201
|
Woyach V, Sherman K, Hillard CJ, Hopp FA, Hogan QH, Dean C. Fatty acid amide hydrolase activity in the dorsal periaqueductal gray attenuates neuropathic pain and associated dysautonomia. Am J Physiol Regul Integr Comp Physiol 2022; 323:R749-R762. [PMID: 36154489 PMCID: PMC9639763 DOI: 10.1152/ajpregu.00073.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 08/29/2022] [Accepted: 09/22/2022] [Indexed: 01/11/2023]
Abstract
The complexity of neuropathic pain and its associated comorbidities, including dysautonomia, make it difficult to treat. Overlap of anatomical regions and pharmacology of sympathosensory systems in the central nervous system (CNS) provide targets for novel treatment strategies. The dorsal periaqueductal gray (dPAG) is an integral component of both the descending pain modulation system and the acute stress response and is critically involved in both analgesia and the regulation of sympathetic activity. Local manipulation of the endocannabinoid signaling system holds great promise to provide analgesia without excessive adverse effects and also influence autonomic output. Inhibition of fatty acid amide hydrolase (FAAH) increases brain concentrations of the endocannabinoid N-arachidonoylethanolamine (AEA) and reduces pain-related behaviors in neuropathic pain models. Neuropathic hyperalgesia and reduced sympathetic tone are associated with increased FAAH activity in the dPAG, which suggests the hypothesis that inhibition of FAAH in the dPAG will normalize pain sensation and autonomic function in neuropathic pain. To test this hypothesis, the effects of systemic or intra-dPAG FAAH inhibition on hyperalgesia and dysautonomia developed after spared nerve injury (SNI) were assessed in male and female rats. Administration of the FAAH inhibitor PF-3845 into the dPAG reduces hyperalgesia behavior and the decrease in sympathetic tone induced by SNI. Prior administration of the CB1 receptor antagonist AM281, attenuated the antihyperalgesic and sympathetic effects of FAAH inhibition. No sex differences were identified. These data support an integrative role for AEA/CB1 receptor signaling in the dPAG contributing to the regulation of both hyperalgesia behavior and altered sympathetic tone in neuropathic pain.
Collapse
Affiliation(s)
- Victoria Woyach
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin
- Research Service, Zablocki Veterans Affairs Medical Center, Milwaukee Wisconsin
| | - Katherine Sherman
- Research Service, Zablocki Veterans Affairs Medical Center, Milwaukee Wisconsin
| | - Cecilia J Hillard
- Department of Pharmacology and Toxicology and Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Francis A Hopp
- Research Service, Zablocki Veterans Affairs Medical Center, Milwaukee Wisconsin
| | - Quinn H Hogan
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin
- Research Service, Zablocki Veterans Affairs Medical Center, Milwaukee Wisconsin
| | - Caron Dean
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin
- Research Service, Zablocki Veterans Affairs Medical Center, Milwaukee Wisconsin
| |
Collapse
|
202
|
Guo S, Li A, Fu X, Li Z, Cao K, Song M, Huang S, Li Z, Yan J, Wang L, Dai X, Feng D, Wang Y, He J, Huo Y, Xu Y. Gene-dosage effect of Pfkfb3 on monocyte/macrophage biology in atherosclerosis. Br J Pharmacol 2022; 179:4974-4991. [PMID: 35834356 PMCID: PMC10420406 DOI: 10.1111/bph.15926] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 06/26/2022] [Accepted: 07/07/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Macrophage-rich atherosclerotic arteries are highly active in glycolysis. PFKFB3, a key glycolytic enzyme, has emerged as a potential therapeutic target in atherosclerosis. Small-molecule inhibitors of PFKFB3, such as 3PO and PFK158, have demonstrated efficacy in hampering atherogenesis in preclinical models. However, genetic studies elucidating the role of Pfkfb3 in atherogenesis need to be conducted to validate pharmacological findings and to unveil potential pharmacological side effects. EXPERIMENTAL APPROACH Apoe-/- mice with global heterozygous or myeloid cell-specific Pfkfb3 deficiency were fed a Western diet (WD), after which atherosclerosis development was determined. Monocyte subsets in atherosclerotic mice and patients were examined by flow cytometry. Monocyte infiltration was assayed by a Ly6Chi monocyte-specific latex labelling procedure. In situ efferocytosis was assessed on mouse aortic root sections. Additionally, metabolic status, macrophage motility, efferocytosis, and involved mechanisms were analysed in peritoneal macrophages. KEY RESULTS Global heterozygous or myeloid cell-specific Pfkfb3 deficiency reduced atherogenesis in Apoe-/- mice. Mechanistic studies showed that PFKFB3 controlled the proliferation and infiltration of proinflammatory monocytes. Moreover, PFKFB3 expression was associated with inflammatory monocyte expansion in patients with atherosclerotic coronary artery disease. Surprisingly, homozygous loss of Pfkfb3 impaired macrophage efferocytosis and exacerbated atherosclerosis in Apoe-/- mice. Mechanistically, PFKFB3-driven glycolysis was shown to be essential for actin polymerization, thus aiding the efferocytotic function of macrophages. CONCLUSION AND IMPLICATIONS Collectively, these findings suggest the existence of a double-edged sword effect of myeloid PFKFB3 on the pathogenesis of atherosclerosis and highlight the need for caution in developing anti-atherosclerotic strategies that target PFKFB3.
Collapse
Affiliation(s)
- Shuai Guo
- School of Basic Medical Sciences, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Anqi Li
- School of Basic Medical Sciences, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Guangzhou Medical University, Guangzhou, China
- Department of Radiology, the Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xiaodong Fu
- School of Basic Medical Sciences, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Zou Li
- School of Basic Medical Sciences, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Kaixiang Cao
- School of Basic Medical Sciences, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Mingchuan Song
- School of Basic Medical Sciences, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Shuqi Huang
- School of Basic Medical Sciences, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Ziling Li
- School of Basic Medical Sciences, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jingwei Yan
- School of Basic Medical Sciences, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Litao Wang
- School of Basic Medical Sciences, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Guangzhou Medical University, Guangzhou, China
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaoyan Dai
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Du Feng
- School of Basic Medical Sciences, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yong Wang
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jun He
- Department of Rehabilitation Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuqing Huo
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Yiming Xu
- School of Basic Medical Sciences, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
203
|
Broekhuizen M, de Vries R, Smits MAW, Dik WA, Schoenmakers S, Koch BCP, Merkus D, Reiss IKM, Danser AHJ, Simons SHP, Hitzerd E. Pentoxifylline as a therapeutic option for pre-eclampsia: a study on its placental effects. Br J Pharmacol 2022; 179:5074-5088. [PMID: 35861684 PMCID: PMC9804511 DOI: 10.1111/bph.15931] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 07/05/2022] [Accepted: 07/11/2022] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND AND PURPOSE Recently pentoxifylline, a non-selective phosphodiesterase inhibitor and adenosine receptor antagonist, has attracted much interest for the treatment of the increased vascular resistance and endothelial dysfunction in pre-eclampsia. We therefore investigated the placental transfer, vascular effects and anti-inflammatory actions of pentoxifylline in healthy and pre-eclamptic human placentas. EXPERIMENTAL APPROACH The placental transfer and metabolism of pentoxifylline were studied using ex vivo placenta perfusion experiments. In wire myography experiments with chorionic plate arteries, pentoxifyllines vasodilator properties were investigated, focusing on the cGMP and cAMP pathways and adenosine receptors. Its effects on inflammatory factors were also studied in placental explants. KEY RESULTS Pentoxifylline transferred from the maternal to foetal circulation, reaching identical concentrations. The placenta metabolized pentoxifylline into its active metabolite lisofylline (M1), which was released into both circulations. In healthy placentas, pentoxifylline potentiated cAMP- and cGMP-induced vasodilation, as well as causing vasodilation by adenosine A1 antagonism and via NO synthase and PKG. Pentoxifylline also reduced inflammatory factors secretion. In pre-eclamptic placentas, we observed that its vasodilator capacity was preserved, however not via NO-PKG but likely through adenosine signalling. Pentoxifylline neither potentiated vasodilation through cAMP and cGMP, nor suppressed the release of inflammatory factors from these placentas. CONCLUSION AND IMPLICATIONS Pentoxifylline is transferred across and metabolized by the placenta. Its beneficial effects on the NO pathway and inflammation are not retained in pre-eclampsia, limiting its application in this disease, although it could be useful for other placenta-related disorders. Future studies might focus on selective A1 receptor antagonists as a new treatment for pre-eclampsia.
Collapse
Affiliation(s)
- Michelle Broekhuizen
- Division of Neonatology, Department of PaediatricsErasmus MC University Medical CenterRotterdamThe Netherlands,Division of Pharmacology and Vascular Medicine, Department of Internal MedicineErasmus MC University Medical CenterRotterdamThe Netherlands,Division of Experimental Cardiology, Department of CardiologyErasmus MC University Medical CenterRotterdamThe Netherlands
| | - Rene de Vries
- Division of Pharmacology and Vascular Medicine, Department of Internal MedicineErasmus MC University Medical CenterRotterdamThe Netherlands
| | - Marja A. W. Smits
- Laboratory Medical Immunology, Department of ImmunologyErasmus MC University Medical CenterRotterdamThe Netherlands
| | - Willem A. Dik
- Laboratory Medical Immunology, Department of ImmunologyErasmus MC University Medical CenterRotterdamThe Netherlands
| | - Sam Schoenmakers
- Department of Obstetrics and GynaecologyErasmus MC University Medical CenterRotterdamThe Netherlands
| | - Birgit C. P. Koch
- Department of PharmacyErasmus MC University Medical CenterRotterdamThe Netherlands
| | - Daphne Merkus
- Division of Experimental Cardiology, Department of CardiologyErasmus MC University Medical CenterRotterdamThe Netherlands
| | - Irwin K. M. Reiss
- Division of Neonatology, Department of PaediatricsErasmus MC University Medical CenterRotterdamThe Netherlands
| | - A. H. Jan Danser
- Division of Pharmacology and Vascular Medicine, Department of Internal MedicineErasmus MC University Medical CenterRotterdamThe Netherlands
| | - Sinno H. P. Simons
- Division of Neonatology, Department of PaediatricsErasmus MC University Medical CenterRotterdamThe Netherlands
| | - Emilie Hitzerd
- Division of Neonatology, Department of PaediatricsErasmus MC University Medical CenterRotterdamThe Netherlands,Division of Pharmacology and Vascular Medicine, Department of Internal MedicineErasmus MC University Medical CenterRotterdamThe Netherlands
| |
Collapse
|
204
|
Liu Z, Mao X, Yang Q, Zhang X, Xu J, Ma Q, Zhou Y, Da Q, Cai Y, Sopeyin A, Dong Z, Hong M, Caldwell RB, Sodhi A, Huo Y. Suppression of myeloid PFKFB3-driven glycolysis protects mice from choroidal neovascularization. Br J Pharmacol 2022; 179:5109-5131. [PMID: 35830274 DOI: 10.1111/bph.15925] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 06/09/2022] [Accepted: 07/05/2022] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND AND PURPOSE Pathological angiogenesis is a major cause of irreversible blindness in individuals with neovascular age-related macular degeneration (nAMD). Macrophages and microglia (MΦ) contribute to aberrant ocular angiogenesis. However, the role of glucose metabolism of MΦ in nAMD is still undefined. Here, we have investigated the involvement of glycolysis, driven by the kinase/phosphatase PFKFB3, in the development of choroidal neovascularization (CNV). EXPERIMENTAL APPROACH CNV was induced in mice with laser photocoagulation. Choroid/retinal pigment epithelium (RPE) complexes and MΦ were isolated for analysis by qRT-PCR, western blot, flow cytometry, immunostaining, metabolic measurements and angiogenesis assays. KEY RESULTS MΦ accumulated within the CNV of murine nAMD models and expressed high levels of glycolysis-related enzymes and M1/M2 polarization markers. This phenotype of hyper-glycolytic and activated MΦ was replicated in bone marrow-derived macrophages stimulated by necrotic RPE in vitro. Myeloid cell-specific knockout of PFKFB3, a key glycolytic activator, attenuated pathological neovascularization in laser-induced CNV, which was associated with decreased expression of MΦ polarization markers and pro-angiogenic factors, along with decreased sprouting of vessels in choroid/RPE complexes. Mechanistically, necrotic RPE increased PFKFB3-driven glycolysis in macrophages, leading to activation of HIF-1α/HIF-2α and NF-κB, and subsequent induction of M1/M2 markers and pro-angiogenic cytokines, finally promoting macrophage reprogramming towards an angiogenic phenotype to facilitate development of CNV. The PFKFB3 inhibitor AZ67 also inhibited activation of HIF-1α/HIF-2α and NF-κB signalling and almost completely prevented laser-induced CNV in mice. CONCLUSIONS AND IMPLICATIONS Modulation of PFKFB3-mediated macrophage glycolysis and activation is a promising strategy for the treatment of nAMD.
Collapse
Affiliation(s)
- Zhiping Liu
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Xiaoxiao Mao
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Qiuhua Yang
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Xiaoyu Zhang
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Jiean Xu
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Qian Ma
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Yaqi Zhou
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Qingen Da
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Yongfeng Cai
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Anu Sopeyin
- Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Zheng Dong
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
- Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia, USA
| | - Mei Hong
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Ruth B Caldwell
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
- Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia, USA
- James and Jean Culver Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Akrit Sodhi
- Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Yuqing Huo
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
- James and Jean Culver Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| |
Collapse
|
205
|
Kallend D, Mason J, Smith PF, Koren MJ, Stoekenbroek R, He Y, Wijngaard P. An evaluation of a supratherapeutic dose of inclisiran on cardiac repolarization in healthy volunteers: A phase I, randomized study. Clin Transl Sci 2022; 15:2663-2672. [PMID: 36196601 PMCID: PMC9652432 DOI: 10.1111/cts.13391] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 08/02/2022] [Accepted: 08/04/2022] [Indexed: 01/25/2023] Open
Abstract
Inclisiran is a small interfering RNA molecule that has been shown to provide an effective and sustained reduction in low-density lipoprotein cholesterol levels. This study aimed to determine whether a supratherapeutic dose of inclisiran affects cardiac repolarization and conduction in healthy volunteers. A phase I, randomized, double-blind, double-dummy, placebo- and positive-controlled, three-way crossover study was performed in 48 healthy volunteers. Volunteers were assigned to three treatments in a randomized sequence: a supratherapeutic dose of inclisiran sodium (900 mg), placebo, or moxifloxacin 400 mg as a positive control, with a minimum 7-day washout period between treatments. Continuous electrocardiogram monitoring was performed from >60 min before dosing until 48 h after dosing. Pharmacokinetics, pharmacodynamics, and safety were also assessed. Inclisiran, at a supratherapeutic dose, did not show a clinically significant effect on the QT interval (Fridericia correction formula [QTcF]; maximal placebo- and baseline-corrected change: 2.5 ms [90% confidence interval: 0.6, 4.5]) near the maximal plasma concentrations at 4 h. In addition, inclisiran did not show any effects on other electrocardiogram intervals or ST- and T-wave morphology. The positive control, moxifloxacin, demonstrated the expected changes in QTcF interval, validating the adequate sensitivity of the study. A supratherapeutic dose of inclisiran sodium (900 mg) had no effect on the QTcF interval or other electrocardiogram parameters, providing additional insight and reassurance regarding the safety profile of inclisiran.
