shiroyama
- Department of Neuropsychiatry, Division of Neuroscience, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| | - Dai Suzuki
- Department of Neuropsychiatry, Division of Neuroscience, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| | - Yuto Ueda
- Department of Neuropsychiatry, Division of Neuroscience, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| |
Collapse
|
1071
|
Schaper-Gerhardt K, Wohlert M, Mommert S, Kietzmann M, Werfel T, Gutzmer R. Stimulation of histamine H 4 receptors increases the production of IL-9 in Th9 polarized cells. Br J Pharmacol 2019; 177:614-622. [PMID: 30589077 DOI: 10.1111/bph.14566] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 10/25/2018] [Accepted: 11/19/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND PURPOSE Th9 cells represent a recently defined subset of CD4+ T-helper cells, characterized by a high production of IL-9. They are found at increased frequency in lesions of atopic dermatitis, where IL-9 is also elevated. As histamine is up-regulated in lesions of inflammatory skin diseases, we investigated the expression profile of histamine receptors and their functional role on Th9 cells. EXPERIMENTAL APPROACH Naïve CD4+ T-cells were purified from human peripheral blood mononuclear cells, using magnetic beads and further differentiated into Th9 cells. During differentiation, cells were additionally stimulated with histamine receptor agonists or left untreated. Histamine receptor expression as well as IL-9 production was measured. KEY RESULTS As proof of a successful differentiation, IL-9 production was measured at mRNA and protein level. Expression of mRNA for histamine H1 , H2 and H4 receptors were up-regulated in differentiated Th9 cells compared to Th0 cells, while no mRNA for the H3 receptor was detectable. Stimulation of Th9 cells with histamine significantly up-regulated expression of mRNA and protein for IL-9 . Experiments with specific histamine receptor agonists and antagonists revealed that this up-regulation was mediated by H4 receptors. CONCLUSIONS AND IMPLICATIONS In summary, our study demonstrates a functional role for histamine H4 receptors on Th9 cells, which might amplify the pro-inflammatory potency of these cells. Together with earlier studies on Th2 and Th17 cells, this study underlines the promising approach for the use of H4 receptor antagonists in inflammatory and allergic diseases such as atopic dermatitis. LINKED ARTICLES This article is part of a themed section on New Uses for 21st Century. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.3/issuetoc.
Collapse
Affiliation(s)
- Katrin Schaper-Gerhardt
- Division of Immunodermatology and Allergy Research, Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| | - Mareike Wohlert
- Division of Immunodermatology and Allergy Research, Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| | - Susanne Mommert
- Division of Immunodermatology and Allergy Research, Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| | - Manfred Kietzmann
- Department of Pharmacology, Toxicology and Pharmacy, Veterinary School Hannover, Hannover, Germany
| | - Thomas Werfel
- Division of Immunodermatology and Allergy Research, Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| | - Ralf Gutzmer
- Division of Immunodermatology and Allergy Research, Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| |
Collapse
|
1072
|
Li N, Li C, Han R, Wang Y, Yang M, Wang H, Tian J. LPM580098, a Novel Triple Reuptake Inhibitor of Serotonin, Noradrenaline, and Dopamine, Attenuates Neuropathic Pain. Front Pharmacol 2019; 10:53. [PMID: 30837867 PMCID: PMC6382704 DOI: 10.3389/fphar.2019.00053] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 01/18/2019] [Indexed: 11/13/2022] Open
Abstract
Background and Purpose: Sedation and somnolence remain serious adverse effects of the existing analgesics (e.g., pregabalin, duloxetine) for neuropathic pain. The available evidence indicates that serotonin (5-HT), noradrenaline (NE), and dopamine (DA) play important roles in modulating the descending inhibitory pain pathway and sleep-wake cycle. The aim of this work was to test the hypothesis that LPM580098, a novel triple reuptake inhibitor (TRI) of 5-HT, NE, and DA, has analgesic effect, and does not induce significant adverse effects associated with central inhibition, such as sedation and somnolence. Methods: The analgesic activity of LPM580098 was assessed on formalin test and spinal nerve ligation (SNL)-induced neuropathic pain model. Locomotor activity, pentobarbital sodium-induced sleeping and rota-rod tests were also conducted. In vitro binding and uptake assays, and Western blotting were performed to examine the potential mechanisms. Results: LPM580098 suppressed the nocifensive behaviors during phase II of the formalin test in mice. In SNL rats, LPM580098 (16 mg kg-1) inhibited mechanical allodynia, thermal hyperalgesia and hyperexcitation of wide-dynamic range (WDR) neurons, in which the effect of LPM580098 was similar to pregabalin (30 mg kg-1). However, pregabalin altered the spontaneous locomotion, affected pentobarbital sodium-induced sleep, and showed a trend to perform motor dysfunction, which were not induced by LPM580098. Mechanistically, LPM580098 inhibited the uptake of 5-HT, NE, and DA, improved pain-induced changes of the synaptic functional plasticity and structural plasticity possibly via downregulating the NR2B/CaMKIIα/GluR1 and Rac1/RhoA signaling pathways. Conclusion: Our results suggest that LPM580098, a novel TRI, is effective in attenuating neuropathic pain without producing unwanted sedation and somnolence associated with central nervous system (CNS) depressants.
Collapse
Affiliation(s)
- Nannan Li
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Chunmei Li
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Rui Han
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Yu Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Mina Yang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Hongbo Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Jingwei Tian
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| |
Collapse
|
1073
|
Pugsley MK, Yong SL, Goldin AL, Hayes ES, Walker MJA. Molecular charge associated with antiarrhythmic actions in a series of amino-2-cyclohexyl ester derivatives. Eur J Pharmacol 2019; 844:241-252. [PMID: 30571955 DOI: 10.1016/j.ejphar.2018.12.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Revised: 11/12/2018] [Accepted: 12/12/2018] [Indexed: 11/15/2022]
Abstract
A series of amino-2-cyclohexyl ester derivatives were studied for their ion channel blocking and antiarrhythmic actions in the rat and a structure-activity analysis was conducted. The compounds are similar in chemical structure except for ionizable amine groups (pK values 6.1-8.9) and the positional arrangements of aromatic naphthyl moieties. Ventricular arrhythmias were produced in rats by coronary-artery occlusion or electrical stimulation. The electrophysiological effects of these compounds on rat heart sodium channels (Nav1.5) expressed in Xenopus laevis oocytes and transient outward potassium currents (Kv4.3) from isolated rat ventricular myocytes were examined. The compounds reduced the incidence of ischemia-related arrhythmias and increased current threshold for induction of ventricular fibrillo-flutter (VFt) dose-dependently. As pK increased compounds showed a diminished effectiveness against ischemia-induced arrhythmias, and were less selective for ischemia- versus electrically-induced arrhythmias. Where tested, compounds produced a concentration-dependent tonic block of Nav1.5 channels. An increased potency for inhibition of Nav1.5 occurred when the external pH (pHo) was reduced to 6.5. Some compounds inhibited Kv4.3 in a pH-independent manner. Overall, the differences in antiarrhythmic and ion channel blocking properties in this series of compounds can be explained by differences in chemical structure. Antiarrhythmic activity for the amino-2-cyclohexyl ester derivatives is likely a function of mixed ion channel blockade in ischemic myocardium. These studies show that drug inhibition of Nav1.5 occurred at lower concentrations than Kv4.3 and was more sensitive to changes in the ionizable amine groups rather than on positional arrangements of the naphthyl constituents. These results offer insight into antiarrhythmic mechanisms of drug-ion channel interactions.
Collapse
Affiliation(s)
- Michael K Pugsley
- Safety Pharmacology & Toxicology, Fairfield, CT 06825, USA; Department of Pharmacology & Therapeutics, Faculty of Medicine, The University of British Columbia, 2176 Health Sciences Mall, Vancouver, BC, Canada, V6T 1Z3.
| | - Sandro L Yong
- Department of Pharmacology & Therapeutics, Faculty of Medicine, The University of British Columbia, 2176 Health Sciences Mall, Vancouver, BC, Canada, V6T 1Z3
| | - Alan L Goldin
- Department of Microbiology & Molecular Genetics, University of California, Irvine, California, CA 92697-4025, USA
| | - Eric S Hayes
- BioCurate Pty Ltd., Parkville, Victoria, Australia
| | - Michael J A Walker
- Department of Pharmacology & Therapeutics, Faculty of Medicine, The University of British Columbia, 2176 Health Sciences Mall, Vancouver, BC, Canada, V6T 1Z3
| |
Collapse
|
1074
|
Stéphanou A, Ballesta A. pH as a potential therapeutic target to improve temozolomide antitumor efficacy : A mechanistic modeling study. Pharmacol Res Perspect 2019; 7:e00454. [PMID: 30705757 PMCID: PMC6349072 DOI: 10.1002/prp2.454] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 11/15/2018] [Accepted: 11/28/2018] [Indexed: 12/13/2022] Open
Abstract
Despite intensive treatments including temozolomide (TMZ) administration, glioblastoma patient prognosis remains dismal and innovative therapeutic strategies are urgently needed. A systems pharmacology approach was undertaken to investigate TMZ pharmacokinetics-pharmacodynamics (PK-PD) incorporating the effect of local pH, tumor spatial configuration and micro-environment. A hybrid mathematical framework was designed coupling ordinary differential equations describing the intracellular reactions, with a spatial cellular automaton to individualize the cells. A differential drug impact on tumor and healthy cells at constant extracellular pH was computationally demonstrated as TMZ-induced DNA damage was larger in tumor cells as compared to normal cells due to less acidic intracellular pH in cancer cells. Optimality of TMZ efficacy defined as maximum difference between damage in tumor and healthy cells was reached for extracellular pH between 6.8 and 7.5. Next, TMZ PK-PD in a solid tumor was demonstrated to highly depend on its spatial configuration as spread cancer cells or fragmented tumors presented higher TMZ-induced damage as compared to compact tumor spheroid. Simulations highlighted that smaller tumors were less acidic than bigger ones allowing for faster TMZ activation and their closer distance to blood capillaries allowed for better drug penetration. For model parameters corresponding to U87 glioma cells, inter-cell variability in TMZ uptake play no role regarding the mean drug-induced damage in the whole cell population whereas this quantity was increased by inter-cell variability in TMZ efflux which was thus a disadvantage in terms of drug resistance. Overall, this study revealed pH as a new potential target to significantly improve TMZ antitumor efficacy.