Collapse
Affiliation(s)
- David Kallend
- Former Employee of The Medicines Company (Schweiz) GmbHZurichSwitzerland
| | - Jay Mason
- Department of Internal Medicine, University of Utah School of MedicineSalt Lake CityUtahUSA
| | | | - Michael J. Koren
- First Coast Heart & Vascular CenterJacksonvilleFloridaUSA
- Jacksonville Center for Clinical ResearchJacksonvilleFloridaUSA
| | | | - YanLing He
- Novartis Institutes for BioMedical Research, Inc.CambridgeMassachusettsUSA
| | - Peter Wijngaard
- Former Employee of Novartis Pharmaceuticals CorporationEast HanoverNew JerseyUSA
| |
Collapse
|
206
|
Lindsay C, Musgaard M, Russell AJ, Sitsapesan R. Statin activation of skeletal ryanodine receptors (RyR1) is a class effect but separable from HMG-CoA reductase inhibition. Br J Pharmacol 2022; 179:4941-4957. [PMID: 35703154 PMCID: PMC9804224 DOI: 10.1111/bph.15893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 04/20/2022] [Accepted: 04/28/2022] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND AND PURPOSE Statins, inhibitors of HMG-CoA reductase, are mainstay treatment for hypercholesterolaemia. However, muscle pain and weakness prevent many patients from benefiting from their cardioprotective effects. We previously demonstrated that simvastatin activates skeletal ryanodine receptors (RyR1), an effect that could be important in initiating myopathy. Using a range of structurally diverse statin analogues, we examined structural features associated with RyR1 activation, aiming to identify statins lacking this property. EXPERIMENTAL APPROACH Compounds were screened for RyR1 activity utilising [3 H]ryanodine binding. Mechanistic insight into RyR1 activity was studied by incorporating RyR1 channels from sheep, mouse or rabbit skeletal muscle into bilayers. KEY RESULTS All UK-prescribed statins activated RyR1 at nanomolar concentrations. Cerivastatin, withdrawn from the market due to life-threatening muscle-related side effects, was more effective than currently-prescribed statins and possessed the unique ability to open RyR1 channels independently of cytosolic Ca2+ . We synthesised the one essential structural moiety that all statins must possess for HMG-CoA reductase inhibition, the R-3,5-dihydroxypentanoic acid unit, and it did not activate RyR1. We also identified five analogues retaining potent HMG-CoA reductase inhibition that inhibited RyR1 and four that lacked the ability to modulate RyR1. CONCLUSION AND IMPLICATIONS That cerivastatin activates RyR1 most strongly supports the hypothesis that RyR1 activation is implicated in statin-induced myopathy. Demonstrating that statin regulation of RyR1 and HMG-CoA reductase are separable effects will allow the role of RyR1 in statin-induced myopathy to be further elucidated by the tool compounds we have identified, allowing development of effective cardioprotective statins with improved patient tolerance.
Collapse
Affiliation(s)
- Chris Lindsay
- Department of PharmacologyUniversity of OxfordOxfordUK
| | - Maria Musgaard
- Structural Bioinformatics and Computational Biochemistry, Department of BiochemistryUniversity of OxfordOxfordUK
- OMass TherapeuticsOxfordUK
| | - Angela J. Russell
- Department of PharmacologyUniversity of OxfordOxfordUK
- Department of Chemistry, Chemistry Research LaboratoryUniversity of OxfordOxfordUK
| | | |
Collapse
|
207
|
Microglia senescence is related to neuropathic pain-associated comorbidities in the spared nerve injury model. Pain 2022; 164:1106-1117. [PMID: 36448971 PMCID: PMC10108589 DOI: 10.1097/j.pain.0000000000002807] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 10/05/2022] [Indexed: 12/02/2022]
Abstract
ABSTRACT The increased presence of senescent cells in different neurological diseases suggests the contribution of senescence in the pathophysiology of neurodegenerative disorders. Microglia can adapt to any type of disturbance of the homeostasis of the central nervous system (CNS) and its altered activity can lead to permanent and unresolvable damage. The aim of this work was to characterize the behavioural phenotype of spared nerve injury (SNI) mice and then associate it to senescence-related mechanisms. In this work we investigated the timing of the onset of anxiety, depression, or memory decline associated with peripheral neuropathic pain, and their correlation with the presence of microglial cellular senescence. SNI mice showed a persistent pain hypersensitivity from 3 days after surgery. 28 days after nerve injury they also developed anxiety, depression, and cognitive impairment. The appearance of these symptoms was coincident to a significant increase of senescence markers, such as β-galactosidase and senescent-associated secretory phenotype (SASP), at microglial level in the spinal cord and hippocampus of SNI animals. These markers were unaltered at previous time points. In murine immortalized microglial cells (BV2) stimulated with LPS 500 ng/mL for 10 days (4h/day) every other day, we observed an increase of β-galactosidase, SASP appearance, a reduction of cell viability and an increase of Senescence-Associated Heterochromatic Foci (SAHF). Therefore, present findings could represent an important step to a better understanding of the pathophysiological cellular mechanisms in comorbidities related to neuropathic pain states.
Collapse
|
208
|
Duran P, Loya-López S, Ran D, Tang C, Calderon-Rivera A, Gomez K, Stratton HJ, Huang S, Xu YM, Wijeratne EMK, Perez-Miller S, Shan Z, Cai S, Gabrielsen AT, Dorame A, Masterson KA, Alsbiei O, Madura CL, Luo G, Moutal A, Streicher J, Zamponi GW, Gunatilaka AAL, Khanna R. The natural product argentatin C attenuates postoperative pain via inhibition of voltage-gated sodium and T-type voltage-gated calcium channels. Br J Pharmacol 2022; 180:1267-1285. [PMID: 36245395 DOI: 10.1111/bph.15974] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 07/07/2022] [Accepted: 07/15/2022] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND AND PURPOSE Postoperative pain occurs in as many as 70% of surgeries performed worldwide. Postoperative pain management still relies on opioids despite their negative consequences, resulting in a public health crisis. Therefore, it is important to develop alternative therapies to treat chronic pain. Natural products derived from medicinal plants are potential sources of novel biologically active compounds for development of safe analgesics. In this study, we screened a library of natural products to identify small molecules that target the activity of voltage-gated sodium and calcium channels that have important roles in nociceptive sensory processing. EXPERIMENTAL APPROACH Fractions derived from the Native American medicinal plant, Parthenium incanum, were assessed using depolarization-evoked calcium influx in rat dorsal root ganglion (DRG) neurons. Further separation of these fractions yielded a cycloartane-type triterpene identified as argentatin C, which was additionally evaluated using whole-cell voltage and current-clamp electrophysiology, and behavioural analysis in a mouse model of postsurgical pain. KEY RESULTS Argentatin C blocked the activity of both voltage-gated sodium and low-voltage-activated (LVA) calcium channels in calcium imaging assays. Docking analysis predicted that argentatin C may bind to NaV 1.7-1.9 and CaV 3.1-3.3 channels. Furthermore, argentatin C decreased Na+ and T-type Ca2+ currents as well as excitability in rat and macaque DRG neurons, and reversed mechanical allodynia in a mouse model of postsurgical pain. CONCLUSION AND IMPLICATIONS These results suggest that the dual effect of argentatin C on voltage-gated sodium and calcium channels supports its potential as a novel treatment for painful conditions.
Collapse
Affiliation(s)
- Paz Duran
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York, USA
| | - Santiago Loya-López
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York, USA
| | - Dongzhi Ran
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, Arizona, USA
| | - Cheng Tang
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York, USA.,NYU Pain Research Center, New York, New York, USA.,Department of Biochemistry and Molecular Biology, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Aida Calderon-Rivera
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York, USA
| | - Kimberly Gomez
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York, USA
| | - Harrison J Stratton
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, Arizona, USA
| | - Sun Huang
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Ya-Ming Xu
- Southwest Center for Natural Products Research, School of Natural Resources and the Environment, College of Agriculture and Life Sciences, The University of Arizona, Tucson, Arizona, USA
| | - E M Kithsiri Wijeratne
- Southwest Center for Natural Products Research, School of Natural Resources and the Environment, College of Agriculture and Life Sciences, The University of Arizona, Tucson, Arizona, USA
| | - Samantha Perez-Miller
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York, USA
| | - Zhiming Shan
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, Arizona, USA
| | - Song Cai
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, Arizona, USA
| | - Anna T Gabrielsen
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, Arizona, USA
| | - Angie Dorame
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, Arizona, USA
| | - Kyleigh A Masterson
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, Arizona, USA
| | - Omar Alsbiei
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, Arizona, USA
| | - Cynthia L Madura
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, Arizona, USA
| | - Guoqin Luo
- Southwest Center for Natural Products Research, School of Natural Resources and the Environment, College of Agriculture and Life Sciences, The University of Arizona, Tucson, Arizona, USA
| | - Aubin Moutal
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, Arizona, USA
| | - John Streicher
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, Arizona, USA
| | - Gerald W Zamponi
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - A A Leslie Gunatilaka
- Southwest Center for Natural Products Research, School of Natural Resources and the Environment, College of Agriculture and Life Sciences, The University of Arizona, Tucson, Arizona, USA
| | - Rajesh Khanna
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York, USA.,NYU Pain Research Center, New York, New York, USA
| |
Collapse
|
209
|
Clark AP, Wei S, Kalola D, Krogh‐Madsen T, Christini DJ. An in silico-in vitro pipeline for drug cardiotoxicity screening identifies ionic pro-arrhythmia mechanisms. Br J Pharmacol 2022; 179:4829-4843. [PMID: 35781252 PMCID: PMC9489646 DOI: 10.1111/bph.15915] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 05/25/2022] [Accepted: 06/24/2022] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND AND PURPOSE Before advancing to clinical trials, new drugs are screened for their pro-arrhythmic potential using a method that is overly conservative and provides limited mechanistic insight. The shortcomings of this approach can lead to the mis-classification of beneficial drugs as pro-arrhythmic. EXPERIMENTAL APPROACH An in silico-in vitro pipeline was developed to circumvent these shortcomings. A computational human induced pluripotent stem cell-derived cardiomyocyte (iPSC-CM) model was used as part of a genetic algorithm to design experiments, specifically electrophysiological voltage clamp (VC) protocols, to identify which of several cardiac ion channels were blocked during in vitro drug studies. Such VC data, along with dynamically clamped action potentials (AP), were acquired from iPSC-CMs before and after treatment with a control solution or a low- (verapamil), intermediate- (cisapride or quinine) or high-risk (quinidine) drug. KEY RESULTS Significant AP prolongation (a pro-arrhythmia marker) was seen in response to quinidine and quinine. The VC protocol identified block of IKr (a source of arrhythmias) by all strong IKr blockers, including cisapride, quinidine and quinine. The protocol also detected block of ICaL by verapamil and Ito by quinidine. Further demonstrating the power of the approach, the VC data uncovered a previously unidentified If block by quinine, which was confirmed with experiments using a HEK-293 expression system and automated patch-clamp. CONCLUSION AND IMPLICATIONS We developed an in silico-in vitro pipeline that simultaneously identifies pro-arrhythmia risk and mechanism of ion channel-blocking drugs. The approach offers a new tool for evaluating cardiotoxicity during preclinical drug screening.
Collapse
Affiliation(s)
| | - Siyu Wei
- Department of Physiology and PharmacologySUNY Downstate Medical CenterBrooklynNew YorkUSA
| | - Darshan Kalola
- Computational Biology Summer ProgramWeill Cornell Medicine & Memorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
| | - Trine Krogh‐Madsen
- Department of Physiology & BiophysicsWeill Cornell MedicineNew YorkNew YorkUSA
- Institute for Computational BiomedicineWeill Cornell MedicineNew YorkNew YorkUSA
| | - David J. Christini
- Department of Biomedical EngineeringCornell UniversityIthacaNew YorkUSA
- Department of Physiology and PharmacologySUNY Downstate Medical CenterBrooklynNew YorkUSA
| |
Collapse
|
210
|
Suzuki Y, Nakagawa S, Endo T, Sotome A, Yuan R, Asano T, Otsuguro S, Maenaka K, Iwasaki N, Kadoya K. High-Throughput Screening Assay Identifies Berberine and Mubritinib as Neuroprotection Drugs for Spinal Cord Injury via Blood-Spinal Cord Barrier Protection. Neurotherapeutics 2022; 19:1976-1991. [PMID: 36178590 PMCID: PMC9723073 DOI: 10.1007/s13311-022-01310-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/19/2022] [Indexed: 12/13/2022] Open
Abstract
Because the breakdown of the blood-brain spinal cord barrier (BBSCB) worsens many central nervous system (CNS) diseases, prevention of BBSCB breakdown has been a major therapeutic target, especially for spinal cord injury (SCI). However, effective drugs that protect BBSCB function have yet to be developed. The purpose of the current study was 1) to develop a high-throughput screening assay (HTSA) to identify candidate drugs to protect BBSCB function, 2) to identify candidate drugs from existing drugs with newly developed HTSA, and 3) to examine the therapeutic effects of candidate drugs on SCI. Our HTSA included a culture of immortalized human brain endothelial cells primed with candidate drugs, stress with H2O2, and evaluation of their viability. A combination of the resazurin-based assay with 0.45 mM H2O2 qualified as a reliable HTSA. Screening of 1,570 existing drugs identified 90 drugs as hit drugs. Through a combination of reproducibility tests, exclusion of drugs inappropriate for clinical translation, and dose dependency tests, berberine, mubritinib, and pioglitazone were identified as a candidate. An in vitro BBSCB functional test revealed that berberine and mubritinib, but not pioglitazone, protected BBSCB from oxygen-glucose deprivation and reoxygenation stress. Additionally, these two drugs minimized BBSCB breakdown 1 day after cervical SCI in mice. Furthermore, berberine and mubritinib reduced neuronal loss and improved gait performance 8 weeks after SCI. Collectively, the current study established a useful HTSA to identify potential neuroprotective drugs by maintaining BBSCB function and demonstrated the neuroprotective effect of berberine and mubritinib after SCI.
Collapse
Affiliation(s)
- Yuki Suzuki
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita 15 jo, Nishi 7 chome, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
| | - Shinsuke Nakagawa
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, 8-19-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Takeshi Endo
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita 15 jo, Nishi 7 chome, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
| | - Akihito Sotome
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita 15 jo, Nishi 7 chome, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
| | - Rufei Yuan
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita 15 jo, Nishi 7 chome, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
| | - Tsuyoshi Asano
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita 15 jo, Nishi 7 chome, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
| | - Satoko Otsuguro
- Center for Research and Education On Drug Discovery, Department of Medical Pharmacology, Hokkaido University, Kita 12 jo, Nishi 6 chome, Kita-ku, Sapporo, Hokkaido, 060-0812, Japan
| | - Katsumi Maenaka
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita 12 jo, Nishi 6 chome, Kita-ku, Sapporo, Hokkaido, 060-0812, Japan
| | - Norimasa Iwasaki
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita 15 jo, Nishi 7 chome, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
| | - Ken Kadoya
- Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Kita 15 jo, Nishi 7 chome, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan.
| |
Collapse
|
211
|
Coppi E, Gibb AJ. Selective block of adenosine A 2A receptors prevents ischaemic-like effects induced by oxygen and glucose deprivation in rat medium spiny neurons. Br J Pharmacol 2022; 179:4844-4856. [PMID: 35817954 PMCID: PMC9796695 DOI: 10.1111/bph.15922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 05/20/2022] [Accepted: 06/26/2022] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND AND PURPOSE Ischaemia is known to cause massive neuronal depolarization, termed anoxic depolarization (AD), due to energy failure and loss of membrane ion gradients. The neuromodulator adenosine accumulates extracellularly during ischaemia and activates four metabotropic receptors: A1 , A2A , A2B and A3 . Striatal medium spiny neurons (MSNs) express high levels of A2A receptors and are particularly vulnerable to ischaemic insults. A2A Receptor blockade reduces acute striatal post-ischaemic damage but the cellular mechanisms involved are still unknown. EXPERIMENTAL APPROACH We performed patch-clamp recordings of MSNs in rat striatal slices subjected to oxygen and glucose deprivation (OGD) to investigate the effects of A2A receptor ligands or ion channel blockers on AD and OGD-induced ionic imbalance, measured as a positive shift in Erev of ramp currents. KEY RESULTS Our data indicate that the A2A receptor antagonist SCH58261 (10 μM) significantly attenuated ionic imbalance and AD appearance in MSNs exposed to OGD. The K+ channel blocker Ba2+ (2 mM) or the Na+ channel blocker tetrodotoxin (1 μM) exacerbated and attenuated, respectively, OGD-induced changes. Spontaneous excitatory post-synaptic current (sEPSC) analysis in MSNs revealed that the A2A receptor agonist CGS21680 (1 μM) prevented OGD-induced decrease of sEPSCs within the first 5 min of the insult, an effect shared by the K+ channel blocker Ba2+ , indicating facilitated glutamate release. CONCLUSION AND IMPLICATIONS Adenosine, released during striatal OGD, activates A2A receptors that may exacerbate OGD-induced damage through K+ channel inhibition. Our results could help to develop A2A receptor-selective therapeutic tools for the treatment of brain ischaemia.