Collapse
Affiliation(s)
| | - Annabelle Ballesta
- INSERM and Paris Sud universityUMRS 935Team “Cancer Chronotherapy and Postoperative Liver Functions”VillejuifFrance
- University of WarwickCoventryUK
| |
Collapse
|
1075
|
Li J, Huang J, Lu J, Guo Z, Li Z, Gao H, Wang P, Luo W, Cai S, Hu Y, Guo K, Wang L, Li Z, Wang M, Zhang X, Liu P. Sirtuin 1 represses PKC-ζ activity through regulating interplay of acetylation and phosphorylation in cardiac hypertrophy. Br J Pharmacol 2019; 176:416-435. [PMID: 30414383 PMCID: PMC6329629 DOI: 10.1111/bph.14538] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 09/20/2018] [Accepted: 10/17/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND AND PURPOSE Activation of PKC-ζ is closely linked to the pathogenesis of cardiac hypertrophy. PKC-ζ can be activated by certain lipid metabolites such as phosphatidylinositol (3,4,5)-trisphosphate and ceramide. However, its endogenous negative regulators are not well defined. Here, the role of the sirtuin1-PKC-ζ signalling axis and the underlying molecular mechanisms were investigated in cardiac hypertrophy. EXPERIMENTAL APPROACH Cellular hypertrophy in cultures of cardiac myocytes, from neonatal Sprague-Dawley rats, was monitored by measuring cell surface area and the mRNA levels of hypertrophic biomarkers. Interaction between sirtuin1 and PKC-ζ was investigated by co-immunoprecipitation and confocal immunofluorescence microscopy. Sirtuin1 activation was enhanced by resveratrol treatment or Ad-sirtuin1 transfection. A model of cardiac hypertrophy in Sprague-Dawley rats was established by abdominal aortic constriction surgery or induced by isoprenaline in vivo. KEY RESULTS Overexpression of PKC-ζ led to cardiac hypertrophy and increased activity of NF-κB, ERK1/2 and ERK5, which was ameliorated by sirtuin1 overexpression. Enhancement of sirtuin1 activity suppressed acetylation of PKC-ζ, hindered its binding to phosphoinositide-dependent kinase 1 and inhibited PKC-ζ phosphorylation in cardiac hypertrophy. Consequently, the downstream pathways of PKC-ζ' were suppressed in cardiac hypertrophy. This regulation loop suggests a new role for sirtuin1 in mediation of cardiac hypertrophy. CONCLUSIONS AND IMPLICATIONS Sirtuin1 is an endogenous negative regulator for PKC-ζ and mediates its activity via regulating the acetylation and phosphorylation in the pathogenesis of cardiac hypertrophy. Targeting the sirtuin1-PKC-ζ signalling axis may suggest a novel therapeutic approach against cardiac hypertrophy.
Collapse
Affiliation(s)
- Jingyan Li
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, National and Local United Engineering Lab of Druggability and New Drugs EvaluationSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Junying Huang
- College of Life SciencesGuangzhou UniversityGuangzhouGuangdongChina
| | - Jing Lu
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, National and Local United Engineering Lab of Druggability and New Drugs EvaluationSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Zhen Guo
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, National and Local United Engineering Lab of Druggability and New Drugs EvaluationSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Zhuoming Li
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, National and Local United Engineering Lab of Druggability and New Drugs EvaluationSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Hui Gao
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, National and Local United Engineering Lab of Druggability and New Drugs EvaluationSun Yat‐sen UniversityGuangzhouGuangdongChina
- Department of Pharmacology, School of MedicineJishou UniversityJishouChina
| | - Panxia Wang
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, National and Local United Engineering Lab of Druggability and New Drugs EvaluationSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Wenwei Luo
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, National and Local United Engineering Lab of Druggability and New Drugs EvaluationSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Sidong Cai
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, National and Local United Engineering Lab of Druggability and New Drugs EvaluationSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Yuehuai Hu
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, National and Local United Engineering Lab of Druggability and New Drugs EvaluationSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Kaiteng Guo
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, National and Local United Engineering Lab of Druggability and New Drugs EvaluationSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Luping Wang
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, National and Local United Engineering Lab of Druggability and New Drugs EvaluationSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Zhenzhen Li
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, National and Local United Engineering Lab of Druggability and New Drugs EvaluationSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Minghui Wang
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, National and Local United Engineering Lab of Druggability and New Drugs EvaluationSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Xiaolei Zhang
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, National and Local United Engineering Lab of Druggability and New Drugs EvaluationSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Peiqing Liu
- Department of Pharmacology and Toxicology, School of Pharmaceutical Sciences, National and Local United Engineering Lab of Druggability and New Drugs EvaluationSun Yat‐sen UniversityGuangzhouGuangdongChina
| |
Collapse
|
1076
|
Bogdándi V, Ida T, Sutton TR, Bianco C, Ditrói T, Koster G, Henthorn HA, Minnion M, Toscano JP, van der Vliet A, Pluth MD, Feelisch M, Fukuto JM, Akaike T, Nagy P. Speciation of reactive sulfur species and their reactions with alkylating agents: do we have any clue about what is present inside the cell? Br J Pharmacol 2019; 176:646-670. [PMID: 29909607 PMCID: PMC6346080 DOI: 10.1111/bph.14394] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 05/18/2018] [Accepted: 06/05/2018] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND AND PURPOSE: Posttranslational modifications of cysteine residues represent a major aspect of redox biology, and their reliable detection is key in providing mechanistic insights. The metastable character of these modifications and cell lysis-induced artifactual oxidation render current state-of-the-art protocols to rely on alkylation-based stabilization of labile cysteine derivatives before cell/tissue rupture. An untested assumption in these procedures is that for all cysteine derivatives, alkylation rates are faster than their dynamic interchange. However, when the interconversion of cysteine derivatives is not rate limiting, electrophilic labelling is under Curtin-Hammett control; hence, the final alkylated mixture may not represent the speciation that prevailed before alkylation. EXPERIMENTAL APPROACH Buffered aqueous solutions of inorganic, organic, cysteine, GSH and GAPDH polysulfide species were used. Additional experiments in human plasma and serum revealed that monobromobimane can extract sulfide from the endogenous sulfur pool by shifting speciation equilibria, suggesting caution should be exercised when interpreting experimental results using this tool. KEY RESULTS In the majority of cases, the speciation of alkylated polysulfide/thiol derivatives depended on the experimental conditions. Alkylation perturbed sulfur speciation in both a concentration- and time-dependent manner and strong alkylating agents cleaved polysulfur chains. Moreover, the labelling of sulfenic acids with dimedone also affected cysteine speciation, suggesting that part of the endogenous pool of products previously believed to represent sulfenic acid species may represent polysulfides. CONCLUSIONS AND IMPLICATIONS We highlight methodological caveats potentially arising from these pitfalls and conclude that current derivatization strategies often fail to adequately capture physiological speciation of sulfur species. LINKED ARTICLES This article is part of a themed section on Chemical Biology of Reactive Sulfur Species. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.4/issuetoc.
Collapse
Affiliation(s)
- Virág Bogdándi
- Department of Molecular Immunology and ToxicologyNational Institute of OncologyBudapestHungary
| | - Tomoaki Ida
- Department of Environmental Medicine and Molecular ToxicologyTohoku University Graduate School of MedicineSendaiJapan
| | - Thomas R Sutton
- Clinical and Experimental Sciences, Faculty of MedicineUniversity Hospital Southampton NHS Foundation Trust, University of SouthamptonSouthamptonUK
| | | | - Tamás Ditrói
- Department of Molecular Immunology and ToxicologyNational Institute of OncologyBudapestHungary
| | - Grielof Koster
- Clinical and Experimental Sciences, Faculty of MedicineUniversity Hospital Southampton NHS Foundation Trust, University of SouthamptonSouthamptonUK
| | - Hillary A Henthorn
- Department of Chemistry and Biochemistry, Materials Science Institute, Institute of Molecular BiologyUniversity of OregonEugeneORUSA
| | - Magda Minnion
- Clinical and Experimental Sciences, Faculty of MedicineUniversity Hospital Southampton NHS Foundation Trust, University of SouthamptonSouthamptonUK
| | - John P Toscano
- Department of ChemistryJohns Hopkins UniversityBaltimoreMDUSA
| | - Albert van der Vliet
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of MedicineUniversity of VermontBurlingtonVTUSA
| | - Michael D Pluth
- Department of Chemistry and Biochemistry, Materials Science Institute, Institute of Molecular BiologyUniversity of OregonEugeneORUSA
| | - Martin Feelisch
- Clinical and Experimental Sciences, Faculty of MedicineUniversity Hospital Southampton NHS Foundation Trust, University of SouthamptonSouthamptonUK
| | - Jon M Fukuto
- Department of ChemistrySonoma State UniversityRohnert ParkCAUSA
| | - Takaaki Akaike
- Department of Environmental Medicine and Molecular ToxicologyTohoku University Graduate School of MedicineSendaiJapan
| | - Péter Nagy
- Department of Molecular Immunology and ToxicologyNational Institute of OncologyBudapestHungary
| |
Collapse
|
1077
|
Masuoka T, Uwada J, Kudo M, Yoshiki H, Yamashita Y, Taniguchi T, Nishio M, Ishibashi T, Muramatsu I. Augmentation of Endogenous Acetylcholine Uptake and Cholinergic Facilitation of Hippocampal Long-Term Potentiation by Acetylcholinesterase Inhibition. Neuroscience 2019; 404:39-47. [PMID: 30708046 DOI: 10.1016/j.neuroscience.2019.01.042] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 01/21/2019] [Accepted: 01/22/2019] [Indexed: 10/27/2022]
Abstract
Hippocampal cholinergic activity enhances long-term potentiation (LTP) of synaptic transmission in intrahippocampal circuits and regulates cognitive function. We recently demonstrated intracellular distribution of functional M1-muscarinic acetylcholine receptors (mAChRs) and neuronal uptake of acetylcholine (ACh) in the central nervous system. Here we examined whether endogenous ACh acts on intracellular M1-mAChRs following its uptake and causes cholinergic facilitation of hippocampal LTP. ACh esterase (AChE) activities and [3H]ACh uptake was measured in rat hippocampal segments. LTP of evoked field excitatory postsynaptic potentials at CA1 synapses was induced by high frequency stimulation in hippocampal slices. Pretreatment with diisopropylfluorophosphate (DFP) irreversibly inhibited AChE, augmented ACh uptake, and significantly enhanced the LTP. This cholinergic facilitation was inhibited by pirenzepine, a membrane-permeable M1 antagonist, while only the early stage of cholinergic facilitation was inhibited by a membrane-impermeable M1 antagonist, muscarinic toxin 7. Tetraethylammonium (TEA) inhibited ACh uptake in hippocampal segments and selectively suppressed late stage cholinergic facilitation without changing the early stage. In contrast, LTP in DFP-untreated slices was not affected by the muscarinic antagonists and TEA. Carbachol (CCh; an AChE-resistant muscarinic agonist) competed with ACh for its uptake and produced cholinergic facilitation of LTP in DFP-untreated slices. The late stage of CCh-induced facilitation was also selectively inhibited by TEA. Our results suggest that when AChE is inactivated by inhibitors, LTP in hippocampal slices is significantly enhanced by endogenous ACh and that cholinergic facilitation is caused by direct activation of cell-surface M1-mAChRs and subsequent activation of intracellular M1-mAChRs after ACh uptake.