Collapse
Affiliation(s)
- Elisabetta Coppi
- Department of Neuroscience, Psychology, Drug Research and Child HealthUniversity of FlorenceFlorenceItaly
| | - Alasdair J. Gibb
- Department of Neuroscience, Physiology and PharmacologyUniversity College LondonLondonUK
| |
Collapse
|
212
|
Liu JQ, Zhao XT, Qin FY, Zhou JW, Ding F, Zhou G, Zhang XS, Zhang ZH, Li ZB. Isoliquiritigenin mitigates oxidative damage after subarachnoid hemorrhage in vivo and in vitro by regulating Nrf2-dependent Signaling Pathway via Targeting of SIRT1. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 105:154262. [PMID: 35896045 DOI: 10.1016/j.phymed.2022.154262] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 05/31/2022] [Accepted: 06/08/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Oxidative stress is a crucial factor leading to subarachnoid hemorrhage (SAH)-induced early brain injury (EBI). Isoliquiritigenin has been verified as a powerful anti-oxidant in a variety of diseases models and can activate sirtuin 1 and nuclear factor-erythroid 2-related factor 2 (Nrf2) pathways. However, the effects of isoliquiritigenin against EBI after SAH and the underlying mechanisms remain elusive. PURPOSE The primary goal of this study is to verify the therapeutic effects of isoliquiritigenin on EBI after SAH and the possible molecular mechanisms. STUDY DESIGN A prechiasmatic cistern SAH model in rats and a hemoglobin incubation SAH model in primary neurons were established. Isoliquiritigenin was administered after SAH induction. EX527 was employed to inhibit sirtuin 1 activation and ML385 was used to suppress Nrf2 signaling. METHODS In our study, neurological scores, brain edema, biochemical estimation, western blotting, and histopathological study were performed to explore the therapeutic action of isoliquiritigenin against SAH. RESULTS Our data revealed that isoliquiritigenin significantly mitigated oxidative damage after SAH as evidenced by decreased reactive oxygen species overproduction and enhanced intrinsic anti-oxidative system. Concomitant with the reduced oxidative insults, isoliquiritigenin improved neurological function and reduced neuronal death in the early period after SAH. Additionally, isoliquiritigenin administration significantly enhanced Nrf2 and sirtuin 1 expressions. Inhibition of Nrf2 by ML385 reversed the anti-oxidative and neuroprotective effects of isoliquiritigenin against SAH. Moreover, inhibiting sirtuin 1 by EX527 pretreatment suppressed isoliquiritigenin-induced Nrf2-dependent pathway and abated the cerebroprotective effects of isoliquiritigenin. In primary cortical neurons, isoliquiritigenin treatment also ameliorated oxidative insults and repressed neuronal degeneration. The beneficial aspects of isoliquiritigenin were attributed to the promotion of sirtuin 1 and Nrf2 signaling pathways and were counteracted by EX527. CONCLUSION Our findings suggest that isoliquiritigenin exerts cerebroprotective effects against SAH-induced oxidative insults by modulating the Nrf2-mediated anti-oxidant signaling in part through sirtuin 1 activation. Isoliquiritigenin might be a new potential drug candidate for SAH.
Collapse
Affiliation(s)
- Jia-Qiang Liu
- The Translational Research Institute for Neurological Disorders of Wannan Medical College, Department of Neurosurgery, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu 241001, PR China
| | - Xin-Tong Zhao
- The Translational Research Institute for Neurological Disorders of Wannan Medical College, Department of Neurosurgery, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu 241001, PR China
| | - Fei-Yun Qin
- The Translational Research Institute for Neurological Disorders of Wannan Medical College, Department of Neurosurgery, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu 241001, PR China
| | - Jia-Wang Zhou
- The Translational Research Institute for Neurological Disorders of Wannan Medical College, Department of Neurosurgery, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu 241001, PR China
| | - Fei Ding
- The Translational Research Institute for Neurological Disorders of Wannan Medical College, Department of Neurosurgery, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu 241001, PR China
| | - Gang Zhou
- The Translational Research Institute for Neurological Disorders of Wannan Medical College, Department of Neurosurgery, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu 241001, PR China
| | - Xiang-Sheng Zhang
- Department of Neurosurgerya, Beijing Friendship Hospital, Capital Medical University, Beijing 100053, China.
| | - Zi-Huan Zhang
- The Translational Research Institute for Neurological Disorders of Wannan Medical College, Department of Neurosurgery, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu 241001, PR China.
| | - Zhen-Bao Li
- The Translational Research Institute for Neurological Disorders of Wannan Medical College, Department of Neurosurgery, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu 241001, PR China.
| |
Collapse
|
213
|
De Pascali F, Ippolito M, Wolfe E, Komolov KE, Hopfinger N, Lemenze D, Kim N, Armen RS, An SS, Scott CP, Benovic JL. β 2 -Adrenoceptor agonist profiling reveals biased signalling phenotypes for the β 2 -adrenoceptor with possible implications for the treatment of asthma. Br J Pharmacol 2022; 179:4692-4708. [PMID: 35732075 PMCID: PMC9474705 DOI: 10.1111/bph.15900] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 04/08/2022] [Accepted: 04/29/2022] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND AND PURPOSE β-Adrenoceptor agonists relieve airflow obstruction by activating β2 -adrenoceptors, which are G protein-coupled receptors (GPCRs) expressed on human airway smooth muscle (HASM) cells. The currently available β-adrenoceptor agonists are balanced agonists, however, and signal through both the stimulatory G protein (Gs )- and β-arrestin-mediated pathways. While Gs signalling is beneficial and promotes HASM relaxation, β-arrestin activation is associated with reduced Gs efficacy. In this context, biased ligands that selectively promote β2 -adrenoceptor coupling to Gs signalling represent a promising strategy to treat asthma. Here, we examined several β-adrenoceptor agonists to identify Gs -biased ligands devoid of β-arrestin-mediated effects. EXPERIMENTAL APPROACH Gs -biased ligands for the β2 -adrenoceptor were identified by high-throughput screening and then evaluated for Gs interaction, Gi interaction, cAMP production, β-arrestin interaction, GPCR kinase (GRK) phosphorylation of the receptor, receptor trafficking, ERK activation, and functional desensitization of the β2 -adrenoceptor. KEY RESULTS We identified ractopamine, dobutamine, and higenamine as Gs -biased agonists that activate the Gs /cAMP pathway upon β2 -adrenoceptor stimulation while showing minimal Gi or β-arrestin interaction. Furthermore, these compounds did not induce any receptor trafficking and had reduced GRK5-mediated phosphorylation of the β2 -adrenoceptor. Finally, we observed minimal physiological desensitization of the β2 -adrenoceptor in primary HASM cells upon treatment with biased agonists. CONCLUSION AND IMPLICATIONS Our work demonstrates that Gs -biased signalling through the β2 -adrenoceptor may prove to be an effective strategy to promote HASM relaxation in the treatment of asthma. Such biased compounds may also be useful in identifying the molecular mechanisms that determine biased signalling and in design of safer drugs.
Collapse
Affiliation(s)
- Francesco De Pascali
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
- These authors contributed equally
| | - Michael Ippolito
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
- These authors contributed equally
| | - Emily Wolfe
- Rutgers Institute for Translational Medicine and Science, New Brunswick, New Jersey and Department of Pharmacology, Rutgers-Robert Wood Johnson Medical School, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Konstantin E. Komolov
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Nathan Hopfinger
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Douglas Lemenze
- Rutgers Institute for Translational Medicine and Science, New Brunswick, New Jersey and Department of Pharmacology, Rutgers-Robert Wood Johnson Medical School, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Nicholas Kim
- Rutgers Institute for Translational Medicine and Science, New Brunswick, New Jersey and Department of Pharmacology, Rutgers-Robert Wood Johnson Medical School, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Roger S. Armen
- Department of Pharmaceutical Sciences, College of Pharmacy, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Steven S. An
- Rutgers Institute for Translational Medicine and Science, New Brunswick, New Jersey and Department of Pharmacology, Rutgers-Robert Wood Johnson Medical School, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Charles P. Scott
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Jeffrey L. Benovic
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
214
|
Stampelou M, Suchankova A, Tzortzini E, Dhingra L, Barkan K, Lougiakis N, Marakos P, Pouli N, Ladds G, Kolocouris A. Dual A1/A3 Adenosine Receptor Antagonists: Binding Kinetics and Structure-Activity Relationship Studies Using Mutagenesis and Alchemical Binding Free Energy Calculations. J Med Chem 2022; 65:13305-13327. [PMID: 36173355 DOI: 10.1021/acs.jmedchem.2c01123] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Drugs targeting adenosine receptors (AR) can provide treatment for diseases. We report the identification of 7-(phenylamino)-pyrazolo[3,4-c]pyridines L2-L10, A15, and A17 as low-micromolar to low-nanomolar A1R/A3R dual antagonists, with 3-phenyl-5-cyano-7-(trimethoxyphenylamino)-pyrazolo[3,4-c]pyridine (A17) displaying the highest affinity at both receptors with a long residence time of binding, as determined using a NanoBRET-based assay. Two binding orientations of A17 produce stable complexes inside the orthosteric binding area of A1R in molecular dynamics (MD) simulations, and we selected the most plausible orientation based on the agreement with alanine mutagenesis supported by affinity experiments. Interestingly, for drug design purposes, the mutation of L2506.51 to alanine increased the binding affinity of A17 at A1R. We explored the structure-activity relationships against A1R using alchemical binding free energy calculations with the thermodynamic integration coupled with the MD simulation (TI/MD) method, applied on the whole G-protein-coupled receptor-membrane system, which showed a good agreement (r = 0.73) between calculated and experimental relative binding free energies.
Collapse
Affiliation(s)
- Margarita Stampelou
- Laboratory of Medicinal Chemistry, Section of Pharmaceutical Chemistry, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou, 15771 Athens, Greece
| | - Anna Suchankova
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, U.K
| | - Efpraxia Tzortzini
- Laboratory of Medicinal Chemistry, Section of Pharmaceutical Chemistry, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou, 15771 Athens, Greece
| | - Lakshiv Dhingra
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, U.K
| | - Kerry Barkan
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, U.K
| | - Nikolaos Lougiakis
- Laboratory of Medicinal Chemistry, Section of Pharmaceutical Chemistry, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou, 15771 Athens, Greece
| | - Panagiotis Marakos
- Laboratory of Medicinal Chemistry, Section of Pharmaceutical Chemistry, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou, 15771 Athens, Greece
| | - Nicole Pouli
- Laboratory of Medicinal Chemistry, Section of Pharmaceutical Chemistry, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou, 15771 Athens, Greece
| | - Graham Ladds
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, U.K
| | - Antonios Kolocouris
- Laboratory of Medicinal Chemistry, Section of Pharmaceutical Chemistry, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou, 15771 Athens, Greece
| |
Collapse
|
215
|
Kim YA, Gu H, Gwon MG, An HJ, Bae S, Leem J, Jung HJ, Park KK, Lee SJ. Synthetic Non-Coding RNA for Suppressing mTOR Translation to Prevent Renal Fibrosis Related to Autophagy in UUO Mouse Model. Int J Mol Sci 2022; 23:11365. [PMID: 36232665 PMCID: PMC9569483 DOI: 10.3390/ijms231911365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/23/2022] [Accepted: 09/24/2022] [Indexed: 11/16/2022] Open
Abstract
The global burden of chronic kidney disease is increasing, and the majority of these diseases are progressive. Special site-targeted drugs are emerging as alternatives to traditional drugs. Oligonucleotides (ODNs) have been proposed as effective therapeutic tools in specific molecular target therapies for several diseases. We designed ring-type non-coding RNAs (ncRNAs), also called mTOR ODNs to suppress mammalian target rapamycin (mTOR) translation. mTOR signaling is associated with excessive cell proliferation and fibrogenesis. In this study, we examined the effects of mTOR suppression on chronic renal injury. To explore the regulation of fibrosis and inflammation in unilateral ureteral obstruction (UUO)-induced injury, we injected synthesized ODNs via the tail vein of mice. The expression of inflammatory-related markers (interleukin-1β, tumor necrosis factor-α), and that of fibrosis (α-smooth muscle actin, fibronectin), was decreased by synthetic ODNs. Additionally, ODN administration inhibited the expression of autophagy-related markers, microtubule-associated protein light chain 3, Beclin1, and autophagy-related gene 5-12. We confirmed that ring-type ODNs inhibited fibrosis, inflammation, and autophagy in a UUO mouse model. These results suggest that mTOR may be involved in the regulation of autophagy and fibrosis and that regulating mTOR signaling may be a therapeutic strategy against chronic renal injury.
Collapse
Affiliation(s)
- Young-Ah Kim
- Department of Pathology, School of Medicine, Daegu Catholic University, Daegu 42472, Korea
| | - Hyemin Gu
- Department of Pathology, School of Medicine, Daegu Catholic University, Daegu 42472, Korea
| | - Mi-Gyeong Gwon
- Department of Pathology, School of Medicine, Daegu Catholic University, Daegu 42472, Korea
| | - Hyun-Jin An
- Department of Pathology, School of Medicine, Daegu Catholic University, Daegu 42472, Korea
| | - Seongjae Bae
- Department of Pathology, School of Medicine, Daegu Catholic University, Daegu 42472, Korea
| | - Jaechan Leem
- Department of Immunology, School of Medicine, Daegu Catholic University, Daegu 42472, Korea
| | - Hyun Jin Jung
- Department of Urology, School of Medicine, Daegu Catholic University, Daegu 42472, Korea
| | - Kwan-Kyu Park
- Department of Pathology, School of Medicine, Daegu Catholic University, Daegu 42472, Korea
| | - Sun-Jae Lee
- Department of Pathology, School of Medicine, Daegu Catholic University, Daegu 42472, Korea
| |
Collapse
|
216
|
Synthetic Amphipathic β-Sheet Temporin-Derived Peptide with Dual Antibacterial and Anti-Inflammatory Activities. Antibiotics (Basel) 2022; 11:antibiotics11101285. [PMID: 36289944 PMCID: PMC9598925 DOI: 10.3390/antibiotics11101285] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 09/15/2022] [Accepted: 09/19/2022] [Indexed: 11/18/2022] Open
Abstract
Temporin family is one of the largest among antimicrobial peptides (AMPs), which act mainly by penetrating and disrupting the bacterial membranes. To further understand the relationship between the physical-chemical properties and their antimicrobial activity and selectivity, an analogue of Temporin L, [Nle1, dLeu9, dLys10]TL (Nle-Phe-Val-Pro-Trp-Phe-Lys-Phe-dLeu-dLys-Arg-Ile-Leu-CONH2) has been developed in the present work. The design strategy consisted of the addition of a norleucine residue at the N-terminus of the lead peptide sequence, [dLeu9, dLys10]TL, previously developed by our group. This modification promoted an increase of peptide hydrophobicity and, interestingly, more efficient activity against both Gram-positive and Gram-negative strains, without affecting human keratinocytes and red blood cells survival compared to the lead peptide. Thus, this novel compound was subjected to biophysical studies, which showed that the peptide [Nle1, dLeu9, dLys10]TL is unstructured in water, while it adopts β-type conformation in liposomes mimicking bacterial membranes, in contrast to its lead peptide forming α-helical aggregates. After its aggregation in the bacterial membrane, [Nle1, dLeu9, dLys10]TL induced membrane destabilization and deformation. In addition, the increase of peptide hydrophobicity did not cause a loss of anti-inflammatory activity of the peptide [Nle1, dLeu9, dLys10]TL in comparison with its lead peptide. In this study, our results demonstrated that positive net charge, optimum hydrophobic−hydrophilic balance, and chain length remain the most important parameters to be addressed while designing small cationic AMPs.