Collapse
Affiliation(s)
- Takayoshi Masuoka
- Department of Pharmacology, School of Medicine, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Ishikawa, 920-0293, Japan.
| | - Junsuke Uwada
- Division of Cellular Signal Transduction, Department of Biochemistry, Asahikawa Medical University, 2-1-1-1, Midorigaoka-Higashi, Asahikawa, Hokkaido, 078-8510, Japan
| | - Makiko Kudo
- Department of Pharmacology, School of Medicine, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Ishikawa, 920-0293, Japan
| | - Hatsumi Yoshiki
- Division of Genomic Science and Microbiology, School of Medicine, University of Fukui, Eiheiji, Fukui 910-1193, Japan
| | - Yuka Yamashita
- Department of Pharmacology, School of Medicine, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Ishikawa, 920-0293, Japan
| | - Takanobu Taniguchi
- Division of Cellular Signal Transduction, Department of Biochemistry, Asahikawa Medical University, 2-1-1-1, Midorigaoka-Higashi, Asahikawa, Hokkaido, 078-8510, Japan
| | - Matomo Nishio
- Department of Pharmacology, School of Medicine, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Ishikawa, 920-0293, Japan
| | - Takaharu Ishibashi
- Department of Pharmacology, School of Medicine, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Ishikawa, 920-0293, Japan
| | - Ikunobu Muramatsu
- Department of Pharmacology, School of Medicine, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Ishikawa, 920-0293, Japan
| |
Collapse
|
1078
|
Chakraborty AD, Gonano LA, Munro ML, Smith LJ, Thekkedam C, Staudacher V, Gamble AB, Macquaide N, Dulhunty AF, Jones PP. Activation of RyR2 by class I kinase inhibitors. Br J Pharmacol 2019; 176:773-786. [PMID: 30588601 DOI: 10.1111/bph.14562] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 11/26/2018] [Accepted: 12/09/2018] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Kinase inhibitors are a common treatment for cancer. Class I kinase inhibitors that target the ATP-binding pocket are particularly prevalent. Many of these compounds are cardiotoxic and can cause arrhythmias. Spontaneous release of Ca2+ via cardiac ryanodine receptors (RyR2), through a process termed store overload-induced Ca2+ release (SOICR), is a common mechanism underlying arrhythmia. We explored whether class I kinase inhibitors could modify the activity of RyR2 and trigger SOICR to determine if this contributes to the cardiotoxic nature of these compounds. EXPERIMENTAL APPROACH The impact of class I and II kinase inhibitors on SOICR was studied in HEK293 cells and ventricular myocytes using single-cell Ca2+ imaging. A specific effect on RyR2 was confirmed using single channel recordings. Ventricular myocytes were also used to determine if drug-induced changes in SOICR could be reversed using anti-SOICR agents. KEY RESULTS Class I kinase inhibitors increased the propensity of SOICR. Single channel recording showed that this was due to a specific effect on RyR2. Class II kinase inhibitors decreased the activity of RyR2 at the single channel level but had little effect on SOICR. The promotion of SOICR mediated by class I kinase inhibitors could be reversed using the anti-SOICR agent VK-II-86. CONCLUSIONS AND IMPLICATIONS Part of the cardiotoxicity of class I kinase inhibitors can be assigned to their effect on RyR2 and increase in SOICR. Compounds with anti-SOICR activity may represent an improved treatment option for patients.
Collapse
Affiliation(s)
- A D Chakraborty
- Department of Physiology, School of Biomedical Sciences, and HeartOtago, University of Otago, Dunedin, New Zealand
| | - L A Gonano
- Department of Physiology, School of Biomedical Sciences, and HeartOtago, University of Otago, Dunedin, New Zealand.,Centro de Investigaciones Cardiovasculares, CONICET La Plata, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, La Plata, Argentina
| | - M L Munro
- Department of Physiology, School of Biomedical Sciences, and HeartOtago, University of Otago, Dunedin, New Zealand
| | - L J Smith
- Department of Physiology, School of Biomedical Sciences, and HeartOtago, University of Otago, Dunedin, New Zealand
| | - C Thekkedam
- Eccles Institute of Neuroscience, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - V Staudacher
- School of Pharmacy, University of Otago, Dunedin, New Zealand
| | - A B Gamble
- School of Pharmacy, University of Otago, Dunedin, New Zealand
| | - N Macquaide
- Institute of Cardiovascular and Medical Sciences and School of Life Sciences, College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, UK
| | - A F Dulhunty
- Eccles Institute of Neuroscience, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - P P Jones
- Department of Physiology, School of Biomedical Sciences, and HeartOtago, University of Otago, Dunedin, New Zealand
| |
Collapse
|
1079
|
Li T, Yang S, She X, Yan Q, Zhang P, Zhu H, Wang F, Luo X, Sun X. Modulation of α-adrenoceptor signalling protects photoreceptors after retinal detachment by inhibiting oxidative stress and inflammation. Br J Pharmacol 2019; 176:801-813. [PMID: 30588605 DOI: 10.1111/bph.14565] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 11/20/2018] [Accepted: 11/29/2018] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND AND PURPOSE Currently available treatments do not halt progression of photoreceptor death and subsequent visual impairment related to retinal detachment (RD) which is observed in various retinal disorders. This study investigated the neuroprotective effects of two adrenoceptor ligands, the α1 -adrenoceptor antagonist doxazosin and the α2 -adrenoceptor agonist guanabenz, against photoreceptor cell death in RD. EXPERIMENTAL APPROACH We used a model of experimental RD in Brown-Norway rats induced by subretinal injection of sodium hyaluronate. Oxidative stress biomarkers and cytokine production were quantified with elisa. Protein expression levels and immunofluorescent labelling were determined in rats with RD and controls for mechanistic elucidation. The effects of systemic (i.p.) administration of doxazosin or guanabenz on photoreceptor apoptosis, retinal histology and electroretinography were evaluated in rats with RD and compared to the effects in vehicle controls. KEY RESULTS Photoreceptors were the major source of RD-induced ROS overproduction in the rat retina through the regulation of NADPH oxidase. Systemic administration of doxazosin or guanabenz decreased the RD-induced production of ROS and proinflammatory cytokines, including IL-1β and the chemokine CCL2, and suppressed retinal gliosis, resulting in attenuation of photoreceptor death and preservation of retinal structures and functions in RD. CONCLUSIONS AND IMPLICATIONS Our findings point to α-adrenoceptors as novel therapeutic targets to provide photoreceptor protection and suggest that both doxazosin and guanabenz, two FDA-approved drugs, could be further explored to treat retinal diseases.
Collapse
Affiliation(s)
- Tong Li
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shiqi Yang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiangjun She
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Quan Yan
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Pengfei Zhang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong Zhu
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China
| | - Fenghua Wang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China
| | - Xueting Luo
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Fundus Diseases, Shanghai, China
| | - Xiaodong Sun
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Fundus Diseases, Shanghai, China.,Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, China
| |
Collapse
|
1080
|
Attili B, Celen S, Ahamed M, Koole M, Haute CVD, Vanduffel W, Bormans G. Preclinical evaluation of [ 18 F]MA3: a CB 2 receptor agonist radiotracer for PET. Br J Pharmacol 2019; 176:1481-1491. [PMID: 30588600 DOI: 10.1111/bph.14564] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 10/31/2018] [Accepted: 11/16/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND AND PURPOSE Non-invasive in vivo imaging of cannabinoid CB2 receptors using PET is pursued to study neuroinflammation. The purpose of this study is to evaluate the in vivo binding specificity of [18 F]MA3, a CB2 receptor agonist, in a rat model with local overexpression of human (h) CB2 receptors. METHODS [18 F]MA3 was produced with good radiochemical yield and radiochemical purity. The radiotracer was evaluated in rats with local overexpression of hCB2 receptors and in a healthy non-human primate using PET. KEY RESULTS Ex vivo autoradiography demonstrated CB2 -specific binding of [18 F]MA3 in rat hCB2 receptor vector injected striatum. In a PET study, increased tracer binding in the hCB2 receptor vector-injected striatum compared to the contralateral control vector-injected striatum was observed. Binding in hCB2 receptor vector-injected striatum was blocked with a structurally non-related CB2 receptor inverse agonist, and a displacement study confirmed the reversibility of tracer binding. This study identified the utility of mutated inactive vector model for evaluation of CB2 receptor agonist PET tracers. [18 F]MA3 PET scans in the non-human primate showed good uptake and fast washout from brain, but no CB2 receptor-specific binding was observed. CONCLUSION AND IMPLICATIONS Evaluation of [18 F]MA3 in a rat model with local overexpression of hCB2 receptors showed CB2 receptor-specific and reversible tracer binding. [18 F]MA3 showed good brain uptake and subsequent washout in a healthy non-human primate, but no specific binding was observed. Further clinical evaluation of [18 F]MA3 in patients with neuroinflammation is warranted. LINKED ARTICLES This article is part of a themed section on 8th European Workshop on Cannabinoid Research. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.10/issuetoc.
Collapse
Affiliation(s)
- Bala Attili
- Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Sofie Celen
- Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Muneer Ahamed
- Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Michel Koole
- Department of Nuclear Medicine and Molecular Imaging, UZ Gasthuisberg, Leuven, Belgium
| | - Chris Van Den Haute
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven, Leuven, Belgium.,Leuven Viral Vector Core, Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Wim Vanduffel
- Laboratory for Neuro- and Psychophysiology, Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Guy Bormans
- Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
1081
|
Lian X, Wu X, Li Z, Zhang Y, Song K, Cai G, Li Q, Lin S, Chen X, Bai XY. The combination of metformin and 2-deoxyglucose significantly inhibits cyst formation in miniature pigs with polycystic kidney disease. Br J Pharmacol 2019; 176:711-724. [PMID: 30515768 DOI: 10.1111/bph.14558] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Revised: 10/25/2018] [Accepted: 11/10/2018] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND AND PURPOSE The pathogenic mechanism of autosomal dominant polycystic kidney disease (ADPKD) is unclear. Similar to tumour cells, polycystic kidney cells are primarily dependent on aerobic glycolysis for ATP production. Compared with rodents, miniature pigs are more similar to humans. This study is the first time to investigate the effects of the combination of metformin and 2-deoxyglucose (2DG) in a pig model of chronic progressive ADPKD. EXPERIMENTAL APPROACH A miniature pig ADPKD model was established by inducible deletion of the PKD1 gene. Blood, urine and kidney biopsy specimens were collected for analysis at specific times. The renal vesicle index was analysed by three-dimensional reconstruction of CT scans. Markers of the mammalian target of rapamycin (mTOR) and ERK signalling pathways and associated metabolism were detected by Western blots and colorimetry. KEY RESULTS The three-dimensional reconstruction of CT scans indicated a markedly lower renal vesicle index in the combination therapy group. Each drug intervention group showed a significantly lower serum creatinine and urinary protein/creatinine ratio. This treatment regimen also inhibited the activities of markers of the proliferation-related mTOR and ERK pathways, and the expression of key enzymes involved in glycolysis, as well as reducing the production of ATP and lactic acid. CONCLUSIONS AND IMPLICATIONS This study showed that the combination of metformin and 2DG blocked the formation of renal cysts and improved the renal function in ADPKD miniature pigs. Our results indicate that the combination of metformin and 2DG may be a promising therapeutic strategy in human ADPKD.
Collapse
Affiliation(s)
- Xiaoying Lian
- Department of Nephrology, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, China.,Department of Nephrology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Xiaoyuan Wu
- Department of Nephrology, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, China.,Department of Nephrology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhongxin Li
- Department of Nephrology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Yingjie Zhang
- Department of Nephrology, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, China
| | - Kangkang Song
- Department of Nephrology, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, China
| | - Guangyan Cai
- Department of Nephrology, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, China
| | - Qinggang Li
- Department of Nephrology, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, China
| | - Shupeng Lin
- Department of Nephrology, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, China
| | - Xiangmei Chen
- Department of Nephrology, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, China
| | - Xue-Yuan Bai
- Department of Nephrology, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
1082
|
An W, Yu Y, Zhang Y, Zhang Z, Yu Y, Zhao X. Exogenous IL-19 attenuates acute ischaemic injury and improves survival in male mice with myocardial infarction. Br J Pharmacol 2019; 176:699-710. [PMID: 30460984 DOI: 10.1111/bph.14549] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 10/21/2018] [Accepted: 10/24/2018] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND AND PURPOSE Myocardial infarction (MI) is one of the leading causes of death in China and often results in the development of heart failure. In this work, we tested the therapeutic role of Interleukin-19 (IL-19) in mice with MI and investigated the underlying molecular mechanism. EXPERIMENTAL APPROACH Mice were subjected to MI by ligation of left anterior descending coronary artery (LAD) and treated with IL-19 (10ng g-1 ; i.p.). KEY RESULTS Protein expression of IL-19 and its receptor in myocardium were upregulated 24 hrs post-MI in male mice. IL-19 treatment decreased infarct and apoptosis in myocardium, accompanied by enhanced haem oxygenase-1 (HO-1) activities and reduced malondialdehyde (MDA) formation. Pretreatment with IL-19 upregulated HO-1 expression in cultured neonatal mouse ventricular myocytes and attenuated oxygen-glucose deprivation (OGD)-induced injuries in vitro. Furthermore, IL-19 preserved cardiac function and improved survival of mice with MI. IL-19 reduced inflammatory infiltrates and suppressed formation of TNF-α, IL-1β, and IL-6. More importantly, IL-19 inhibited polarization toward proinflammatory M1 macrophages and stimulated M2 macrophage polarization in myocardium of mice with MI. IL-19 enhanced protein levels of vascular endothelial growth factor (VEGF) and promoted angiogenesis in myocardium of mice with MI. In addition, IL-19 treatment increased DNA-binding of the transcription factor STAT3 in myocardium of mice with MI. CONCLUSIONS AND IMPLICATIONS Treatment with exogenous IL-19 attenuated acute ischemic injury and improved survival of mice with MI. The mechanisms underlying these effects involved induction of HO-1, M2 macrophage polarization, angiogenesis, and STAT3 activation.
Collapse
Affiliation(s)
- Weishuai An
- Department of Cardiovasology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Yongsheng Yu
- Department of Cardiovasology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Yuefan Zhang
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Zhigang Zhang
- Department of Cardiology, Putuo Center Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yunhua Yu
- Department of Geriatrics, Fuzhou General Hospital, Fujian Medical University, Fuzhou, China
| | - Xianxian Zhao
- Department of Cardiovasology, Changhai Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|
1083
|
Coleman RA, Woodrooffe AJ, Clark KL, Toris CB, Fan S, Wang JW, Woodward DF. The affinity, intrinsic activity and selectivity of a structurally novel EP 2 receptor agonist at human prostanoid receptors. Br J Pharmacol 2019; 176:687-698. [PMID: 30341781 PMCID: PMC6365485 DOI: 10.1111/bph.14525] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Revised: 09/14/2018] [Accepted: 09/26/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND AND PURPOSE Prostanoid EP2 receptor agonists exhibit several activities including ocular hypotension, tocolysis and anti-inflammatory activity. This report describes the affinity and selectivity of a structurally novel, non-prostanoid EP2 receptor agonist, PGN-9856, and its therapeutic potential. EXPERIMENTAL APPROACH The pharmacology of a series of non-prostanoid EP2 receptor agonists was determined according to functional and radioligand binding studies, mostly using human recombinant prostanoid receptor transfectants. The selectivity of PGN-9856, as the preferred compound, was subsequently determined by using a diverse variety of non-prostanoid target proteins. The therapeutic potential of PGN-9856 was addressed by determining its activity in relevant primate cell, tissue and disease models. KEY RESULTS PGN-9856 was a selective and high affinity (pKi ≥ 8.3) ligand at human recombinant EP2 receptors. In addition to high affinity binding, it was a potent and full EP2 receptor agonist with a high level of selectivity at EP1 , EP3 , EP4 , DP, FP, IP and TP receptors. In cells overexpressing human recombinant EP2 receptors, PGN-9856 displayed a potency (pEC50 ≥ 8.5) and a maximal response (increase in cAMP) comparable to that of the endogenous agonist PGE2 . PGN-9856 exhibited no appreciable affinity (up 10 μM) for a range of 53 other receptors, ion channels and enzymes. Finally, PGN-9856 exhibited tocolytic, anti-inflammatory and long-acting ocular hypotensive properties consistent with its potent EP2 receptor agonist properties. CONCLUSIONS AND IMPLICATIONS PGN-9856 is a potent, selective and efficacious prostanoid EP2 receptor agonist with diverse potential therapeutic applications: tocolytic, anti-inflammatory and notably anti-glaucoma.
Collapse
Affiliation(s)
| | | | | | - C B Toris
- University of Nebraska Medical Center, Omaha, NE, USA
| | - S Fan
- University of Nebraska Medical Center, Omaha, NE, USA
| | - J W Wang
- JeniVision Inc., Irvine, CA, USA
| | | |
Collapse
|
1084
|
Feng L, Sun Y, Song P, Xu L, Wu X, Wu X, Shen Y, Sun Y, Kong L, Wu X, Xu Q. Seselin ameliorates inflammation via targeting Jak2 to suppress the proinflammatory phenotype of macrophages. Br J Pharmacol 2019; 176:317-333. [PMID: 30338847 PMCID: PMC6295420 DOI: 10.1111/bph.14521] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 08/23/2018] [Accepted: 09/24/2018] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND AND PURPOSE Sepsis is a serious clinical condition with a high mortality rate. Anti inflammatory agents have been found to be beneficial for the treatment of sepsis. Here, we have evaluated the anti-inflammatory activity of seselin in models of sepsis and investigated the underlying molecular mechanism(s). EXPERIMENTAL APPROACH In vivo therapeutic effects of seselin was evaluated in two models of sepsis, caecal ligation and puncture or injection of LPS, in C57BL/6 mice. In vitro, anti-inflammatory activity of seselin was assessed with macrophages stimulated with LPS and IFN-γ. Anti inflammatory actions were analysed with immunohistochemical methods, ELISA and Western blotting. Flow cytometry was used to assess markers of macrophage phenotype (pro- or anti-inflammatory). Other methods used included co-immunoprecipitation, cellular thermal shift assay and molecular docking. KEY RESULTS In vivo, seselin clearly ameliorated sepsis induced by caecal ligation and puncture. In lung tissue from septic mice and in cultured macrophages, seselin down-regulated levels of proinflammatory factors and activity of STAT1 and p65, the master signal pathway molecules for polarization of macrophages into the proinflammatory phenotype. Importantly, adoptive transfer of bone marrow-derived macrophages, pretreated with seselin, lowered systemic proinflammatory factors in mice challenged with LPS. The underlying mechanism was that seselin targeted Jak2 to block interaction with IFNγ receptors and downstream STAT1. CONCLUSIONS AND IMPLICATIONS Seselin exhibited anti-inflammatory activity through its action on Jak2. These results indicated a possible application of seselin to the treatment of inflammatory disease via blocking the development of the proinflammatory phenotype of macrophages.
Collapse
Affiliation(s)
- Lili Feng
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Life SciencesNanjing UniversityNanjingChina
| | - Yi Sun
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Life SciencesNanjing UniversityNanjingChina
| | - Pingping Song
- Jiangsu Centre for Research and Development of Medicinal Plants, Institute of Botany Jiangsu ProvinceChinese Academy of SciencesNanjingChina
| | - Lisha Xu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Life SciencesNanjing UniversityNanjingChina
| | - Xingxin Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Life SciencesNanjing UniversityNanjingChina
| | - Xuefeng Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Life SciencesNanjing UniversityNanjingChina
| | - Yan Shen
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Life SciencesNanjing UniversityNanjingChina
| | - Yang Sun
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Life SciencesNanjing UniversityNanjingChina
| | - Lingdong Kong
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Life SciencesNanjing UniversityNanjingChina
| | - Xudong Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Life SciencesNanjing UniversityNanjingChina
| | - Qiang Xu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Life SciencesNanjing UniversityNanjingChina
| |
Collapse
|
1085
|
Huang Z, Wang L, Wang J, Feng W, Yang Z, Ni S, Huang Y, Li H, Yang Y, Wang M, Hu R, Wan H, Wen C, Xian S, Lu L. Hispaglabridin B, a constituent of liquorice identified by a bioinformatics and machine learning approach, relieves protein-energy wasting by inhibiting forkhead box O1. Br J Pharmacol 2019; 176:267-281. [PMID: 30270561 PMCID: PMC6295407 DOI: 10.1111/bph.14508] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 08/22/2018] [Accepted: 08/26/2018] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND AND PURPOSE Liquorice is the root of Glycyrrhiza glabra, which is a popular food in Europe and China that has previously shown benefits for skeletal fatigue and nutrient metabolism. However, the mechanism and active ingredients remain largely unclear. The aim of this study was to investigate the active ingredients of liquorice for muscle wasting and elucidate the underlying mechanisms. EXPERIMENTAL APPROACH RNA-Seq and bioinformatics analysis were applied to predict the main target of liquorice. A machine learning model and a docking tool were used to predict active ingredients. Isotope labelling experiments, immunostaining, Western blots, qRT-PCR, ChIP-PCR and luciferase reporters were utilized to test the pharmacological effects in vitro and in vivo. The reverse effects were verified through recombination-based overexpression. KEY RESULTS The liposoluble constituents of liquorice improved muscle wasting by inhibiting protein catabolism and fibre atrophy. We further identified FoxO1 as the target of liposoluble constituents of liquorice. In addition, hispaglabridin B (HB) was predicted as an inhibitor of FoxO1. Further studies determined that HB improved muscle wasting by inhibiting catabolism in vivo and in vitro. HB also markedly suppressed the transcriptional activity of FoxO1, with decreased expression of the muscle-specific E3 ubiquitin ligases MuRF1 and Atrogin-1. CONCLUSIONS AND IMPLICATIONS HB can serve as a novel natural food extract for preventing muscle wasting in chronic kidney disease and possibly other catabolic conditions.