Collapse
|
217
|
Kuzmenkov AI, Peigneur S, Nasburg JA, Mineev KS, Nikolaev MV, Pinheiro-Junior EL, Arseniev AS, Wulff H, Tytgat J, Vassilevski AA. Apamin structure and pharmacology revisited. Front Pharmacol 2022; 13:977440. [PMID: 36188602 PMCID: PMC9523135 DOI: 10.3389/fphar.2022.977440] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 08/05/2022] [Indexed: 12/02/2022] Open
Abstract
Apamin is often cited as one of the few substances selectively acting on small-conductance Ca2+-activated potassium channels (KCa2). However, published pharmacological and structural data remain controversial. Here, we investigated the molecular pharmacology of apamin by two-electrode voltage-clamp in Xenopus laevis oocytes and patch-clamp in HEK293, COS7, and CHO cells expressing the studied ion channels, as well as in isolated rat brain neurons. The microtitre broth dilution method was used for antimicrobial activity screening. The spatial structure of apamin in aqueous solution was determined by NMR spectroscopy. We tested apamin against 42 ion channels (KCa, KV, NaV, nAChR, ASIC, and others) and confirmed its unique selectivity to KCa2 channels. No antimicrobial activity was detected for apamin against Gram-positive or Gram-negative bacteria. The NMR solution structure of apamin was deposited in the Protein Data Bank. The results presented here demonstrate that apamin is a selective nanomolar or even subnanomolar-affinity KCa2 inhibitor with no significant effects on other molecular targets. The spatial structure as well as ample functional data provided here support the use of apamin as a KCa2-selective pharmacological tool and as a template for drug design.
Collapse
Affiliation(s)
- Alexey I. Kuzmenkov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | | | - Joshua A. Nasburg
- Department of Pharmacology, University of California, Davis, Davis, CA, United States
| | - Konstantin S. Mineev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Moscow Institute of Physics and Technology, Moscow Region, Dolgoprudny, Russia
| | - Maxim V. Nikolaev
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, Saint Petersburg, Russia
| | | | - Alexander S. Arseniev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Moscow Institute of Physics and Technology, Moscow Region, Dolgoprudny, Russia
| | - Heike Wulff
- Department of Pharmacology, University of California, Davis, Davis, CA, United States
| | - Jan Tytgat
- Toxicology and Pharmacology, KU Leuven, Leuven, Belgium
| | - Alexander A. Vassilevski
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Moscow Institute of Physics and Technology, Moscow Region, Dolgoprudny, Russia
- *Correspondence: Alexander A. Vassilevski,
| |
Collapse
|
218
|
Zhang Y, Yang M, Yuan Q, He Q, Ping H, Yang J, Zhang Y, Fu X, Liu J. Piperine ameliorates ischemic stroke-induced brain injury in rats by regulating the PI3K/AKT/mTOR pathway. JOURNAL OF ETHNOPHARMACOLOGY 2022; 295:115309. [PMID: 35597410 DOI: 10.1016/j.jep.2022.115309] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/10/2022] [Accepted: 04/16/2022] [Indexed: 06/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Piperine (PIP), a main active component isolated from Piper nigrum L., exerts neuroprotective effects in a rat model of ischemic stroke (IS). However, studies on the effects of PIP on neuroprotection and autophagy after IS are limited. AIM OF THE STUDY This study aimed to prove the protective effects of PIP against brain IS and elucidate its underlying mechanisms. MATERIALS AND METHODS Specific pathogen-free male Sprague-Dawley rats were selected to establish a permanent middle cerebral artery occlusion model. The experiment was randomly divided into six groups: sham group, model group, PIP intervention group (10, 20, and 30 mg/kg group), and nimodipine group (Nimo group, 12 mg/kg). Neurological function score, postural reflex score, body swing score, balance beam test, and grip strength test were used to detect behavioral changes of rats. The area of cerebral infarction was detected by TTC staining, and the number and morphological changes of neurons were observed by Nissl and HE staining. In addition, the ultrastructure of hippocampal dentate gyrus neurons was observed using a transmission electron microscope. Western blot was used to detect the expression of PI3K/AKT/mTOR signaling pathway proteins and autophagy-related proteins, namely, Beclin1 and LC3, in the hippocampus and cortex. Cell experiments established an in vitro model of oxygen-glucose deprivation (OGD) with the HT22 cell line to verify the mechanism. The experiment was divided into five groups: control group, OGD group, OGD + PIP 20 μg/mL group, OGD + PIP 30 μg/mL group, and OGD + PIP 40 μg/mL group. CCK-8 was used to measure cell activity, and Western blot was used to measure the expression of PI3K/AKT/mTOR signaling pathway proteins and autophagy-related proteins (Beclin1 and LC3). RESULTS Compared with the model group, the neurological function scores, body swing scores, and postural reflex scores of rats in the 10, 20, and 30 mg/kg PIP intervention groups and Nimo groups decreased, whereas the balance beam score and grip test scores increased (all p < 0.05). After 10, 20, and 30 mg/kg PIP and Nimo intervention, the cerebral infarction area of pMCAO rats was reduced (p < 0.01), and Nissl and HE staining results showed that the number of neurons survived in the 30 mg/kg PIP and Nimo intervention groups increased. Cell morphology and structure were significantly improved (p < 0.05). Most of the hippocampal dentate gyrus neurons and their organelles gradually returned to normal in the 30 mg/kg PIP and Nimo intervention groups, with less neuronal damage. The expression levels of p-mTOR, p-AKT, and p-PI3K in the hippocampus and cortex of the 30 mg/kg PIP and Nimo intervention groups decreased, whereas the expression level of PI3K increased (all p < 0.05). In addition, the expression level of autophagy-related proteins, namely, Beclin1 and LC3-II, in the 30 mg/kg PIP and Nimo intervention groups decreased (all p < 0.05). Results of CCK-8 showed that after 1 h of OGD, the 30 and 40 μg/mL PIP intervention groups had higher cell viability than the OGD group (p < 0.01). Western blot results showed that compared with the OGD group, the expression level of p-mTOR, p-AKT, and p-PI3K in the 30 and 40 μg/mL PIP intervention groups decreased, and the expression level of PI3K increased (all p < 0.05). Moreover, the expression level of autophagy-related proteins, namely, Beclin1 and LC3-II, in the 30 and 40 μg/mL PIP intervention groups decreased (all p < 0.05). CONCLUSIONS This study shows that PIP is a potential compound with neuroprotective effects. PIP can inhibit the PI3K/AKT/mTOR pathway and autophagy. Its inhibition of autophagy is possibly related to modulating the PI3K/AKT/mTOR pathway. These findings provide new insights into the use of PIP for the treatment of IS and its underlying mechanism.
Collapse
Affiliation(s)
- Yiwei Zhang
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China
| | - Miao Yang
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China
| | - Qianqian Yuan
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China
| | - Qianxiong He
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China
| | - Honglu Ping
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, China
| | - Jianrong Yang
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China
| | - Yiqiang Zhang
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China
| | - Xueyan Fu
- School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China.
| | - Juan Liu
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China.
| |
Collapse
|
219
|
Heinrich M, Jalil B, Abdel-Tawab M, Echeverria J, Kulić Ž, McGaw LJ, Pezzuto JM, Potterat O, Wang JB. Best Practice in the chemical characterisation of extracts used in pharmacological and toxicological research-The ConPhyMP-Guidelines. Front Pharmacol 2022; 13:953205. [PMID: 36176427 PMCID: PMC9514875 DOI: 10.3389/fphar.2022.953205] [Citation(s) in RCA: 119] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 07/18/2022] [Indexed: 12/22/2022] Open
Abstract
Background: Research on medicinal plants and extracts derived from them differs from studies performed with single compounds. Extracts obtained from plants, algae, fungi, lichens or animals pose some unique challenges: they are multicomponent mixtures of active, partially active and inactive substances, and the activity is often not exerted on a single target. Their composition varies depending on the method of preparation and the plant materials used. This complexity and variability impact the reproducibility and interpretation of pharmacological, toxicological and clinical research. Objectives: This project develops best practice guidelines to ensure reproducibility and accurate interpretations of studies using medicinal plant extracts. The focus is on herbal extracts used in pharmacological, toxicological, and clinical/intervention research. Specifically, the consensus-based statement focuses on defining requirements for: 1) Describing the plant material/herbal substances, herbal extracts and herbal medicinal products used in these studies, and 2) Conducting and reporting the phytochemical analysis of the plant extracts used in these studies in a reproducible and transparent way. The process and methods: We developed the guidelines through the following process: 1) The distinction between the three main types of extracts (extract types A, B, and C), initially conceptualised by the lead author (MH), led the development of the project as such; 2) A survey among researchers of medicinal plants to gather global perspectives, opportunities, and overarching challenges faced in characterising medicinal plant extracts under different laboratory infrastructures. The survey responses were central to developing the guidelines and were reviewed by the core group; 3) A core group of 9 experts met monthly to develop the guidelines through a Delphi process; and. 4) The final draft guidelines, endorsed by the core group, were also distributed for feedback and approval to an extended advisory group of 20 experts, including many journal editors. Outcome: The primary outcome is the "Consensus statement on the Phytochemical Characterisation of Medicinal Plant extracts" (ConPhyMP) which defines the best practice for reporting the starting plant materials and the chemical methods recommended for defining the chemical compositions of the plant extracts used in such studies. The checklist is intended to be an orientation for authors in medicinal plant research as well as peer reviewers and editors assessing such research for publication.
Collapse
Affiliation(s)
- Michael Heinrich
- Pharmacognosy and Phytotherapy, UCL School of Pharmacy, London, United Kingdom
| | - Banaz Jalil
- Pharmacognosy and Phytotherapy, UCL School of Pharmacy, London, United Kingdom
| | - Mona Abdel-Tawab
- Central Laboratory of German Pharmacists, Eschborn, Germany/Institute of Pharmaceutical Chemistry, Johann-Wolfgang-Goethe University, Frankfurt, Germany
| | - Javier Echeverria
- Departamento de Ciencias del Ambiente, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Žarko Kulić
- Preclinical Research and Development, Dr. Willmar Schwabe GmbH & Co. KG, Karlsruhe, Germany
| | - Lyndy J. McGaw
- Phytomedicine Programme, Department of Paraclinical Sciences, Faculty of Veterinary Science, University of Pretoria, Pretoria, South Africa
| | - John M. Pezzuto
- College of Pharmacy and Health Sciences, Western New England University, Springfield, MA, United States
| | - Olivier Potterat
- Division of Pharmaceutical Biology, University of Basel, Basel, Switzerland
| | - Jia-Bo Wang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| |
Collapse
|
220
|
Permixon®, hexane-extracted Serenoa repens, inhibits human prostate and bladder smooth muscle contraction and exerts growth-related functions in human prostate stromal cells. Life Sci 2022; 308:120931. [PMID: 36084760 DOI: 10.1016/j.lfs.2022.120931] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/25/2022] [Accepted: 09/01/2022] [Indexed: 11/24/2022]
Abstract
AIMS Recently, the European Association of Urology recommended hexane-extracted fruit of Serenoa repens (HESr) in their guidelines on management of non-neurogenic male lower urinary tracts symptoms (LUTS). Despite previously lacking recommendations, Permixon® is the most investigated HESr in clinical trials, where it proved effective for male LUTS. In contrast, underlying mechanisms were rarely addressed and are only marginally understood. We therefore investigated effects of Permixon® on human prostate and detrusor smooth muscle contraction and on growth-related functions in prostate stromal cells. MAIN METHODS Permixon® capsules were dissolved using n-hexane. Contractions of human prostate and detrusor tissues were induced in organ bath. Proliferation (EdU assay), growth (colony formation), apoptosis and cell death (flow cytometry), viability (CCK-8) and actin organization (phalloidin staining) were studied in cultured human prostate stromal cells (WPMY-1). KEY FINDINGS Permixon® inhibited α1-adrenergic and thromboxane-induced contractions in prostate tissues, and methacholine-and thromboxane-induced contractions in detrusor tissues. Endothelin-1-induced contractions were not inhibited. Neurogenic contractions were inhibited in both tissues in a concentration-dependent manner. In WPMY-1 cells, Permixon® caused concentration-dependent breakdown of actin polymerization, inhibited colony formation, reduced cell viability, and proliferation, without showing cytotoxic or pro-apoptotic effects. SIGNIFICANCE Our results provide a novel basis that allows, for the first time, to fully explain the ubiquitous beneficial effects of HESr in clinical trials. HESr may inhibit at least neurogenic, α1-adrenergic and thromboxane-induced smooth muscle contraction in the prostate and detrusor, and in parallel, prostate stromal cell growth. Together, this may explain symptom improvements by Permixon® in previous clinical trials.
Collapse
|
221
|
Caffino L, Mottarlini F, Targa G, Verheij MMM, Homberg J, Fumagalli F. Long access to cocaine self-administration dysregulates the glutamate synapse in the nucleus accumbens core of serotonin transporter knockout rats. Br J Pharmacol 2022; 179:4254-4264. [PMID: 33880773 PMCID: PMC9544393 DOI: 10.1111/bph.15496] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 03/15/2021] [Accepted: 04/08/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND AND PURPOSE It is well established that the nucleus accumbens and glutamate play a critical role in the motivation to take drugs of abuse. We have previously demonstrated that rats with ablation of the serotonin (5-HT) transporter (SERT-/- rats) show increased cocaine intake reminiscent of compulsivity. EXPERIMENTAL APPROACH By comparing SERT-/- to SERT+/+ rats, we set out to explore whether SERT deletion influences glutamate neurotransmission under control conditions as well as after short access (1 h/session) or long access (6 h/session) to cocaine self-administration. KEY RESULTS Rats were killed at 24 h after the final self-administration session for ex vivo molecular analyses of the glutamate system (vesicular and glial transporters, post-synaptic subunits of NMDA and AMPA receptors and their related scaffolding proteins). Such analyses were undertaken in the nucleus accumbens core. In cocaine-naïve animals, SERT deletion evoked widespread abnormalities in markers of glutamatergic neurotransmission that, overall, indicate a reduction of glutamate signalling. These results suggest that 5-HT is pivotal for the maintenance of accumbal glutamate homeostasis. We also found that SERT deletion altered glutamate homeostasis mainly after long access, but not short access, to cocaine. CONCLUSION AND IMPLICATIONS Our findings reveal that SERT deletion may sensitize the glutamatergic synapses of the nucleus accumbens core to the long access but not short access, intake of cocaine. LINKED ARTICLES This article is part of a themed issue on New discoveries and perspectives in mental and pain disorders. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v179.17/issuetoc.