Collapse
Affiliation(s)
- Zeng‐Yan Huang
- The First Affiliated HospitalGuangzhou University of Chinese MedicineGuangzhouChina
- Lingnan Medical Research CenterGuangzhou University of Chinese MedicineGuangzhouChina
| | - Ling‐Jun Wang
- The First Affiliated HospitalGuangzhou University of Chinese MedicineGuangzhouChina
- Lingnan Medical Research CenterGuangzhou University of Chinese MedicineGuangzhouChina
| | - Jia‐Jia Wang
- The First Affiliated HospitalGuangzhou University of Chinese MedicineGuangzhouChina
- Lingnan Medical Research CenterGuangzhou University of Chinese MedicineGuangzhouChina
| | - Wen‐Jun Feng
- The First Affiliated HospitalGuangzhou University of Chinese MedicineGuangzhouChina
| | - Zhong‐Qi Yang
- The First Affiliated HospitalGuangzhou University of Chinese MedicineGuangzhouChina
- Lingnan Medical Research CenterGuangzhou University of Chinese MedicineGuangzhouChina
| | - Shi‐Hao Ni
- The First Affiliated HospitalGuangzhou University of Chinese MedicineGuangzhouChina
- Lingnan Medical Research CenterGuangzhou University of Chinese MedicineGuangzhouChina
| | - Yu‐Sheng Huang
- The First Affiliated HospitalGuangzhou University of Chinese MedicineGuangzhouChina
- Lingnan Medical Research CenterGuangzhou University of Chinese MedicineGuangzhouChina
| | - Huan Li
- The First Affiliated HospitalGuangzhou University of Chinese MedicineGuangzhouChina
- Lingnan Medical Research CenterGuangzhou University of Chinese MedicineGuangzhouChina
| | - Yi Yang
- The First Affiliated HospitalGuangzhou University of Chinese MedicineGuangzhouChina
- Lingnan Medical Research CenterGuangzhou University of Chinese MedicineGuangzhouChina
| | - Ming‐Qing Wang
- School of Traditional Chinese MedicineSouthern Medical UniversityGuangzhouChina
- Peninsula School of MedicineUniversity of PlymouthPlymouthUK
| | - Rong Hu
- School of Traditional Chinese MedicineSouthern Medical UniversityGuangzhouChina
| | - Heng Wan
- Department of EndocrinologyThe Third Affiliated Hospital of Southern Medical UniversityGuangzhouChina
| | - Chan‐Juan Wen
- Department of RadiologyNan Fang Hospital of Southern Medical UniversityGuangzhouChina
| | - Shao‐Xiang Xian
- The First Affiliated HospitalGuangzhou University of Chinese MedicineGuangzhouChina
- Lingnan Medical Research CenterGuangzhou University of Chinese MedicineGuangzhouChina
| | - Lu Lu
- The First Affiliated HospitalGuangzhou University of Chinese MedicineGuangzhouChina
- Lingnan Medical Research CenterGuangzhou University of Chinese MedicineGuangzhouChina
| |
Collapse
|
1086
|
Broad LM, Sanger HE, Mogg AJ, Colvin EM, Zwart R, Evans DA, Pasqui F, Sher E, Wishart GN, Barth VN, Felder CC, Goldsmith PJ. Identification and pharmacological profile of SPP1, a potent, functionally selective and brain penetrant agonist at muscarinic M 1 receptors. Br J Pharmacol 2019; 176:110-126. [PMID: 30276808 PMCID: PMC6284335 DOI: 10.1111/bph.14510] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 09/14/2018] [Accepted: 09/18/2018] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE We aimed to identify and develop novel, selective muscarinic M1 receptor agonists as potential therapeutic agents for the symptomatic treatment of Alzheimer's disease. EXPERIMENTAL APPROACH We developed and utilized a novel M1 receptor occupancy assay to drive a structure activity relationship in a relevant brain region while simultaneously tracking drug levels in plasma and brain to optimize for central penetration. Functional activity was tracked in relevant native in vitro assays allowing translational (rat-human) benchmarking of structure-activity relationship molecules to clinical comparators. KEY RESULTS Using this paradigm, we identified a series of M1 receptor selective molecules displaying desirable in vitro and in vivo properties and optimized key features, such as central penetration while maintaining selectivity and a partial agonist profile. From these compounds, we selected spiropiperidine 1 (SPP1). In vitro, SPP1 is a potent, partial agonist of cortical and hippocampal M1 receptors with activity conserved across species. SPP1 displays high functional selectivity for M1 receptors over native M2 and M3 receptor anti-targets and over a panel of other targets. Assessment of central target engagement by receptor occupancy reveals SPP1 significantly and dose-dependently occupies rodent cortical M1 receptors. CONCLUSIONS AND IMPLICATIONS We report the discovery of SPP1, a novel, functionally selective, brain penetrant partial orthosteric agonist at M1 receptors, identified by a novel receptor occupancy assay. SPP1 is amenable to in vitro and in vivo study and provides a valuable research tool to further probe the role of M1 receptors in physiology and disease.
Collapse
Affiliation(s)
- Lisa M Broad
- Eli Lilly and Company, Lilly Research CentreWindleshamSurreyUK
| | - Helen E Sanger
- Eli Lilly and Company, Lilly Research CentreWindleshamSurreyUK
| | - Adrian J Mogg
- Eli Lilly and Company, Lilly Research CentreWindleshamSurreyUK
| | - Ellen M Colvin
- Eli Lilly and Company, Lilly Research CentreWindleshamSurreyUK
| | - Ruud Zwart
- Eli Lilly and Company, Lilly Research CentreWindleshamSurreyUK
| | - David A Evans
- Eli Lilly and Company, Lilly Research CentreWindleshamSurreyUK
| | | | - Emanuele Sher
- Eli Lilly and Company, Lilly Research CentreWindleshamSurreyUK
| | | | - Vanessa N Barth
- Eli Lilly and Company, Lilly Corporate CenterIndianapolisINUSA
| | | | | |
Collapse
|
1087
|
Heblinski M, Bladen C, Connor M. Regulation of heterologously expressed 5-HT 1B receptors coupling to potassium channels in AtT-20 cells. Br J Pharmacol 2018; 176:451-465. [PMID: 30447001 DOI: 10.1111/bph.14547] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 10/08/2018] [Accepted: 10/17/2018] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND AND PURPOSE 5-HT1B receptors are widely expressed GPCRs and a target of triptans, the most commonly prescribed anti-migraine drugs. There is very limited information about the acute, agonist-induced regulation of 5-HT1B receptor signalling and so we sought to characterize this in a neuron-like system. EXPERIMENTAL APPROACH Epitope-tagged human 5-HT1B receptors were expressed in mouse AtT20 cells. 5-HT1B receptor signalling was assessed using whole-cell patch-clamp recordings of endogenous G protein-gated inwardly rectified potassium (GIRK) channels, and receptor localization measured using immunofluorescence. KEY RESULTS 5-HT (EC50 65 nM) and sumatriptan (EC50 165 nM) activated GIRK channels in AtT20 cells expressing 5-HT1B receptors. Continuous application of both 5-HT (EC50 120 nM) and sumatriptan (EC50 280 nM) produced profound desensitization of 5-HT1B receptor signalling within a few minutes. Complete recovery from desensitization was observed after 10 min. Both 5-HT and sumatriptan induced significant heterologous desensitization of SRIF (somatostatin)-activated GIRK currents, with the 5-HT-induced heterologous desensitization being blocked by the protein kinase inhibitor staurosporine. Both agonists induced modest 5-HT1B receptor internalization, with a time course much slower than receptor desensitization. CONCLUSIONS AND IMPLICATIONS In AtT-20 cells, 5-HT1B receptors undergo rapid and reversible desensitization at concentrations of agonist similar to those required to activate the receptor. Desensitization is incomplete, and the continued signalling of the receptor in the presence of the agonist may lead to cellular adaptations. Finally, 5-HT1B receptor activation causes significant heterologous desensitization, which may lead to a reduced effectiveness of unrelated drugs in vivo.
Collapse
Affiliation(s)
- Marika Heblinski
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Christopher Bladen
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Mark Connor
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| |
Collapse
|
1088
|
Dekker SJ, Dohmen F, Vermeulen NPE, Commandeur JNM. Characterization of kinetics of human cytochrome P450s involved in bioactivation of flucloxacillin: inhibition of CYP3A-catalysed hydroxylation by sulfaphenazole. Br J Pharmacol 2018; 176:466-477. [PMID: 30447161 PMCID: PMC6329626 DOI: 10.1111/bph.14548] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 10/19/2018] [Accepted: 10/27/2018] [Indexed: 01/18/2023] Open
Abstract
Background and Purpose The aim of this study was to characterize the human cytochrome P450s (CYPs) involved in oxidative bioactivation of flucloxacillin to 5‐hydroxymethyl flucloxacillin, a metabolite with high cytotoxicity towards biliary epithelial cells. Experimental Approach The CYPs involved in hydroxylation of flucloxacillin were characterized using recombinant human CYPs, pooled liver microsomes in the presence of CYP‐specific inhibitors and by correlation analysis using a panel of liver microsomes from 16 donors. Key Results Recombinant CYPs showing the highest specific activity were CYP3A4, CYP3A7 and to lower extent CYP2C9 and CTP2C8. Michaelis–Menten enzyme kinetics were determined for pooled human liver microsomes, recombinant CYP3A4, CYP3A7 and CYP2C9. Surprisingly, sulfaphenazole appeared to be a potent inhibitor of 5′‐hydroxylation of flucloxacillin by both recombinant CYP3A4 and CYP3A7. Conclusions and Implications The combined results show that the 5′‐hydroxylation of flucloxacillin is primarily catalysed by CYP3A4, CYP3A7 and CYP2C9. The large variability of the hepatic expression of these enzymes could affect the formation of 5′‐hydroxymethyl flucloxacillin, which may determine the differences in susceptibility to flucloxacillin‐induced liver injury. Additionally, the strong inhibition in CYP3A‐catalysed flucloxacillin metabolism by sulfaphenazole suggests that unanticipated drug–drug interactions could occur with coadministered drugs.
Collapse
Affiliation(s)
- Stefan J Dekker
- Division of Molecular Toxicology, Amsterdam Institute for Molecules Medicine and Systems (AIMMS), Vrije Universiteit, Amsterdam, The Netherlands
| | - Floor Dohmen
- Division of Molecular Toxicology, Amsterdam Institute for Molecules Medicine and Systems (AIMMS), Vrije Universiteit, Amsterdam, The Netherlands
| | - Nico P E Vermeulen
- Division of Molecular Toxicology, Amsterdam Institute for Molecules Medicine and Systems (AIMMS), Vrije Universiteit, Amsterdam, The Netherlands
| | - Jan N M Commandeur
- Division of Molecular Toxicology, Amsterdam Institute for Molecules Medicine and Systems (AIMMS), Vrije Universiteit, Amsterdam, The Netherlands
| |
Collapse
|
1089
|
Zhou X, Zhang R, Zou Z, Shen X, Xie T, Xu C, Dong J, Liao L. Hypoglycaemic effects of glimepiride in sulfonylurea receptor 1 deficient rat. Br J Pharmacol 2018; 176:478-490. [PMID: 30471094 DOI: 10.1111/bph.14553] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 10/30/2018] [Accepted: 11/02/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND AND PURPOSE Sulfonylureas (SUs) have been suggested to have an insulin-independent blood glucose-decreasing activity due to an extrapancreatic effect. However, a lack of adequate in vivo evidence makes this statement controversial. Here, we aimed to evaluate whether glimepiride has extrapancreatic blood glucose-lowering activity in vivo. EXPERIMENTAL APPROACH Sulfonylurea receptor 1 deficient (SUR1-/- ) rats were created by means of transcription activator-like effector nucleases (TALEN)-mediated gene targeting technology. Type 2 diabetic models were established by feeding a high-fat diet and administering a low-dose of streptozotocin. These rats were then randomly divided into four groups: glimepiride, gliclazide, metformin and saline. All rats were treated for 2 weeks. KEY RESULTS Glimepiride decreased blood glucose levels and increased insulin sensitivity without elevating insulin levels. Gliclazide showed similar effects as glimepiride. Both agents were weaker than metformin. Further mechanistic investigations revealed that glimepiride increased hepatic glycogen synthesis and decreased gluconeogenesis, which were accompanied by the activation of Akt in the liver. Moreover, glimepiride increased both total and membrane glucose transporter 4 (GLUT4) levels in muscle and fat, which might be attributed to insulin receptor-independent IRS1/Akt activation. CONCLUSION AND IMPLICATIONS Glimepiride possesses an extrapancreatic blood glucose-lowering effect in vivo, which might be attributed to its direct effect on insulin-sensitive tissues. Therefore, the combination of glimepiride with multiple insulin injections should not be excluded per se.
Collapse
Affiliation(s)
- Xiaojun Zhou
- Department of Endocrinology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Rui Zhang
- Department of Endocrinology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Zhiwei Zou
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, China
| | - Xue Shen
- Department of Endocrinology, Shandong Provincial Qianfoshan Hospital, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Tianyue Xie
- Department of Endocrinology, Shandong Provincial Qianfoshan Hospital, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chunmei Xu
- Department of Endocrinology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Jianjun Dong
- Department of Endocrinology, Qilu Hospital of Shandong University, Jinan, China
| | - Lin Liao
- Department of Endocrinology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| |
Collapse
|
1090
|
Yoest KE, Cummings JA, Becker JB. Oestradiol influences on dopamine release from the nucleus accumbens shell: sex differences and the role of selective oestradiol receptor subtypes. Br J Pharmacol 2018; 176:4136-4148. [PMID: 30381823 DOI: 10.1111/bph.14531] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 10/15/2018] [Accepted: 10/18/2018] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND AND PURPOSE Females are more sensitive than males to both the acute and prolonged effects of psychomotor stimulants. In females, this is regulated by oestradiol, which enhances dopamine release in the dorsal striatum. In this study, we tested the acute effect of oestradiol on dopamine release in the nucleus accumbens (NAc) shell after cocaine administration and investigated which oestradiol receptors (ERs) contribute to sex differences in the response to cocaine. EXPERIMENTAL APPROACH The ability of oestradiol benzoate (EB) to acutely modulate the effect of cocaine on phasic dopamine release in the NAc shell was measured by fast-scan cyclic voltammetry in anaesthetized male and female rats. The roles of ER subtypes, ERα and ERβ, was determined with selective agonists. KEY RESULTS EB acutely enhanced the effect of cocaine on stimulated dopamine release from the NAc shell in females but not in male rats only at levels of stimulation expected to optimally saturate dopamine transporters. Enhanced dopamine release after cocaine administration was also observed in females after selective activation of ERβ but not ERα. EB attenuated the effect of cocaine on NAc shell dopamine reuptake in males but not in females. CONCLUSIONS AND IMPLICATIONS Oestradiol acutely and rapidly regulates dopamine release in females and dopamine reuptake in males. In females, oestradiol rapidly enhances the effect of cocaine on dopamine release, likely via activation of ERβ. The effect of oestradiol in males is not seen with selective receptor subtype activation, a topic deserving of further study. LINKED ARTICLES This article is part of a themed section on The Importance of Sex Differences in Pharmacology Research. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.21/issuetoc.
Collapse
Affiliation(s)
- Katie E Yoest
- Department of Psychology, University of Michigan, Ann Arbor, MI, USA
| | | | - Jill B Becker
- Department of Psychology, University of Michigan, Ann Arbor, MI, USA.,Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA.,Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
1091
|
An H, Kim J, Kim W, Gwon M, Gu HM, Jeon MJ, Han S, Pak SC, Lee C, Park IS, Park K. Therapeutic effects of bee venom and its major component, melittin, on atopic dermatitis in vivo and in vitro. Br J Pharmacol 2018; 175:4310-4324. [PMID: 30187459 PMCID: PMC6240132 DOI: 10.1111/bph.14487] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2017] [Revised: 07/22/2018] [Accepted: 07/27/2018] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND AND PURPOSE Atopic dermatitis (AD) is a multifactorial skin condition with complex interactions of innate and adaptive immune responses. There are several existing therapies for AD, including topical glucocorticosteroids, emollients, phototherapies, calcineurin inhibitors and immunosuppressants, such as cyclosporine A. Although these therapies reduce inflammation, they also cause serious side effects. Therefore, it is necessary to develop new therapeutic approaches for AD treatment without side effects. There are several studies on natural materials or toxins, such as herbs, ginseng extract and snake venom, for AD treatment. However, treatment of AD with bee venom and its major component, melittin has rarely been studied. EXPERIMENTAL APPROACH Effects of bee venom and melittin were studied in a model of AD in vivo induced by 1-chloro-2,4-dinitrobenzene (DNCB) in female Balb/c mice and in cultures of human keratinocytes, stimulated by TNF-α/IFN-γ. The potential pharmacological effects of bee venom and melittin on these in vivo and in vitro AD-like skin disease models were studied. KEY RESULTS Bee venom and melittin exhibited potent anti-atopic activities, shown by decreased AD-like skin lesions, induced by DNCB in mice. In vitro studies using TNF-α/IFN-γ-stimulated human keratinocytes showed that bee venom and melittin inhibited the increased expression of chemokines, such as CCL17 and CCL22, and pro-inflammatory cytokines, including IL-1β, IL-6 and IFN-γ, through the blockade of the NF-κB and STAT signalling pathways. CONCLUSIONS AND IMPLICATIONS Our results suggest that bee venom and melittin would be suitable for epicutaneous application, as topical administration is often appropriate for the treatment of AD.
Collapse
Affiliation(s)
- Hyun‐Jin An
- Department of Pathology, College of MedicineCatholic University of DaeguDaeguKorea
| | - Jung‐Yeon Kim
- Department of Pathology, College of MedicineCatholic University of DaeguDaeguKorea
| | - Woon‐Hae Kim
- Department of Pathology, College of MedicineCatholic University of DaeguDaeguKorea
| | - Mi‐Gyeong Gwon
- Department of Pathology, College of MedicineCatholic University of DaeguDaeguKorea
| | - Hye Min Gu
- Department of Pathology, College of MedicineCatholic University of DaeguDaeguKorea
| | - Min Ji Jeon
- Department of Pathology, College of MedicineCatholic University of DaeguDaeguKorea
| | - Sang‐Mi Han
- Department of Agricultural BiologyNational Academy of Agricultural ScienceJeonju‐siKorea
| | - Sok Cheon Pak
- School of Biomedical SciencesCharles Sturt UniversityBathurstNSWAustralia
| | - Chong‐Kee Lee
- Department of Immunology, College of MedicineCatholic University of DaeguDaeguKorea
| | - In Sook Park
- Department of Oral and Maxillofacial Surgery, Department of Dentistry, College of MedicineCatholic University of DaeguDaeguKorea
| | - Kwan‐Kyu Park
- Department of Pathology, College of MedicineCatholic University of DaeguDaeguKorea
| |
Collapse
|
1092
|
Fan J, Li D, Chen HS, Huang JG, Xu JF, Zhu WW, Chen JG, Wang F. Metformin produces anxiolytic-like effects in rats by facilitating GABA A receptor trafficking to membrane. Br J Pharmacol 2018; 176:297-316. [PMID: 30318707 DOI: 10.1111/bph.14519] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 09/15/2018] [Accepted: 09/19/2018] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND AND PURPOSE Altered function or expression of GABAA receptors contributes to anxiety disorders. Benzodiazepines are widely prescribed for the treatment of anxiety. However, the long-term use of benzodiazepines increases the risk of developing drug dependence and tolerance. Thus, it is urgent to explore new therapeutic approaches. Metformin is widely used to treat Type 2 diabetes and other metabolic syndromes. However, the role of metformin in psychiatric disorders, especially anxiety, remains largely unknown. EXPERIMENTAL APPROACH We examined the effects of metformin on anxiety-like behaviour of rats in open field test and elevated plus maze test. We also observed the effect of metformin (10 μM, in vitro; 100 mg·kg-1 , in vivo) on the trafficking of GABAA receptors, as mechanisms underlying the anxiolytic effects of metformin. KEY RESULTS Metformin (100 mg·kg-1 , i.p. 30 min) displayed a robust and rapid anxiolytic effect, without tolerance. Metformin up-regulated the surface expression of GABAA receptors and increased miniature inhibitory postsynaptic currents (mIPSCs). AMP-activated protein kinase (AMPK) activated by metformin-induced stimulation of forkhead box O3a (FoxO3a) transcriptional activity, followed by increased expression of GABAA receptor-associated protein (GABARAP) and its binding to GABAA receptors finally resulted in the membrane insertion of GABAA receptors. CONCLUSIONS AND IMPLICATIONS Metformin increased mIPSCs by up-regulating the membrane insertion of GABAA receptors, via a pathway involving AMPK, FoxO3a, and the GABAA receptor-associated protein. Thus metformin has a potential new use in the treatment of anxiety disorders.
Collapse
Affiliation(s)
- Jun Fan
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Di Li
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong-Sheng Chen
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian-Geng Huang
- Department of Pharmaceutics, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jun-Feng Xu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wen-Wen Zhu
- Department of Pharmaceutics, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian-Guo Chen
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,The Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan, China.,Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation (HUST), Wuhan, China.,Laboratory of Neuropsychiatric Diseases, The Institute of Brain Research, Huazhong University of Science and Technology, Wuhan, China.,The Collaborative-Innovation Center for Brain Science, Wuhan, China
| | - Fang Wang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,The Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan, China.,Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation (HUST), Wuhan, China.,Laboratory of Neuropsychiatric Diseases, The Institute of Brain Research, Huazhong University of Science and Technology, Wuhan, China.,The Collaborative-Innovation Center for Brain Science, Wuhan, China
| |
Collapse
|
1093
|
Li DJ, Tong J, Zeng FY, Guo M, Li YH, Wang H, Wang P. Nicotinic ACh receptor α7 inhibits PDGF-induced migration of vascular smooth muscle cells by activating mitochondrial deacetylase sirtuin 3. Br J Pharmacol 2018; 176:4388-4401. [PMID: 30270436 DOI: 10.1111/bph.14506] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 09/07/2018] [Accepted: 09/13/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND AND PURPOSE PDGF-BB is an angiogenic factor involved in cardiovascular diseases. Here, we investigated the possible effects of activation of the nicotinic ACh receptor α7 subtype (α7nAChR) on PDGF-BB-induced proliferation and migration in vascular smooth muscle cells (VSMCs). EXPERIMENTAL APPROACH PNU-282987, a selective α7nAChR pharmacological agonist, was used to activate α7nAChR. The influences of α7nAChR activation on PDGF-BB-induced proliferation and migration, as well as the phosphorylation of focal adhesion kinase (FAK)/Src, a pro-migration signalling pathway, were determined in VSMCs. A variety of biochemical assays were applied to explore the underlying molecular mechanisms. KEY RESULTS PDGF-BB induced pronounced migration and proliferation of VSMCs. Activation of α7nAChRs by PNU-282987 blocked PDGF-BB-induced migration but not proliferation in wild-type (WT) VSMCs, whereas this effect was absent in α7nAChR-knockout VSMCs. Accordingly, PNU-282987 attenuated PDGF-BB-induced phosphorylation of FAKTyr397 and SrcTyr416 in WT VSMCs. Mechanistically, PNU-282987 suppressed PDGF-BB-induced oxidative stress, as demonstrated by the alterations in ROS, H2 O2 content, superoxide anion and total antioxidant activity. A sirtuin 3 (SIRT3) inhibitor 3-(1H-1,2,3-triazol-4-yl) pyridine or shRNA-mediated SIRT3 knockdown abolished the inhibitory effect of PNU-282987. PNU-282987 did not modulate SIRT3 protein expression but enhanced mitochondrial SIRT3 deacetylase activity. In line with this action, PNU-282987 enhanced the deacetylation of mitochondrial FoxO3. Lastly, PNU-282987 corrected the PDGF-BB-induced mitochondrial dysfunction by increasing mitochondrial citrate synthase activity, ATP content and nicotinamide adenine dinucleotide pool. CONCLUSIONS Pharmacological activation of α7nAChRs inhibits PDGF-BB-induced VSMC migration by activating the mitochondrial deacetylase SIRT3, implying an important role for α7nAChRs in mitochondria biology and PDGF-related diseases. LINKED ARTICLES This article is part of a themed section on Mitochondrial Pharmacology: Featured Mechanisms and Approaches for Therapy Translation. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.22/issuetoc.