Collapse
Affiliation(s)
- Lucia Caffino
- Department of Pharmacological and Biomolecular SciencesUniversità degli Studi di MilanoMilanItaly
| | - Francesca Mottarlini
- Department of Pharmacological and Biomolecular SciencesUniversità degli Studi di MilanoMilanItaly
| | - Giorgia Targa
- Department of Pharmacological and Biomolecular SciencesUniversità degli Studi di MilanoMilanItaly
| | - Michel M. M. Verheij
- Department of Cognitive Neuroscience, Division of Molecular Neurogenetics, Donders Institute for Brain, Cognition and BehaviourRadboud University Nijmegen Medical CentreNijmegenThe Netherlands
| | - Judith Homberg
- Department of Cognitive Neuroscience, Division of Molecular Neurogenetics, Donders Institute for Brain, Cognition and BehaviourRadboud University Nijmegen Medical CentreNijmegenThe Netherlands
| | - Fabio Fumagalli
- Department of Pharmacological and Biomolecular SciencesUniversità degli Studi di MilanoMilanItaly
| |
Collapse
|
222
|
Wang L, Yang Z, Wang S, Que Y, Shu X, Wu F, Liu G, Li S, Hu P, Chen H, Shi J, Tong X. Substitution of SERCA2 Cys 674 accelerates aortic aneurysm by inducing endoplasmic reticulum stress and promoting cell apoptosis. Br J Pharmacol 2022; 179:4423-4439. [PMID: 35491240 DOI: 10.1111/bph.15864] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 04/08/2022] [Accepted: 04/10/2022] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE The Cys674 residue (C674) in the sarcoplasmic/endoplasmic reticulum Ca2+ ATPase 2 (SERCA2) is key to maintaining its enzyme activity. The irreversible oxidation of C674 occurs broadly in aortic aneurysms. Substitution of C674 promotes a phenotypic transition of aortic smooth muscle cells (SMCs) and exacerbates angiotensin II-induced aortic aneurysm. However, its underlying mechanism remains enigmatic. EXPERIMENTAL APPROACH Heterozygous SERCA2 C674S knock-in (SKI) mice, in which half of C674 was replaced by serine, were used to mimic partially irreversible oxidation of C674 thiol. The aortas of SKI mice and their littermate wild-type mice under an LDL receptor-deficient background were collected for histological and immunohistochemical analysis. Cultured aortic SMCs were used for protein expression, apoptosis analysis, and cell function studies. KEY RESULTS The substitution of SERCA2 C674 caused endoplasmic reticulum (ER) stress and induced SMC apoptosis. The inhibition of ER stress by 4-phenylbutyric acid (4-PBA) in SKI aortic SMCs decreased the expression of marker proteins for cell apoptosis as well as phenotypic transition, and prevented cell apoptosis, proliferation, migration, and macrophage adhesion to SMCs. 4-PBA also ameliorated angiotensin II-induced aortic aneurysm in SKI mice. CONCLUSIONS AND IMPLICATIONS The irreversible oxidation of SERCA2 C674 promotes the development of aortic aneurysm by inducing ER stress and subsequent SMC apoptosis. Our study illustrates that ER stress caused by oxidative inactivation of C674 is related to the pathogenesis of aortic aneurysm. Therefore, ER stress and SERCA2 are potential therapeutic targets for treating aortic aneurysm.
Collapse
Affiliation(s)
- Langtao Wang
- School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Zhen Yang
- School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Sai Wang
- School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Yumei Que
- School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Xi Shu
- School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Fuhua Wu
- School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Gang Liu
- School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
- Henan Key Laboratory of Medical Tissue Regeneration, College of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Siqi Li
- School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Pingping Hu
- School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Hao Chen
- Chongqing General Hospital, University of Chinese Academy of Science, Chongqing, China
| | - Jian Shi
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | - Xiaoyong Tong
- School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| |
Collapse
|
223
|
Almquist J, Kuruvilla D, Mai T, Tummala R, White WI, Tang W, Roskos L, Chia YL. Nonlinear Population Pharmacokinetics of Anifrolumab in Healthy Volunteers and Patients With Systemic Lupus Erythematosus. J Clin Pharmacol 2022; 62:1106-1120. [PMID: 35383948 PMCID: PMC9540432 DOI: 10.1002/jcph.2055] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 03/27/2022] [Indexed: 12/03/2022]
Abstract
We characterized the population pharmacokinetics of anifrolumab, a type I interferon receptor-blocking antibody. Pharmacokinetic data were analyzed from the anifrolumab (intravenous [IV], every 4 weeks) arms from 5 clinical trials in patients with systemic lupus erythematosus (SLE) (n = 664) and healthy volunteers (n = 6). Population pharmacokinetic modeling was performed using a 2-compartment model with parallel linear and nonlinear elimination pathways. The impact of covariates (demographics, interferon gene signature [IFNGS, high/low], disease characteristics, renal/hepatic function, SLE medications, and antidrug antibodies) on pharmacokinetics was evaluated. Time-varying clearance (CL) was characterized using an empirical sigmoidal time-dependent function. Anifrolumab exposure increased more than dose-proportionally from 100 to 1000 mg IV every 4 weeks. Based on population pharmacokinetics modeling, the baseline median linear CL was 0.193 L/day in IFNGS-high patients and 0.153 L/day in IFNGS-low/healthy volunteers. After a year, median anifrolumab linear CL decreased by 8.4% from baseline. Body weight and IFNGS were significant pharmacokinetic covariates, whereas age, sex, race, disease activity, SLE medications, and presence of antidrug antibodies had no significant effect on anifrolumab pharmacokinetics. Anifrolumab at a concentration of 300 mg IV every 4 weeks was predicted to be below the lower limit of quantitation in 95% of patients ≈10 weeks after a single dose and ≈16 weeks after stopping dosing at steady state. To conclude, anifrolumab exhibited nonlinear pharmacokinetics and time-varying linear CL; doses ≥300 mg IV every 4 weeks provided sustained anifrolumab concentrations. This study provides further evidence to support the use of anifrolumab 300 mg IV every 4 weeks in patients with moderate to severe SLE.
Collapse
Affiliation(s)
- Joachim Almquist
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences, R&DAstraZenecaGothenburgSweden
| | - Denison Kuruvilla
- BioPharmaceuticals R&DAstraZenecaSouth San FranciscoCaliforniaUSA
- Present address:
GenentechSouth San FranciscoCaliforniaUSA
| | - Tu Mai
- BioPharmaceuticals R&DAstraZenecaSouth San FranciscoCaliforniaUSA
- Present address:
GenentechSouth San FranciscoCaliforniaUSA
| | - Raj Tummala
- Clinical Development, Late Respiratory & Immunology, BioPharmaceuticals R&DAstraZenecaGaithersburgMarylandUSA
| | - Wendy I. White
- Clinical Pharmacology and Quantitative PharmacologyClinical Pharmacology & Safety Sciences, BioPharmaceuticals R&DAstraZenecaGaithersburgMarylandUSA
| | - Weifeng Tang
- Clinical Pharmacology and Quantitative PharmacologyClinical Pharmacology & Safety Sciences, R&DAstraZenecaGaithersburgMarylandUSA
| | - Lorin Roskos
- BioPharmaceuticals R&DAstraZenecaSouth San FranciscoCaliforniaUSA
- Present address:
ExelixisAlamedaCaliforniaUSA
| | - Yen Lin Chia
- BioPharmaceuticals R&DAstraZenecaSouth San FranciscoCaliforniaUSA
- Present address:
SeagenSouth San FranciscoCaliforniaUSA
| |
Collapse
|
224
|
Cristiano C, Cuozzo M, Coretti L, Liguori F, Cimmino F, Turco L, Avagliano C, Aviello G, Mollica M, Lembo F, Russo R. Oral sodium butyrate supplementation ameliorates paclitaxel-induced behavioral and intestinal dysfunction. Biomed Pharmacother 2022; 153:113528. [DOI: 10.1016/j.biopha.2022.113528] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/04/2022] [Accepted: 08/08/2022] [Indexed: 12/16/2022] Open
|
225
|
Ardalan M, Chumak T, Quist A, Hermans E, Hoseinpoor Rafati A, Gravina G, Jabbari Shiadeh SM, Svedin P, Alabaf S, Hansen B, Wegener G, Westberg L, Mallard C. Reelin cells and sex-dependent synaptopathology in autism following postnatal immune activation. Br J Pharmacol 2022; 179:4400-4422. [PMID: 35474185 PMCID: PMC9545289 DOI: 10.1111/bph.15859] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 04/07/2022] [Accepted: 04/10/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Autism spectrum disorders (ASD) are heterogeneous neurodevelopmental disorders with considerably increased risk in male infants born preterm and with neonatal infection. Here, we investigated the role of postnatal immune activation on hippocampal synaptopathology by targeting Reelin+ cells in mice with ASD-like behaviours. EXPERIMENTAL APPROACH C57/Bl6 mouse pups of both sexes received lipopolysaccharide (LPS, 1 mg·kg-1 ) on postnatal day (P) 5. At P45, animal behaviour was examined by marble burying and sociability test, followed by ex vivo brain MRI diffusion kurtosis imaging (DKI). Hippocampal synaptogenesis, number and morphology of Reelin+ cells, and mRNA expression of trans-synaptic genes, including neurexin-3, neuroligin-1, and cell-adhesion molecule nectin-1, were analysed at P12 and P45. KEY RESULTS Social withdrawal and increased stereotypic activities in males were related to increased mean diffusivity on MRI-DKI and overgrowth in hippocampus together with retention of long-thin immature synapses on apical dendrites, decreased volume and number of Reelin+ cells as well as reduced expression of trans-synaptic and cell-adhesion molecules. CONCLUSION AND IMPLICATIONS The study provides new insights into sex-dependent mechanisms that may underlie ASD-like behaviour in males following postnatal immune activation. We identify GABAergic interneurons as core components of dysmaturation of excitatory synapses in the hippocampus following postnatal infection and provide cellular and molecular substrates for the MRI findings with translational value.
Collapse
Affiliation(s)
- Maryam Ardalan
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Department of Clinical Medicine, Translational Neuropsychiatry UnitAarhus UniversityAarhusDenmark
| | - Tetyana Chumak
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Alexandra Quist
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Eva Hermans
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Department of Developmental Origins of Disease, Utrecht Brain Center and Wilhelmina Children's HospitalUtrecht UniversityUtrechtNetherlands
| | - Ali Hoseinpoor Rafati
- Department of Clinical Medicine, Translational Neuropsychiatry UnitAarhus UniversityAarhusDenmark
| | - Giacomo Gravina
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Seyedeh Marziyeh Jabbari Shiadeh
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Department of Clinical Medicine, Translational Neuropsychiatry UnitAarhus UniversityAarhusDenmark
| | - Pernilla Svedin
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Setareh Alabaf
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Brian Hansen
- Department of Clinical Medicine, Center of Functionally Integrative Neuroscience‐SKSAarhus UniversityAarhusDenmark
| | - Gregers Wegener
- Department of Clinical Medicine, Translational Neuropsychiatry UnitAarhus UniversityAarhusDenmark
| | - Lars Westberg
- Department of Pharmacology, Institute of Neuroscience and Physiology, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Carina Mallard
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| |
Collapse
|
226
|
Farkas DJ, Foss JD, Ward SJ, Rawls SM. Kratom alkaloid mitragynine: Inhibition of chemotherapy-induced peripheral neuropathy in mice is dependent on sex and active adrenergic and opioid receptors. IBRO Neurosci Rep 2022; 13:198-206. [PMID: 36093282 PMCID: PMC9459671 DOI: 10.1016/j.ibneur.2022.08.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 08/26/2022] [Indexed: 11/06/2022] Open
Abstract
Mitragynine (MG) is an alkaloid found in Mitragyna speciosa (kratom) that is used as an herbal remedy for pain relief and opioid withdrawal. MG acts at μ-opioid and α-adrenergic receptors in vitro, but the physiological relevance of this activity in the context of neuropathic pain remains unknown. The purpose of the present study was to characterize the effects of MG in a mouse model of chemotherapy-induced peripheral neuropathy (CIPN), and to investigate the potential impact of sex on MG's therapeutic efficacy. Inhibition of oxaliplatin-induced mechanical hypersensitivity was measured following intraperitoneal administration of MG. Both male and female C57BL/6J mice were used to characterize potential sex-differences in MG's therapeutic efficacy. Pharmacological mechanisms of MG were characterized through pretreatment with the opioid and adrenergic antagonists naltrexone, prazosin, yohimbine, and propranolol (1, 2.5, 5 mg/kg). Oxaliplatin produced significant mechanical allodynia of equal magnitude in both male and females, which was dose-dependently attenuated by repeated MG exposure. MG was more potent in males vs females, and the highest dose of MG (10 mg/kg) exhibited greater anti-allodynic efficacy in males. Mechanistically, activity at µ-opioid, α1- and α2-adrenergic receptors, but not β-adrenergic receptors contributed to the effects of MG against oxaliplatin-induced mechanical hypersensitivity. Repeated MG exposure significantly attenuated oxaliplatin-induced mechanical hypersensitivity with greater potency and efficacy in males, which has crucial implications in the context of individualized pain management. The opioid and adrenergic components of MG indicate that it shares pharmacological properties with clinical neuropathic pain treatments.
Collapse
Affiliation(s)
- Daniel J. Farkas
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, 3500 North Broad Street, Philadelphia, PA 19140, USA,Corresponding author.
| | - Jeffery D. Foss
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, 3500 North Broad Street, Philadelphia, PA 19140, USA
| | - Sara Jane Ward
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, 3500 North Broad Street, Philadelphia, PA 19140, USA,Department of Pharmacology, Lewis Katz School of Medicine, Temple University, 3500 North Broad Street, Philadelphia, PA 19140, USA
| | - Scott M. Rawls
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, 3500 North Broad Street, Philadelphia, PA 19140, USA,Department of Pharmacology, Lewis Katz School of Medicine, Temple University, 3500 North Broad Street, Philadelphia, PA 19140, USA
| |
Collapse
|
227
|
Soluble guanylate cyclase stimulator riociguat improves spatial memory in mice via peripheral mechanisms. Neurosci Lett 2022; 788:136840. [PMID: 35985509 DOI: 10.1016/j.neulet.2022.136840] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 07/10/2022] [Accepted: 08/11/2022] [Indexed: 01/02/2023]
Abstract
Soluble guanylate cyclase (sGC) - cyclic guanosine monophosphate (cGMP) signalling is important for healthy memory function and a healthy vascular system. Targeting sGC-cGMP signalling can therefore be a potential strategy to enhance memory processes. sGC can be targeted by using agonists, such as sGC stimulator riociguat. Therefore, this study aimed to target sGC using riociguat to investigate its acute effects on memory function and neuronal plasticity in mice. The effects of riociguat on long-term memory and a biperiden-induced memory deficit model for assessing short-term memory were tested in the object location task, and working memory was tested in the Y-maze continuous alternation task. Pharmacokinetic measurements were performed within brain tissue of mice, and hippocampal plasticity measures were assessed using western blotting. Acute oral administration with a low dose of 0.03 mg/kg riociguat was able to enhance working-, short-, and long-term spatial memory. Under cerebral vasoconstriction higher doses of riociguat were still effective on memory. Pharmacokinetic measurements revealed poor brain penetration of riociguat and its metabolite M-1. Increased activation of VASP was found, while no effects were found on other memory-related hippocampal plasticity measures. Memory enhancing effects of riociguat are most likely regulated by vascular peripheral effects on cGMP signalling. Yet, further research is needed to investigate the possible contribution of hemodynamic or metabolic effects of sGC stimulators on memory performance.