Collapse
Affiliation(s)
- Dong-Jie Li
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China.,Department of Pharmacology, School of Medicine, Tongji University, Shanghai, China.,Ninghai First Hospital, Zhejiang, China
| | - Jie Tong
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China.,Department of Pharmacology, School of Medicine, Tongji University, Shanghai, China.,Ninghai First Hospital, Zhejiang, China
| | - Fei-Yan Zeng
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China.,Department of Pharmacology, School of Medicine, Tongji University, Shanghai, China.,Ninghai First Hospital, Zhejiang, China
| | - Mengqi Guo
- Key Laboratory of Molecular Pharmacology and Drug Evaluation, School of Pharmacy, Yantai University, Yantai, China
| | - Yong-Hua Li
- Department of Anesthesiology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Hongbo Wang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation, School of Pharmacy, Yantai University, Yantai, China
| | - Pei Wang
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China.,Key Laboratory of Molecular Pharmacology and Drug Evaluation, School of Pharmacy, Yantai University, Yantai, China.,Department of Pharmacology, School of Medicine, Tongji University, Shanghai, China.,Department of Pharmacology, School of Pharmacy, Second Military Medical University, Shanghai, China
| |
Collapse
|
1094
|
Wu B, Feng J, Yu L, Wang Y, Chen Y, Wei Y, Han J, Feng X, Zhang Y, Di S, Ma Z, Fan C, Ha X. Icariin protects cardiomyocytes against ischaemia/reperfusion injury by attenuating sirtuin 1-dependent mitochondrial oxidative damage. Br J Pharmacol 2018; 175:4137-4153. [PMID: 30051466 PMCID: PMC6177614 DOI: 10.1111/bph.14457] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 07/02/2018] [Accepted: 07/12/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND AND PURPOSE Icariin, a major active ingredient in traditional Chinese medicines, is attracting increasing attention because of its unique pharmacological effects against ischaemic heart disease. The histone deacetylase, sirtuin-1, plays a protective role in ischaemia/reperfusion (I/R) injury, and this study was designed to investigate the protective role of icariin in models of cardiac I/R injury and to elucidate the potential involvement of sirtuin-1. EXPERIMENTAL APPROACH I/R injury was simulated in vivo (mouse hearts), ex vivo (isolated rat hearts) and in vitro (neonatal rat cardiomyocytes and H9c2 cells). Prior to I/R injury, animals or cells were exposed to icariin, with or without inhibitors of sirtuin-1 (sirtinol and SIRT1 siRNA). KEY RESULTS In vivo and in vitro, icariin given before I/R significantly improved post-I/R heart contraction and limited the infarct size and leakage of creatine kinase-MB and LDH from the damaged myocardium. Icariin also attenuated I/R-induced mitochondrial oxidative damage, decreasing malondialdehyde content and increasing superoxide dismutase activity and expression of Mn-superoxide dismutase. Icariin significantly improved mitochondrial membrane homeostasis by increasing mitochondrial membrane potential and cytochrome C stabilization, which further inhibited cell apoptosis. Sirtuin-1 was significantly up-regulated in hearts treated with icariin, whereas Ac-FOXO1 was simultaneously down-regulated. Importantly, sirtinol and SIRT1 siRNA either blocked icariin-induced cardioprotection or disrupted icariin-mediated mitochondrial homeostasis. CONCLUSIONS AND IMPLICATIONS Pretreatment with icariin protected cardiomyocytes from I/R-induced oxidative stress through activation of sirtuin-1 /FOXO1 signalling.
Collapse
Affiliation(s)
- Bing Wu
- Department of GeriatricsLanzhou General Hospital of the People's Liberation ArmyLanzhouChina
- Department of CardiologyTangdu Hospital, Fourth Military Medical UniversityXi'anChina
| | - Jian‐yu Feng
- Department of Cardiovascular Surgery, Xijing HospitalFourth Military Medical UniversityXi'anChina
| | - Li‐ming Yu
- Department of Cardiovascular SurgeryGeneral Hospital of Shenyang Military Area CommandShenyangChina
| | - Yan‐chun Wang
- Department of GeriatricsLanzhou General Hospital of the People's Liberation ArmyLanzhouChina
| | - Yong‐qing Chen
- Department of CardiologyLanzhou General Hospital of the People's Liberation ArmyLanzhouChina
| | - Yan Wei
- Department of ophthalmologyLanzhou General Hospital of the People's Liberation ArmyLanzhouChina
| | - Jin‐song Han
- Department of Cardiovascular SurgeryGeneral Hospital of Shenyang Military Area CommandShenyangChina
| | - Xiao Feng
- Department of Cardiovascular Surgery, Xijing HospitalFourth Military Medical UniversityXi'anChina
| | - Yu Zhang
- Department of Cardiovascular SurgeryLanzhou General Hospital of the People's Liberation ArmyLanzhouChina
| | - Shou‐yin Di
- Department of Thoracic SurgeryTangdu Hospital, Fourth Military Medical UniversityXi'anChina
| | - Zhi‐qiang Ma
- Department of Thoracic SurgeryTangdu Hospital, Fourth Military Medical UniversityXi'anChina
| | - Chong‐xi Fan
- Department of Thoracic SurgeryTangdu Hospital, Fourth Military Medical UniversityXi'anChina
- Department of Biomedical EngineeringFourth Military Medical UniversityXi'anChina
| | - Xiao‐qin Ha
- Department of Clinical LaboratoryLanzhou General Hospital of the People's Liberation ArmyLanzhouChina
| |
Collapse
|
1095
|
|
1096
|
Aboumsallem JP, Mishra M, Amin R, Muthuramu I, Kempen H, De Geest B. Successful treatment of established heart failure in mice with recombinant HDL (Milano). Br J Pharmacol 2018; 175:4167-4182. [PMID: 30079544 PMCID: PMC6177616 DOI: 10.1111/bph.14463] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 07/09/2018] [Accepted: 07/12/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND AND PURPOSE The pleiotropic properties of HDL may exert beneficial effects on the myocardium. The effect of recombinant HDLMilano on established heart failure was evaluated in C57BL/6 mice. EXPERIMENTAL APPROACH Mice were subjected to transverse aortic constriction (TAC) or sham operation at the age of 14 weeks. Eight weeks later, TAC and sham mice were each randomized into three different groups. Reference groups were killed at day 56 after the operation for baseline analysis. Five i.p. injections of recombinant HDLMilano (MDCO-216), 100 mg·kg-1 , or an equivalent volume of control buffer were administered with a 48 h interval starting at day 56. Endpoint analyses in the control buffer groups and in the MDCO-216 groups were executed at day 65. KEY RESULTS Lung weight in MDCO-216 TAC mice was 25.3% lower than in reference TAC mice and 27.9% lower than in control buffer TAC mice and was similar in MDCO-216 sham mice. MDCO-216 significantly decreased interstitial fibrosis and increased relative vascularity compared to reference TAC mice and control buffer TAC mice. The peak rate of isovolumetric relaxation in MDCO-216 TAC mice was 30.4 and 36.3% higher than in reference TAC mice and control buffer TAC mice respectively. Nitro-oxidative stress and myocardial apoptosis were significantly reduced in MDCO-216 TAC mice compared to control buffer TAC mice. CONCLUSIONS AND IMPLICATIONS MDCO-216 improves diastolic function, induces regression of interstitial fibrosis and normalizes lung weight in mice with established heart failure. Recombinant HDL may emerge as a treatment modality in heart failure.
Collapse
Affiliation(s)
- Joseph Pierre Aboumsallem
- Centre for Molecular and Vascular Biology, Department of Cardiovascular SciencesCatholic University of LeuvenLeuvenBelgium
| | - Mudit Mishra
- Centre for Molecular and Vascular Biology, Department of Cardiovascular SciencesCatholic University of LeuvenLeuvenBelgium
| | - Ruhul Amin
- Centre for Molecular and Vascular Biology, Department of Cardiovascular SciencesCatholic University of LeuvenLeuvenBelgium
| | - Ilayaraja Muthuramu
- Centre for Molecular and Vascular Biology, Department of Cardiovascular SciencesCatholic University of LeuvenLeuvenBelgium
| | - Herman Kempen
- The Medicines Company (Schweiz) GmbHZürichSwitzerland
| | - Bart De Geest
- Centre for Molecular and Vascular Biology, Department of Cardiovascular SciencesCatholic University of LeuvenLeuvenBelgium
| |
Collapse
|
1097
|
Liu T, Wang P, Cong M, Zhao X, Zhang D, Xu H, Liu L, Jia J, You H. Diethyldithiocarbamate, an anti-abuse drug, alleviates steatohepatitis and fibrosis in rodents through modulating lipid metabolism and oxidative stress. Br J Pharmacol 2018; 175:4480-4495. [PMID: 30266038 DOI: 10.1111/bph.14503] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 08/23/2018] [Accepted: 09/15/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND PURPOSE Diethyldithiocarbamate (DDC) is a major metabolite of disulfiram that is a potential drug for alcoholism treatment. In the present study, we attempted to explore the possible effect of DDC on non-alcoholic fatty liver disease (NAFLD) and related fibrosis in vivo. EXPERIMENTAL APPROACH C57BL/6 mice and Sprague Dawley (SD) rats received a methionine/choline-deficient (MCD) diet to establish the model of NAFLD with or without DDC treatment. The livers and serum were assessed for histological changes and parameters related to lipid metabolism, liver injury, inflammation and fibrosis. Apoptosis and macrophage related markers were assessed by immunohistochemistry (IHC). KEY RESULTS DDC significantly reduced hepatic steatosis in rats with NAFLD, induced by the MCD diet. DDC reduced the oxidative stress and endoplasmic reticulum stress-related parameters in mice with non-alcoholic steatohepatitis, induced by the MCD diet. IHC for Bax and cleaved caspase-3 showed that DDC inhibited the apoptosis of hepatocytes in the liver. DDC significantly reduced ballooning and Mallory-Denk bodies (MDB) in hepatocytes, accompanied by suppression of serum alanine aminotransferase, aspartate aminotransferase and MDB formation-related genes. DDC significantly alleviated hepatic inflammation, accompanied by suppression of inflammation-related genes. DDC suppressed the infiltration of macrophages, particularly inducible NOS-positive pro-inflammatory macrophages. In addition, DDC significantly alleviated liver fibrosis. Microarray analyses showed that DDC strongly affected lipid metabolism and oxidative stress-related processes and pathways. CONCLUSION AND IMPLICATIONS DDC improves hepatic steatosis, ballooning, inflammation and fibrosis in rodent models of NAFLD through modulating lipid metabolism and oxidative stress.