Collapse
|
228
|
7-Chloro-4-(Phenylselanyl) Quinoline Is a Novel Multitarget Therapy to Combat Peripheral Neuropathy and Comorbidities Induced by Paclitaxel in Mice. Mol Neurobiol 2022; 59:6567-6589. [DOI: 10.1007/s12035-022-02991-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 08/07/2022] [Indexed: 10/15/2022]
|
229
|
Elik A, Altunay N. Chemometric approach for the spectrophotometric determination of chloramphenicol in various food matrices: Using natural deep eutectic solvents. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2022; 276:121198. [PMID: 35367727 DOI: 10.1016/j.saa.2022.121198] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/09/2022] [Accepted: 03/24/2022] [Indexed: 06/14/2023]
Abstract
A novel, simple and green temperature controlled-natural deep eutectic solvent emulsification liquid-liquid microextraction (TC-NADES-LLME) coupled with UV-vis spectrophotometry was optimized for preconcentration and measurement of chloramphenicol (CAP) in eggs, milks honeys and chicken meat. Four different NADES were prepared and investigated for the efficient extraction of CAP. The important parameters (pH, NADES-3 vol, Fe(III) amount and extraction temperature) affecting the extraction efficiency of the TC-NADES-LLME procedure were investigated and optimized using a chemometric approach. In this study, Fe(III), NADES-3 and extraction temperature were used as complexing agent, extraction solvent and emulator accelerator, respectively. Using optimized values, the linear range of the TC-NADES-LLME procedure was in the range of 0.1-300 µg L-1 with a coefficient of determination of 0.9991. The detection limit and enhancement factor were 0.03 µg L-1 and 285, respectively. The precision of the method has been confirmed in repeatability and reproducibility studies. Relative standard deviation of these studieswas<4.2 %. The matrix effect was investigated by adding three different CAP concentrations to the selected samples, and the results indicated the low matrix effect of the method. The TC-NADES-LLME procedure was successfully applied to determine and extract CAP in the selected samples.
Collapse
Affiliation(s)
- Adil Elik
- Sivas Cumhuriyet University, Faculty of Science, Department of Chemistry, Sivas, Turkey
| | - Nail Altunay
- Sivas Cumhuriyet University, Faculty of Science, Department of Chemistry, Sivas, Turkey.
| |
Collapse
|
230
|
Somalo-Barranco G, Serra C, Lyons D, Piggins HD, Jockers R, Llebaria A. Design and Validation of the First Family of Photo-Activatable Ligands for Melatonin Receptors. J Med Chem 2022; 65:11229-11240. [PMID: 35930058 PMCID: PMC9421648 DOI: 10.1021/acs.jmedchem.2c00717] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
![]()
Melatonin is a neurohormone released in a circadian manner
with
peak levels at night. Melatonin mediates its effects mainly through
G protein-coupled MT1 and MT2 receptors. Drugs
acting on melatonin receptors are indicated for circadian rhythm-
and sleep-related disorders. Tools to study the activation of these
receptors with high temporal resolution are lacking. Here, we synthesized
a family of light-activatable caged compounds by attaching o-nitrobenzyl (o-NB) or coumarin photocleavable
groups to melatonin indolic nitrogen. All caged compounds showed the
expected decrease in binding affinity for MT1 and MT2. The o-NB derivative MCS-0382 showed the
best uncaging and biological properties, with 250-fold increase in
affinity and potency upon illumination. Generation of melatonin from
MCS-0382 was further demonstrated by its ability to modulate the excitation
of SCN neurons in rat brain slices. MCS-0382 is available to study
melatonin effects in a temporally controlled manner in cellular and
physiological settings.
Collapse
Affiliation(s)
- Gloria Somalo-Barranco
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 Paris, France.,MCS, Laboratory of Medicinal Chemistry & Synthesis, Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), 08034 Barcelona, Spain
| | - Carme Serra
- MCS, Laboratory of Medicinal Chemistry & Synthesis, Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), 08034 Barcelona, Spain.,SIMChem, Synthesis of High Added Value Molecules, Institute of Advanced Chemistry of Catalonia (IQAC-CSIC), 08034 Barcelona, Spain
| | - David Lyons
- School of Physiology, Pharmacology and Neuroscience, Faculty of Life Sciences, University of Bristol, BS8 1TD Bristol, U.K
| | - Hugh D Piggins
- School of Physiology, Pharmacology and Neuroscience, Faculty of Life Sciences, University of Bristol, BS8 1TD Bristol, U.K
| | - Ralf Jockers
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 Paris, France
| | - Amadeu Llebaria
- MCS, Laboratory of Medicinal Chemistry & Synthesis, Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), 08034 Barcelona, Spain
| |
Collapse
|
231
|
Maier M, Olthoff S, Hill K, Zosel C, Magauer T, Wein LA, Schaefer M. KS0365, a novel activator of the transient receptor potential vanilloid 3 (TRPV3) channel, accelerates keratinocyte migration. Br J Pharmacol 2022; 179:5290-5304. [PMID: 35916168 DOI: 10.1111/bph.15937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 06/09/2022] [Accepted: 07/06/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Ca2+ signalling mediated by the thermosensitive, non-selective, Ca2+ -permeable transient receptor potential channel TRPV3 is assumed to play a critical role in regulating several aspects of skin functions, such as keratinocyte proliferation, differentiation, skin barrier formation and wound healing. Studying the function of TRPV3 in skin homeostasis, however, is still constrained by a lack of potent and selective pharmacological modulators of TRPV3. EXPERIMENTAL APPROACH By screening an in-house compound library using fluorometric intracellular Ca2+ assays, we identified two chemically related hits. The more potent and efficient TRPV3 activator KS0365 was further evaluated in fluo-4-assisted Ca2+ assays, different Ca2+ imaging approaches, electrophysiological studies, cytotoxicity and migration assays. KEY RESULTS KS0365 activated recombinant and native mouse TRPV3 more potently and with a higher efficacy compared to 2-APB and did not activate TRPV1, TRPV2 or TRPV4 channels. The activation of TRPV3 by KS0365 super-additively accelerated the EGF-induced keratinocyte migration, which was inhibited by the TRP channel blocker ruthenium red or by siRNA-mediated TRPV3 knockdown. Moreover, KS0365 induced strong Ca2+ responses in migrating front cells and in leading edges of keratinocytes. CONCLUSIONS AND IMPLICATIONS The selective TRPV3 activator KS0365 triggers increases in [Ca2+ ]i with most prominent signals in the leading edge, and accelerates migration of keratinocytes. TRPV3 activators may promote reepithelialization upon skin wounding.
Collapse
Affiliation(s)
- Marion Maier
- Leipzig University, Medical Faculty, Rudolf-Boehm-Institute of Pharmacology and Toxicology, Leipzig, Germany
| | - Stefan Olthoff
- Leipzig University, Medical Faculty, Rudolf-Boehm-Institute of Pharmacology and Toxicology, Leipzig, Germany
| | - Kerstin Hill
- Leipzig University, Medical Faculty, Rudolf-Boehm-Institute of Pharmacology and Toxicology, Leipzig, Germany
| | - Carolin Zosel
- Leipzig University, Medical Faculty, Rudolf-Boehm-Institute of Pharmacology and Toxicology, Leipzig, Germany
| | - Thomas Magauer
- Leopold-Franzens-University Innsbruck, Institute of Organic Chemistry and Center for Molecular Biosciences, Innsbruck, Austria
| | - Lukas Anton Wein
- Leopold-Franzens-University Innsbruck, Institute of Organic Chemistry and Center for Molecular Biosciences, Innsbruck, Austria
| | - Michael Schaefer
- Leipzig University, Medical Faculty, Rudolf-Boehm-Institute of Pharmacology and Toxicology, Leipzig, Germany
| |
Collapse
|
232
|
Ghovanloo MR, Estacion M, Higerd-Rusli GP, Zhao P, Dib-Hajj S, Waxman SG. Inhibition of sodium conductance by cannabigerol contributes to a reduction of dorsal root ganglion neuron excitability. Br J Pharmacol 2022; 179:4010-4030. [PMID: 35297036 DOI: 10.1111/bph.15833] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 03/02/2022] [Accepted: 03/05/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND AND PURPOSE Cannabigerol (CBG), a non-psychotropic phytocannabinoid and a precursor of ∆9 -tetrahydrocannabinol and cannabidiol, has been suggested to act as an analgesic. A previous study reported that CBG (10 μM) blocks voltage-gated sodium (Nav ) currents in CNS neurons, although the underlying mechanism is not well understood. Genetic and functional studies have validated Nav 1.7 channels as an opportune target for analgesic drug development. The effects of CBG on Nav 1.7 channels, which may contribute to its analgesic properties, have not been previously investigated. EXPERIMENTAL APPROACH To determine the effects of CBG on Nav channels, we used stably transfected HEK cells and primary dorsal root ganglion (DRG) neurons to characterize compound effects using experimental and computational techniques. These included patch-clamp, multielectrode array, and action potential modelling. KEY RESULTS CBG is a ~10-fold state-dependent Nav channel inhibitor (KI -KR : ~2-20 μM) with an average Hill-slope of ~2. We determined that, at lower concentrations, CBG predominantly blocks sodium Gmax and slows recovery from inactivation. However, as the concentration is increased, CBG also induces a hyperpolarizing shift in the half-voltage of inactivation. Our modelling and multielectrode array recordings suggest that CBG attenuates DRG excitability. CONCLUSIONS AND IMPLICATIONS Inhibition of Nav 1.7 channels in DRG neurons may underlie CBG-induced neuronal hypoexcitability. As most Nav 1.7 channels are inactivated at the resting membrane potential of DRG neurons, they are more likely to be inhibited by lower CBG concentrations, suggesting functional selectivity against Nav 1.7 channels, compared with other Nav channels (via Gmax block).
Collapse
Affiliation(s)
- Mohammad-Reza Ghovanloo
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA
- Center for Neuroscience and Regeneration Research, Yale University, West Haven, Connecticut, USA
- Neuro-Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, USA
| | - Mark Estacion
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA
- Center for Neuroscience and Regeneration Research, Yale University, West Haven, Connecticut, USA
- Neuro-Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, USA
| | - Grant P Higerd-Rusli
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA
- Center for Neuroscience and Regeneration Research, Yale University, West Haven, Connecticut, USA
- Neuro-Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, USA
| | - Peng Zhao
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA
- Center for Neuroscience and Regeneration Research, Yale University, West Haven, Connecticut, USA
- Neuro-Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, USA
| | - Sulayman Dib-Hajj
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA
- Center for Neuroscience and Regeneration Research, Yale University, West Haven, Connecticut, USA
- Neuro-Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, USA
| | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA
- Center for Neuroscience and Regeneration Research, Yale University, West Haven, Connecticut, USA
- Neuro-Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, USA
| |
Collapse
|
233
|
Curtis MJ, Alexander SPH, Cirino G, George CH, Kendall DA, Insel PA, Izzo AA, Ji Y, Panettieri RA, Patel HH, Sobey CG, Stanford SC, Stanley P, Stefanska B, Stephens GJ, Teixeira MM, Vergnolle N, Ahluwalia A. Planning experiments: Updated guidance on experimental design and analysis and their reporting III. Br J Pharmacol 2022; 179:3907-3913. [PMID: 35673806 DOI: 10.1111/bph.15868] [Citation(s) in RCA: 406] [Impact Index Per Article: 135.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Scientists who plan to publish in British Journal of Pharmacology (BJP) must read this article before undertaking a study. This editorial provides guidance for the design of experiments. We have published previously two guidance documents on experimental design and analysis (Curtis et al., 2015; Curtis et al., 2018). This update clarifies and simplifies the requirements on design and analysis for BJP manuscripts. This editorial also details updated requirements following an audit and discussion on best practice by the BJP editorial board. Explanations for the requirements are provided in the previous articles. Here, we address new issues that have arisen in the course of handling manuscripts and emphasise three aspects of design that continue to present the greatest challenge to authors: randomisation, blinded analysis and balance of group sizes.
Collapse
Affiliation(s)
| | | | | | | | | | - Paul A Insel
- University of California, San Diego, California, USA
| | | | - Yong Ji
- Nanjing Medical University, Nanjing, China
| | | | - Hemal H Patel
- University of California, San Diego, California, USA
| | | | | | | | | | | | | | | | - Amrita Ahluwalia
- William Harvey Research Institute, Queen Mary University of London, London, UK
| |
Collapse
|
234
|
Silva GBAD, Souza DS, Menezes-Filho JER, Silva-Neto JAD, Cruz JDS, Roman-Campos DR, Quintans-Júnior LJ, Vasconcelos CMLD. (-)-Carvone Modulates Intracellular Calcium Signaling with Antiarrhythmic Action in Rat Hearts. Arq Bras Cardiol 2022; 119:294-304. [PMID: 35946691 PMCID: PMC9363060 DOI: 10.36660/abc.20210499] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 12/08/2021] [Indexed: 11/23/2022] Open
Abstract
Fundamento: A (-)-carvona é um monoterpeno encontrado em óleos essenciais com atividade antioxidante e anti-inflamátoria. Objetivos: O objetivo deste estudo foi analisar a propriedade antiarrítmica da (-)-carvona no coração de rato e seus efeitos sobre a sinalização de Ca+2 intracelular. Métodos: Os efeitos da (-)-carvona foram avaliados sobre a contratilidade atrial (0,01 – 4 mM) e ventricular (0,5 mM), e no eletrocardiograma (0,5mM). A fração de encurtamento, a corrente de cálcio do tipo L (ICa,L) e a sinalização de Ca+2 foram medidas no cardiomiócito isolado (0,5 mM). O efeito antiarrítmico foi avaliado no modelo de arritmia induzida por sobrecarga de cálcio (0,5 mM) (n = 5). Um p < 0,05 foi adotado como nível de significância estatística. Resultados: No átrio, a (-)-carvona causou inotropismo negativo de maneira concentração-dependente (EC50 0,44 ± 0,11 mM) e diminuiu o inotropismo positivo induzido pelo CaCl2 (0,1 – 8,0 mM) e BAY K8644 (5 - 500 nM), um agonista de canal de cálcio do tipo L. Em coração isolado, a (-)-carvona (0,5mM) reduziu a contratilidade ventricular em 73% e a frequência cardíaca (em 46%), aumentou o Pri (30,7%, tempo desde o início da onda P até a onda R) e o QTc (9,2%, uma medida de despolarização e repolarização dos ventrículos), sem mudar a duração do complexo QRS. A (-)-carvona diminuiu a fração de encurtamento (61%), a (ICa,L) (79%) e o transiente intracelular de Ca+2 (38%). Além disso, a (-)-carvona apresentou ação antiarrítmica, identificada pela redução do escore de arritmia (85%) e ocorrência de fibrilação ventricular. Conclusão: A (-)-carvona reduz a entrada de Ca+2 através de canais de Ca+2 do tipo L e, assim, diminui a contratilidade cardíaca e o Ca+2 intracelular e apresenta promissora atividade antiarrítmica no coração de ratos.
Collapse
Affiliation(s)
| | | | | | | | - Jader Dos Santos Cruz
- Universidade Federal de Minas Gerais - Instituto de Ciências Biológicas, Belo Horizonte, MG - Brasil
| | | | | | | |
Collapse
|
235
|
Shi Z, Li X, Zhang YM, Zhou YY, Gan XF, Fan QY, He CQ, Shi T, Zhang SY. Constitutive androstane receptor (CAR) mediates pyrene-induced mice liver inflammatory response with increased serum amyloid A proteins and Th17 cells. Br J Pharmacol 2022; 179:5209-5221. [PMID: 35906855 DOI: 10.1111/bph.15934] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 07/12/2022] [Accepted: 07/20/2022] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE The constitutive androstane receptor (CAR), a known xenobiotic sensor, plays an important role in drug metabolism by regulating numerous genes. We previously reported that pyrene, an environmental pollutant, is a CAR activator and induces mouse hepatotoxicity via CAR. Here, we investigate the molecular mechanism of inflammatory response in pyrene-caused mice liver injury. EXPERIMENTAL APPROACH Effects of pyrene on the liver were investigated in wild-type and CAR knockout (KO) mice. Levels of pyrene and its metabolite were analyzed by high performance liquid chromatography (HPLC). KEY RESULTS Serum amyloid A proteins (SAAs) were dramatically induced in the liver and serum of pyrene-exposed wild-type mice. Interleukin 17 (IL-17)-producing helper T cells (Th17 cells) and IL-17 levels were significantly increased in the liver of pyrene-exposed wild-type mice. Hepatic mRNA levels of inflammatory cytokines including IL-1β, IL-6 and TNFα, and serum IL-6 levels were significantly elevated in pyrene-treated wild-type mice. However, the above induction was not observed in CAR KO mice. CONCLUSION AND IMPLICATIONS We demonstrate that CAR plays a crucial role in pyrene-caused mice liver inflammatory response with increased SAAs and Th17 cells. Our results suggest that serum SAAs may be a convenient biomarker for early diagnosis of liver inflammatory response caused by polycyclic aromatic hydrocarbons (PAHs) including pyrene. CAR and Th17 cells may be potential targets for novel therapeutic strategy for xenobiotic-induced liver inflammatory response.