Collapse
Affiliation(s)
- Tianhui Liu
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University.,Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis and National Clinical Research Center of Digestive Disease, Beijing, China
| | - Ping Wang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University.,Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis and National Clinical Research Center of Digestive Disease, Beijing, China
| | - Min Cong
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University.,Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis and National Clinical Research Center of Digestive Disease, Beijing, China
| | - Xinyan Zhao
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University.,Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis and National Clinical Research Center of Digestive Disease, Beijing, China
| | - Dong Zhang
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Hufeng Xu
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Lin Liu
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University.,Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis and National Clinical Research Center of Digestive Disease, Beijing, China
| | - Jidong Jia
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University.,Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis and National Clinical Research Center of Digestive Disease, Beijing, China
| | - Hong You
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University.,Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis and National Clinical Research Center of Digestive Disease, Beijing, China
| |
Collapse
|
1098
|
Kang LJ, Kwon ES, Lee KM, Cho C, Lee JI, Ryu YB, Youm TH, Jeon J, Cho MR, Jeong SY, Lee SR, Kim W, Yang S. 3'-Sialyllactose as an inhibitor of p65 phosphorylation ameliorates the progression of experimental rheumatoid arthritis. Br J Pharmacol 2018; 175:4295-4309. [PMID: 30152858 PMCID: PMC6240131 DOI: 10.1111/bph.14486] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 07/25/2018] [Accepted: 08/10/2018] [Indexed: 12/25/2022] Open
Abstract
Background and Purpose 3′‐Sialyllactose (3′‐SL) is a safe compound that is present in high levels in human milk. Although it has anti‐inflammatory properties and supports immune homeostasis, its effect on collagen‐induced arthritis (CIA) is unknown. In this study, we investigated the prophylactic and therapeutic effect of 3′‐SL on the progression of rheumatoid arthritis (RA) in in vitro and in vivo models. Experimental Approach The anti‐arthritic effect of 3′‐SL was analysed with fibroblast‐like synoviocytes in vitro and an in vivo mouse model of CIA. RT‐PCR, Western blotting and ELISA were performed to evaluate its effects in vitro. Histological analysis of ankle and knee joints of mice with CIA was performed using immunohistochemistry, as well as safranin‐O and haematoxylin staining. Key Results 3′‐SL markedly alleviated the severity of CIA in the mice by reducing paw swelling, clinical scores, incidence rate, serum levels of inflammatory cytokines and autoantibody production. Moreover, 3′‐SL reduced synovitis and pannus formation and suppressed cartilage destruction by blocking secretion of chemokines, pro‐inflammatory cytokines, https://en.wikipedia.org/wiki/Matrix_metalloproteinases and osteoclastogenesis via NF‐κB signalling. Notably, phosphorylation of p65, which is a key protein in the NF‐κB signalling pathway, was totally blocked by 3′‐SL in the RA models. Conclusions and Implications 3′‐SL ameliorated pathogenesis of CIA by suppressing catabolic factor expression, proliferation of inflammatory immune cells and osteoclastogenesis. These effects were mediated via blockade of the NF‐κB signalling pathway. Therefore, 3′‐SL exerted prophylactic and therapeutic effects and could be a novel therapeutic agent for the treatment of RA.
Collapse
Affiliation(s)
- Li-Jung Kang
- Department of Pharmacology, Ajou University School of Medicine, Suwon, Korea.,Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Korea.,CIRNO, Sungkyunkwan University, Suwon, Korea
| | - Eun-Soo Kwon
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea
| | | | - Chanmi Cho
- Department of Pharmacology, Ajou University School of Medicine, Suwon, Korea.,Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Korea.,CIRNO, Sungkyunkwan University, Suwon, Korea
| | - Jae-In Lee
- Natural Product Material Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup, Korea
| | - Young Bae Ryu
- Natural Product Material Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup, Korea
| | - Tae Hyun Youm
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea.,Laboratory of Physiology, College of Pharmacy, Chungnam National University, Daejeon, Korea
| | - Jimin Jeon
- Department of Pharmacology, Ajou University School of Medicine, Suwon, Korea.,Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Korea.,CIRNO, Sungkyunkwan University, Suwon, Korea
| | - Mi Ra Cho
- Rheumatism Research Center, The Catholic University of Korea, College of Medicine, Seoul, Korea
| | - Seon-Yong Jeong
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Korea.,Department of Medical Genetics, Ajou University School of Medicine, Suwon, Korea
| | - Sang-Rae Lee
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, Korea.,Department of Functional Genomics, University of Science and Technology, Daejeon, Korea
| | - Wook Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, Korea
| | - Siyoung Yang
- Department of Pharmacology, Ajou University School of Medicine, Suwon, Korea.,Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Korea.,CIRNO, Sungkyunkwan University, Suwon, Korea
| |
Collapse
|
1099
|
Zhang C, Deng J, Liu D, Tuo X, Xiao L, Lai B, Yao Q, Liu J, Yang H, Wang N. Nuciferine ameliorates hepatic steatosis in high-fat diet/streptozocin-induced diabetic mice through a PPARα/PPARγ coactivator-1α pathway. Br J Pharmacol 2018; 175:4218-4228. [PMID: 30129056 DOI: 10.1111/bph.14482] [Citation(s) in RCA: 137] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 07/17/2018] [Accepted: 07/27/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND AND PURPOSE Nuciferine, an alkaloid found in Nelumbo nucifera leaves, alleviates dyslipidemia in vivo. However, whether it improves liver injury in diabetic conditions and the underlying mechanism is unclear. The present study aimed to investigate the effects of nuciferine on lipid and glucose metabolism in a murine model of Type 2 diabetes mellitus (T2DM) and to determine the underlying mechanisms of these effects. EXPERIMENTAL APPROACH A murine model of T2DM was induced by high-fat diet (HFD) feeding combined with streptozocin (STZ) injections, and the diabetic mice were treated with nuciferine in their food. The underlying mechanism of the anti-steatotic effect of nuciferine was further explored in HepG2 hepatocytes cultured with palmitic acid. Major signalling profiles involved in fatty acid oxidation were then evaluated, using Western blot, RT-qPCR and si-RNA techniques, along with immunohistochemistry. KEY RESULTS Nuciferine restored impaired glucose tolerance and insulin resistance in diabetic mice. Hepatic levels of total cholesterol, triglycerides and LDL were decreased, as were the number of lipid droplets, by nuciferine treatment. Furthermore, nuciferine up-regulated β-oxidation related genes in livers of diabetic mice. Luciferase reporter cell assay showed that nuciferine directly reversed palmitic acid-induced inhibition of PPARα transcriptional activity. Silencing PPARγ coactivator-1α (PGC1α) expression in HepG2 cells abolished the effects of nuciferine in accelerating β-oxidation. CONCLUSIONS AND IMPLICATIONS Nuciferine improved lipid profile and attenuated hepatic steatosis in HFD/STZ-induced diabetic mice by activating the PPARα/PGC1α pathway. Nuciferine may be a potentially important candidate in improving hepatic steatosis and the management of T2DM.
Collapse
Affiliation(s)
- Chao Zhang
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China.,Department of Nutrition and Food Safety, College of Public Health, Xi'an Jiaotong University, Xi'an, China
| | - Jianjun Deng
- Shaanxi Key Laboratory of Degradable Biomedical Materials, School of Chemical Engineering, Northwest University, Xi'an, China
| | - Dan Liu
- Department of Nutrition and Food Safety, College of Public Health, Xi'an Jiaotong University, Xi'an, China
| | - Xingxia Tuo
- Department of Nutrition and Food Safety, College of Public Health, Xi'an Jiaotong University, Xi'an, China
| | - Lei Xiao
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Baochang Lai
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Qinyu Yao
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Jia Liu
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Haixia Yang
- Department of Nutrition and Food Safety, College of Public Health, Xi'an Jiaotong University, Xi'an, China
| | - Nanping Wang
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China.,The Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| |
Collapse
|
1100
|
Tan WQ, Fang QQ, Shen XZ, Giani JF, Zhao TV, Shi P, Zhang LY, Khan Z, Li Y, Li L, Xu JH, Bernstein EA, Bernstein KE. Angiotensin-converting enzyme inhibitor works as a scar formation inhibitor by down-regulating Smad and TGF-β-activated kinase 1 (TAK1) pathways in mice. Br J Pharmacol 2018; 175:4239-4252. [PMID: 30153328 DOI: 10.1111/bph.14489] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 07/19/2018] [Accepted: 08/16/2018] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND AND PURPOSE Angiotensin-converting enzyme (ACE), an important part of the renin-angiotensin system, is implicated in stimulating the fibrotic processes in the heart, lung, liver and kidney, while an ACE inhibitor (ACEI) promotes physiological tissue repair in these organs. The mechanism is closely related to TGF-β1 pathways. However, the reported effects of applying ACEIs during scar formation are unclear. Hence, we explored the anti-fibrotic effects of an ACEI and the molecular mechanisms involved in a mouse scar model. EXPERIMENTAL APPROACH After a full-thickness skin wound operation, ACE wild-type mice were randomly assigned to receive either ramipril, losartan or hydralazine p.o. ACE knockout (KO) mice and negative control mice only received vehicle (water). Wound/scar widths during wound healing and histological examinations were recorded at the final day. The ability of ACEI to reduce fibrosis via TGF-β1 signalling was evaluated in vitro and in vivo. KEY RESULTS ACE KO mice and mice that received ramipril showed narrower wound/scar width, reduced fibroblast proliferation, decreased collagen and TGF-β1 expression. ACEI attenuated the phosphorylation of small mothers against decapentaplegic (Smad2/3) and TGF-β-activated kinase 1 (TAK1) both in vitro and in vivo. The expression of ACE-related peptides varied in murine models with different drug treatments. CONCLUSIONS AND IMPLICATIONS ACEI showed anti-fibrotic properties in scar formation by mediating downstream peptides to suppress TGF-β1/Smad and TGF-β1/TAK1 pathways. These findings suggest that dual inhibition of Smad and TAK1 signalling by ACEI is a useful strategy for the development of new anti-fibrotic agents.
Collapse
Affiliation(s)
- Wei-Qiang Tan
- Department of Plastic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China.,Department of Plastic Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang Province, China.,Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Qing-Qing Fang
- Department of Plastic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China.,Department of Plastic Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang Province, China
| | - Xiao Z Shen
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Department of Physiology, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Jorge F Giani
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Tuantuan V Zhao
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Peng Shi
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Li-Yun Zhang
- Department of Plastic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China.,Department of Plastic Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang Province, China
| | - Zakir Khan
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - You Li
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Liang Li
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ji-Hua Xu
- Department of Plastic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China.,Department of Plastic Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang Province, China
| | - Ellen A Bernstein
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Kenneth E Bernstein
- Department of Plastic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China.,Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|