Collapse
Affiliation(s)
- Zhe Shi
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Xue Li
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Yu-Man Zhang
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Yi-Yao Zhou
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Xiu-Feng Gan
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Qiao-Ying Fan
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Chen-Qing He
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Tong Shi
- School of Medicine, Tongji University, Shanghai, China
| | - Shu-Yun Zhang
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, China.,School of Medicine, Taizhou University, Taizhou, China
| |
Collapse
|
236
|
Fouda MA, Mohamed YF, Fernandez R, Ruben PC. Anti-inflammatory effects of cannabidiol against lipopolysaccharides in cardiac sodium channels. Br J Pharmacol 2022; 179:5259-5272. [PMID: 35906756 DOI: 10.1111/bph.15936] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 07/13/2022] [Accepted: 07/24/2022] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND Sepsis, caused by a dysregulated host response to infections, can lead to cardiac arrhythmias. However, the mechanisms underlying sepsis-induced inflammation, and how inflammation provokes cardiac arrhythmias, are not well understood. We hypothesized that CBD may ameliorate lipopolysaccharides (LPS)-induced cardiotoxicity via Toll-like receptor 4 (TLR-4) and cardiac sodium channels (Nav1.5). METHODS AND RESULTS We incubated human immune cells (THP-1 macrophages) with LPS for 24 hours, then extracted the THP-1 incubation media. ELISA assay showed that LPS (1 or 5 μg/ml), in a concentration-dependent manner, or MPLA (TLR-4 agonist, 5 μg/ml) stimulated the THP-1 cells to release inflammatory cytokines (TNF-α and IL-6). Prior incubation (4 hours) with cannabidiol (CBD: 5 μM) or C34 (TLR-4 antagonist: 5 μg/ml) inhibited LPS and MPLA-induced release of both IL-6 and TNF-α. Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CM) were subsequently incubated for 24 hours in the media extracted from THP-1 cells incubated with LPS, MPLA alone, or in combination with CBD or C34. Voltage-clamp experiments showed a right shift in the voltage dependence of Nav1.5 activation, steady state fast inactivation (SSFI), increased persistent current and prolonged in silico action potential duration in hiSPC-CM incubated in the LPS or MPLA-THP-1 media. Co-incubation with CBD or C34 rescued the biophysical dysfunction caused by LPS and MPLA. CONCLUSION Our results suggest that CBD may protect against sepsis-induced inflammation and subsequent arrhythmias through (i) inhibition of the release of inflammatory cytokines, antioxidant and anti-apoptotic effects and/or (ii) direct effect on Nav1.5.
Collapse
Affiliation(s)
- Mohamed A Fouda
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, Canada.,Department of Pharmacology and Toxicology, Alexandria University, Alexandria, Egypt
| | - Yasmine Fathy Mohamed
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, Canada.,Department of Microbiology and Immunology, Alexandria University, Alexandria, Egypt
| | - Rachel Fernandez
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, Canada
| | - Peter C Ruben
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, Canada
| |
Collapse
|
237
|
Uhl FE, Vanherle L, Meissner A. Cystic fibrosis transmembrane regulator correction attenuates heart failure-induced lung inflammation. Front Immunol 2022; 13:928300. [PMID: 35967318 PMCID: PMC9365932 DOI: 10.3389/fimmu.2022.928300] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 07/05/2022] [Indexed: 11/13/2022] Open
Abstract
Heart failure (HF) affects 64 million people worldwide. Despite advancements in prevention and therapy, quality of life remains poor for many HF patients due to associated target organ damage. Pulmonary manifestations of HF are well-established. However, difficulties in the treatment of HF patients with chronic lung phenotypes remain as the underlying patho-mechanistic links are still incompletely understood. Here, we aim to investigate the cystic fibrosis transmembrane regulator (CFTR) involvement in lung inflammation during HF, a concept that may provide new mechanism-based therapies for HF patients with pulmonary complications. In a mouse model of HF, pharmacological CFTR corrector therapy (Lumacaftor (Lum)) was applied systemically or lung-specifically for 2 weeks, and the lungs were analyzed using histology, flow cytometry, western blotting, and qPCR. Experimental HF associated with an apparent lung phenotype characterized by vascular inflammation and remodeling, pronounced tissue inflammation as evidenced by infiltration of pro-inflammatory monocytes, and a reduction of pulmonary CFTR+ cells. Moreover, the elevation of a classically-activated phenotype of non-alveolar macrophages coincided with a cell-specific reduction of CFTR expression. Pharmacological correction of CFTR with Lum mitigated the HF-induced downregulation of pulmonary CFTR expression and increased the proportion of CFTR+ cells in the lung. Lum treatment diminished the HF-associated elevation of classically-activated non-alveolar macrophages, while promoting an alternatively-activated macrophage phenotype within the lungs. Collectively, our data suggest that downregulation of CFTR in the HF lung extends to non-alveolar macrophages with consequences for tissue inflammation and vascular structure. Pharmacological CFTR correction possesses the capacity to alleviate HF-associated lung inflammation.
Collapse
Affiliation(s)
- Franziska E. Uhl
- Department of Experimental Medical Science, Lund University, Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - Lotte Vanherle
- Department of Experimental Medical Science, Lund University, Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - Anja Meissner
- Department of Experimental Medical Science, Lund University, Lund, Sweden
- Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
- Department of Physiology, Institute of Theoretical Medicine, Medical Faculty, University of Augsburg, Augsburg, Germany
- *Correspondence: Anja Meissner,
| |
Collapse
|
238
|
Shelkar GP, Gandhi PJ, Liu J, Dravid SM. Cocaine preference and neuroadaptations are maintained by astrocytic NMDA receptors in the nucleus accumbens. SCIENCE ADVANCES 2022; 8:eabo6574. [PMID: 35867797 PMCID: PMC9307248 DOI: 10.1126/sciadv.abo6574] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 06/09/2022] [Indexed: 06/15/2023]
Abstract
Cocaine-associated memories induce cravings and interfere with the ability of users to cease cocaine use. Reducing the strength of cue-drug memories by facilitating extinction may have therapeutic value for the treatment of cocaine addiction. Here, we demonstrate the expression of GluN1/2A/2C NMDA receptor currents in astrocytes in the nucleus accumbens core. Selective ablation of GluN1 subunit from astrocytes in the nucleus accumbens enhanced extinction of cocaine preference memory but did not affect cocaine conditioning or reinstatement. Repeated cocaine exposure up-regulated GluN2C subunit expression and increased astrocytic NMDA receptor currents. Furthermore, intra-accumbal inhibition of GluN2C/2D-containing receptors and GluN2C subunit deletion facilitated extinction of cocaine memory. Cocaine-induced neuroadaptations including dendritic spine maturation and AMPA receptor recruitment were absent in GluN2C knockout mice. Impaired retention of cocaine preference memory in GluN2C knockout mice was restored by exogenous administration of recombinant glypican 4. Together, these results identify a previously unknown astrocytic GluN2C-containing NMDA receptor mechanism underlying maintenance of cocaine preference memory.
Collapse
Affiliation(s)
- Gajanan P. Shelkar
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, 2500 California Plaza, Omaha, NE, USA
| | - Pauravi J. Gandhi
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, 2500 California Plaza, Omaha, NE, USA
| | - Jinxu Liu
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, 2500 California Plaza, Omaha, NE, USA
| | | |
Collapse
|
239
|
Ballout J, Claßen R, Richter K, Grau V, Diener M. Ionotropic P2X
4
and P2X
7
receptors in the regulation of ion transport across rat colon. Br J Pharmacol 2022; 179:4992-5011. [DOI: 10.1111/bph.15928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 05/18/2022] [Accepted: 07/09/2022] [Indexed: 11/29/2022] Open
Affiliation(s)
- Jasmin Ballout
- Institute for Veterinary Physiology and Biochemistry Justus Liebig University Giessen Germany
| | - Rebecca Claßen
- Institute for Veterinary Physiology and Biochemistry Justus Liebig University Giessen Germany
| | - Katrin Richter
- Laboratory of Experimental Surgery, Departement of General Surgery, German Centre for Lung Research (DZL) Justus Liebig University Giessen Germany
| | - Veronika Grau
- Laboratory of Experimental Surgery, Departement of General Surgery, German Centre for Lung Research (DZL) Justus Liebig University Giessen Germany
| | - Martin Diener
- Institute for Veterinary Physiology and Biochemistry Justus Liebig University Giessen Germany
| |
Collapse
|
240
|
Khalaf MM, Abo-Youssef AM, Malak MN, Hamzawy MA. Novel therapeutic modalities target cell signaling of Renin-Angiotensin system/NF-κB-induced cell cycle arrest and apoptosis in urethane-induced lung cancer in mice: An in vivo study. J Biochem Mol Toxicol 2022; 36:e23162. [PMID: 35822566 DOI: 10.1002/jbt.23162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 05/20/2022] [Accepted: 07/01/2022] [Indexed: 11/07/2022]
Abstract
BACKGROUND Lung cancer has risen to the top of the list of cancer-related deaths worldwide. Aliskiren is a direct renin inhibitor. AIM This study aims to investigate the impact of cell signaling of Renin-Angiotensin system (RAS)/NF-κB on lung cancer by investigating the potential therapeutic effects of aliskiren for lung cancer treatment in urethane-induced lung cancer in mice. METHODS Male BALB/c mice were randomly assigned to one of five treatment groups for 150 days, including (1) normal control; (2) aliskiren (25 mg/kg/i.p) daily, (3) urethane at a dose of 1.5 g/kg (i.p) at Day 1 and 60 (nonsmall cell lung cancer[NSCLC] group) (4) NSCLC mice received carboplatin (15 mg/kg/i.p) every other day for the last 4 successive weeks and (5) NSCLC mice treated with aliskiren daily. Tumor size was determined based on blood sampling, and lungs were isolated for biochemical analysis, western blot analysis assay, and histopathological examination. RESULTS Urethane demonstrated significant changes in all biochemical and molecular parameters and histological patterns. Aliskiren-treated mice had significantly lower levels of NF-κB p65, Bcl-2, cyclin D1, ICAM-1, MMP-2, and Nrf2, with an increase in the catalytic activity of caspase-3 due to its RAS inhibitory mechanism. The combined urethane administration with aliskiren demonstrated a significant improvement in the histopathological examination. CONCLUSION RAS/NF-B cell signaling is a potential therapeutic target for preventing and treating lung adenocarcinoma, evidenced by the fundamental cytotoxic mechanism and attenuation of metastasis and angiogenesis induced by the treatment of NSCLC mice with aliskiren.
Collapse
Affiliation(s)
- Marwa M Khalaf
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Amira M Abo-Youssef
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Marina N Malak
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Mohamed A Hamzawy
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Fayoum University, Fayoum, Egypt
| |
Collapse
|
241
|
Moral-Sanz J, Fernandez-Rojo MA, Colmenarejo G, Kurdyukov S, Brust A, Ragnarsson L, Andersson Å, Vila SF, Cabezas-Sainz P, Wilhelm P, Vela-Sebastian A, Fernández-Carrasco I, Chin YKY, López-Mancheño Y, Smallwood TB, Clark RJ, Fry BG, King GF, Ramm GA, Alewood PF, Lewis RJ, Mulvenna JP, Boyle GM, Sanchez LE, Neely GG, Miles JJ, Ikonomopoulou MP. The structural conformation of the tachykinin domain drives the anti-tumoral activity of an octopus peptide in melanoma BRAF V600E. Br J Pharmacol 2022; 179:4878-4896. [PMID: 35818835 DOI: 10.1111/bph.15923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 04/22/2022] [Accepted: 05/05/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Over the past decades, targeted therapies and immunotherapy have vastly improved survival and reduced the morbidity of patients with BRAF-mutated melanoma. However, drug resistance and relapse hinder overall success. Therefore, there is an urgent need for novel compounds with therapeutic efficacy against BRAF- melanoma. This prompted us to investigate the antiproliferative profile of a tachykinin-peptide from the Octopus kaurna, Octpep-1 in melanoma. EXPERIMENTAL APPROACH We evaluated the cytotoxicity of Octpep-1 by MTT assay. Mechanistic insights on viability and cellular damage caused by Octpep-1 were gained via flow cytometry and bioenergetics. Structural and pharmacological characterization was conducted by molecular modelling, molecular biology, CRISPR/Cas9 technology, high-throughput mRNA and calcium flux analysis. In-vivo efficacy was validated in two independent xerograph animal models (mice and zebrafish). KEY RESULTS Octpep-1 selectively reduced the proliferative capacity of human melanoma BRAFV600E -mutated cells with minimal effects on fibroblasts. In melanoma-treated cells, Octpep-1 increased ROS with unaltered mitochondrial membrane potential and promoted non-mitochondrial and mitochondrial respiration with inefficient ATP coupling. Despite similarities with tachykinin peptides, knock-out or pharmacological blockade of tachykinin receptors suggested that Octpep-1 acts via a tachykinin-independent mechanism. Molecular modelling revealed that the cytotoxicity of Octpep-1 depends upon the α-helix and polyproline conformation in the C-terminal region of the peptide. Indeed, a truncated form of the C-terminal end of Octpep-1 displayed enhanced potency and efficacy against melanoma. Octpep-1 reduced the progression of tumors in xenograft melanoma mice and zebrafish, confirming its therapeutic potential in human BRAF-mutated melanoma. CONCLUSION AND IMPLICATIONS We unravel the intrinsic anti-tumoral properties of a tachykinin peptide, possessing a pharmacology independent of tachykinin-receptors. This peptide mediates the selective cytotoxicity in BRAF-mutated melanoma in-vitro and prevents tumor progression in-vivo, providing the foundation for a potential therapy against melanoma.
Collapse
Affiliation(s)
- Javier Moral-Sanz
- Translational Venomics Group, Madrid Institute for Advanced Studies in Food, Madrid, Spain
| | - Manuel A Fernandez-Rojo
- Hepatic Regenerative Medicine Group, Madrid Institute for Advanced Studies in Food, Madrid, Spain.,Hepatic Fibrosis Group, Department of Cell and Molecular Biology, QIMR Berghofer Medical Research Institute, Queensland, Australia.,Diamantina Institute, The University of Queensland, St. Lucia, QLD, Australia
| | - Gonzalo Colmenarejo
- Biostatistics & Bioinformatics Unit, Madrid Institute for Advances Studies in Food, Madrid, Spain
| | - Sergey Kurdyukov
- Dr. John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre, Centenary Institute, and School of Life and Environmental Sciences, University of Sydney, Camperdown, NSW, Australia
| | - Andreas Brust
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
| | - Lotten Ragnarsson
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
| | - Åsa Andersson
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
| | - Sabela F Vila
- Translational Venomics Group, Madrid Institute for Advanced Studies in Food, Madrid, Spain.,Department of Zoology, Genetics and Physical Anthropology, University of Santiago de Compostela, Lugo, Spain
| | - Pablo Cabezas-Sainz
- Department of Zoology, Genetics and Physical Anthropology, University of Santiago de Compostela, Lugo, Spain
| | - Patrick Wilhelm
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
| | - Ana Vela-Sebastian
- Translational Venomics Group, Madrid Institute for Advanced Studies in Food, Madrid, Spain
| | | | - Yanni K Y Chin
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia.,Centre for Advanced Imaging, The University of Queensland, St Lucia, QLD, Australia
| | - Yaiza López-Mancheño
- Hepatic Regenerative Medicine Group, Madrid Institute for Advanced Studies in Food, Madrid, Spain
| | - Taylor B Smallwood
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD, Australia
| | - Richard J Clark
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia.,School of Biomedical Sciences, The University of Queensland, St Lucia, QLD, Australia
| | - Bryan G Fry
- School of Biological Sciences, The University of Queensland, St Lucia, QLD, Australia
| | - Glenn F King
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia.,Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, QLD, Australia
| | - Grant A Ramm
- Hepatic Fibrosis Group, Department of Cell and Molecular Biology, QIMR Berghofer Medical Research Institute, Queensland, Australia.,Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Paul F Alewood
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
| | - Richard J Lewis
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia
| | - Jason P Mulvenna
- Infectious Diseases Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Glen M Boyle
- Department of Cell and Molecular Biology, Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Laura E Sanchez
- Department of Zoology, Genetics and Physical Anthropology, University of Santiago de Compostela, Lugo, Spain
| | - G Gregory Neely
- Dr. John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre, Centenary Institute, and School of Life and Environmental Sciences, University of Sydney, Camperdown, NSW, Australia
| | - John J Miles
- Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.,James Cook University, Centre for Biodiscovery and Molecular Development of Therapeutics and Centre for Biosecurity in Tropical Infectious Diseases, Cairns, Australia.,The Australian Institute of Tropical Health and Medicine (AITHM), James Cook University, Cairns, QLD, Australia.,Centre for Molecular Therapeutics, James Cook University, Cairns, QLD, Australia.,Centre for Tropical Bioinformatics and Molecular Biology, James Cook University, Cairns, QLD, Australia
| | - Maria P Ikonomopoulou
- Translational Venomics Group, Madrid Institute for Advanced Studies in Food, Madrid, Spain.,Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, Australia.,Department of Cell and Molecular Biology, Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| |
Collapse
|
242
|
Huang X, Lin X, Wang L, Xie Y, Que Y, Li S, Hu P, Tong X. Substitution of SERCA2 Cys 674 aggravates cardiac fibrosis by promoting the transformation of cardiac fibroblasts to cardiac myofibroblasts. Biochem Pharmacol 2022; 203:115164. [PMID: 35809651 DOI: 10.1016/j.bcp.2022.115164] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 06/28/2022] [Accepted: 06/28/2022] [Indexed: 11/02/2022]
Abstract
Sarcoplasmic/endoplasmic reticulum Ca2+ ATPase 2 (SERCA2) is vital to maintain intracellular calcium homeostasis, and its redox Cys674 (C674) is the key to regulating activity. Our goal was to investigate whether the redox state of SERCA2 C674 is critical for cardiac fibrosis and the mechanisms involved. Heterozygous SERCA2 C674S knock-in (SKI) mice, in which half of C674 was substituted by serine, were used to mimic the partial loss of the reactive C674 thiol in pathological conditions. In cardiac fibroblasts, the substitution of C674 thiol increased Ca2+ levels in cytoplasm and mitochondria, and intracellular ROS levels, and activated calcineurin/nuclear factor of activated T-lymphocytes (NFAT) pathway, increased the protein expression of profibrotic factors TGF beta 1 (TGF-β1), alpha smooth muscle actin, collagen I and collagen III, and promoted the transformation of cardiac fibroblasts to cardiac myofibroblasts, which could be reversed by calcineurin/NFAT inhibitor, SERCA2 agonist, or ROS scavenger. Activation of SERCA2 or scavenging ROS is beneficial to alleviate cardiac fibrosis caused by the substitution of C674. In conclusion, the partial loss of the reactive C674 thiol in the SERCA2 exacerbates cardiac fibrosis by activating the calcineurin/NFAT/TGF-β1 pathway to promote the transformation of cardiac fibroblasts to cardiac myofibroblasts, which highlights the importance of C674 redox state in maintaining the homeostasis of cardiac fibroblasts. SERCA2 is a potential therapeutic target for the treatment of cardiac fibrosis.
Collapse
Affiliation(s)
- Xiaoyang Huang
- School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Xiaojuan Lin
- School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Langtao Wang
- School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Yufei Xie
- School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Yumei Que
- School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Siqi Li
- School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Pingping Hu
- School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Xiaoyong Tong
- School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China.
| |
Collapse
|
243
|
Hu XQ, Song R, Dasgupta C, Romero M, Juarez R, Hanson J, Blood AB, Wilson SM, Zhang L. MicroRNA-210-mediated mitochondrial reactive oxygen species confer hypoxia-induced suppression of spontaneous transient outward currents in ovine uterine arteries. Br J Pharmacol 2022; 179:4640-4654. [PMID: 35776536 PMCID: PMC9474621 DOI: 10.1111/bph.15914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 05/26/2022] [Accepted: 06/22/2022] [Indexed: 12/05/2022] Open
Abstract
Background and Purpose Hypoxia during pregnancy is associated with increased uterine vascular resistance and elevated blood pressure both in women and female sheep. A previous study demonstrated a causal role of microRNA‐210 (miR‐210) in gestational hypoxia‐induced suppression of Ca2+ sparks/spontaneous transient outward currents (STOCs) in ovine uterine arteries, but the underlying mechanisms remain undetermined. We tested the hypothesis that miR‐210 perturbs mitochondrial metabolism and increases mitochondrial reactive oxygen species (mtROS) that confer hypoxia‐induced suppression of STOCs in uterine arteries. Experimental Approach Resistance‐sized uterine arteries were isolated from near‐term pregnant sheep and were treated ex vivo in normoxia and hypoxia (10.5% O2) for 48 h. Key Results Hypoxia increased mtROS and suppressed mitochondrial respiration in uterine arteries, which were also produced by miR‐210 mimic to normoxic arteries and blocked by antagomir miR‐210‐LNA in hypoxic arteries. Hypoxia or miR‐210 mimic inhibited Ca2+ sparks/STOCs and increased uterine arterial myogenic tone, which were inhibited by the mitochondria‐targeted antioxidant MitoQ. Hypoxia and miR‐210 down‐regulated iron–sulfur cluster scaffold protein (ISCU) in uterine arteries and knockdown of ISCU via siRNAs suppressed mitochondrial respiration, increased mtROS, and inhibited STOCs. In addition, blockade of mitochondrial electron transport chain with antimycin and rotenone inhibited large‐conductance Ca2+‐activated K+ channels, decreased STOCs and increased uterine arterial myogenic tone. Conclusion and Implications This study demonstrates a novel mechanistic role for the miR‐210‐ISCU‐mtROS axis in inhibiting Ca2+ sparks/STOCs in the maladaptation of uterine arteries and provides new insights into the understanding of mitochondrial perturbations in the pathogenesis of pregnancy complications resulted from hypoxia.
Collapse
Affiliation(s)
- Xiang-Qun Hu
- Lawrence D. Longo MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Rui Song
- Lawrence D. Longo MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Chiranjib Dasgupta
- Lawrence D. Longo MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Monica Romero
- Lawrence D. Longo MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Rucha Juarez
- Lawrence D. Longo MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Jenna Hanson
- Lawrence D. Longo MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Arlin B Blood
- Lawrence D. Longo MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Sean M Wilson
- Lawrence D. Longo MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Lubo Zhang
- Lawrence D. Longo MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA
| |
Collapse
|
244
|
Pesavento S, Bilel S, Murari M, Gottardo R, Arfè R, Tirri M, Panato A, Tagliaro F, Marti M. Zebrafish larvae: A new model to study behavioural effects and metabolism of fentanyl, in comparison to a traditional mice model. MEDICINE, SCIENCE, AND THE LAW 2022; 62:188-198. [PMID: 35040690 DOI: 10.1177/00258024221074568] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
In an effort to find alternatives to study in vivo the so-called New Psychoactive Substances (NPS), the present work was undertaken to investigate the use of zebrafish larvae as animal model in pharmaco-toxicology, providing behavioural and metabolism information. For this purpose, fentanyl, the progenitor of an extremely dangerous group of NPS, was administered at different doses to zebrafish larvae (1, 10, 50, 100 µM) in comparison to mice (0.1, 1, 6, 15 mg/kg), as a well-established animal model. A behavioural assay was performed at the time of the peak effect of fentanyl, showing that the results in larvae are consistent with those observed in mice. On the other hand, several morphological abnormalities (namely yolk sac edema, abnormal pericardial edema, jaw defect and spinal curvature) were found in larvae mostly at high fentanyl doses (50, 100 µM). Larva extract and mice urine were analyzed by using liquid chromatography coupled to high resolution mass spectrometry to identify the metabolic pathways of fentanyl. The main metabolites detected were norfentanyl and hydroxyfentanyl in both the tested models. In conclusion, the present study provides evidence that fentanyl effects on zebrafish larvae and metabolism are similar to rodents and consequently support the hypothesis of using zebrafish larvae as a suitable rapid screening tool to investigate new drugs, and particularly NPS.
Collapse
Affiliation(s)
- S Pesavento
- Unit of Forensic Medicine, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - S Bilel
- Department of Translational Medicine, Section of Legal Medicine and LTTA Centre, 9299University of Ferrara, Italy
| | - M Murari
- Unit of Forensic Medicine, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - R Gottardo
- Unit of Forensic Medicine, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - R Arfè
- Department of Translational Medicine, Section of Legal Medicine and LTTA Centre, 9299University of Ferrara, Italy
| | - M Tirri
- Department of Translational Medicine, Section of Legal Medicine and LTTA Centre, 9299University of Ferrara, Italy
| | - A Panato
- Unit of Forensic Medicine, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - F Tagliaro
- Unit of Forensic Medicine, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
- "World-Class Research Center" Digital biodesign and personalized healthcare", Sechenov First Moscow State Medical University, Moscow, Russia
| | - M Marti
- Department of Translational Medicine, Section of Legal Medicine and LTTA Centre, 9299University of Ferrara, Italy
- Collaborative Center of the National Early Warning System, Department for Anti-Drug Policies, Presidency of the Council of Ministers, Italy
| |
Collapse
|
245
|
Jeong SB, Das R, Kim DH, Lee S, Oh HI, Jo S, Lee Y, Kim J, Park S, Choi DK, Moon UY, Kwon OB, Namkung W, Lee S, Cho BC, Woo J, Seo Y. Anticancer effect of verteporfin on non-small cell lung cancer via downregulation of ANO1. Biomed Pharmacother 2022; 153:113373. [PMID: 35785700 DOI: 10.1016/j.biopha.2022.113373] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 06/17/2022] [Accepted: 06/29/2022] [Indexed: 11/19/2022] Open
Abstract
Anoctamin 1 (ANO1) is a calcium-activated chloride channel found in various cell types and is overexpressed in non-small cell lung cancer (NSCLC), a major cause of cancer-related mortality. With the rising interest in development of druggable compounds for NSCLC, there has been a corresponding rise in interest in ANO1, a novel drug target for NSCLC. However, as ANO1 inhibitors that have been discovered simultaneously exhibit both the functions of an inhibition of ANO1 channel as well as a reduction of ANO1 protein levels, it is unclear which of the two functions directly causes the anticancer effect. In this study, verteporfin, a chemical compound that reduces ANO1 protein levels was identified through high-throughput screening. Verteporfin did not inhibit ANO1-induced chloride secretion but reduced ANO1 protein levels in a dose-dependent manner with an IC50 value of ~300 nM. Moreover, verteporfin inhibited neither P2Y receptor-induced intracellular Ca2+ mobilization nor cystic fibrosis transmembrane conductance regulator (CFTR) channel activity, and molecular docking studies revealed that verteporfin bound to specific sites of ANO1 protein. Confirming that verteporfin reduces ANO1 protein levels, we then investigated the molecular mechanisms involved in its effect on NSCLC cells. Interestingly, verteporfin decreased ANO1 protein levels, the EGFR-STAT3 pathway as well as ANO1 mRNA expression. Verteporfin reduced the viability of ANO1-expressing cells (PC9, and gefitinib-resistant PC9) and induced apoptosis by increasing caspase-3 activity and PARP-1 cleavage. However, it did not affect hERG channel activity. These results show that the anticancer mechanism of verteporfin is caused via the down-regulation of ANO1.
Collapse
Affiliation(s)
- Sung Baek Jeong
- New Drug Development Center, Daegu Gyeongbuk Medical Innovation Foundation, Daegu, the Republic of Korea.
| | - Raju Das
- Department of Physiology, Dongguk University College of Medicine, Gyeongju, the Republic of Korea.
| | - Dong-Hyun Kim
- New Drug Development Center, Daegu Gyeongbuk Medical Innovation Foundation, Daegu, the Republic of Korea.
| | - Sion Lee
- New Drug Development Center, Daegu Gyeongbuk Medical Innovation Foundation, Daegu, the Republic of Korea.
| | - Hye In Oh
- Underwood Division Economics, Underwood International College, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, the Republic of Korea.
| | - Sungwoo Jo
- College of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, the Republic of Korea.
| | - Yechan Lee
- College of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, the Republic of Korea.
| | - Jeongdong Kim
- College of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, the Republic of Korea.
| | - SeonJu Park
- Chuncheon Center, Korea Basic Science Institute (KBSI), Chuncheon 24341, the Republic of Korea.
| | - Dong Kyu Choi
- New Drug Development Center, Daegu Gyeongbuk Medical Innovation Foundation, Daegu, the Republic of Korea.
| | - Uk Yeol Moon
- New Drug Development Center, Daegu Gyeongbuk Medical Innovation Foundation, Daegu, the Republic of Korea.
| | - Oh-Bin Kwon
- New Drug Development Center, Daegu Gyeongbuk Medical Innovation Foundation, Daegu, the Republic of Korea.
| | - Wan Namkung
- College of Pharmacy and Yonsei Institute of Pharmaceutical Sciences, Yonsei University, 85 Songdogwahak-ro, Yeonsu-gu, Incheon 21983, the Republic of Korea; Interdisciplinary Program of Integrated OMICS for Biomedical Science Graduate School, Yonsei University, Seoul 03722, the Republic of Korea.
| | - Sungwoo Lee
- New Drug Development Center, Daegu Gyeongbuk Medical Innovation Foundation, Daegu, the Republic of Korea.
| | - Byoung Chul Cho
- Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, the Republic of Korea.
| | - Joohan Woo
- Department of Physiology, Dongguk University College of Medicine, Gyeongju, the Republic of Korea; Channelopathy Research Center (CRC), Dongguk University College of Medicine, 32 Dongguk-ro, Ilsan Dong-gu, Goyang, Gyeonggi-do 10326, the Republic of Korea.
| | - Yohan Seo
- New Drug Development Center, Daegu Gyeongbuk Medical Innovation Foundation, Daegu, the Republic of Korea.
| |
Collapse
|
246
|
Treatment with the soluble guanylate cyclase activator BAY 60–2770 normalizes bladder function in an in vivo rat model of chronic prostatitis. Eur J Pharmacol 2022; 927:175052. [DOI: 10.1016/j.ejphar.2022.175052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 05/17/2022] [Accepted: 05/19/2022] [Indexed: 12/30/2022]
|
247
|
Wang Y, Tan L, Jiao K, Xue C, Tang Q, Jiang S, Ren Y, Chen H, El-Aziz TMA, Abdelazeem KNM, Yu Y, Zhao F, Zhu MX, Cao Z. Scutellarein Attenuates Atopic Dermatitis by Selectively Inhibiting Transient Receptor Potential Vanilloid 3. Br J Pharmacol 2022; 179:4792-4808. [PMID: 35771623 DOI: 10.1111/bph.15913] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 06/08/2022] [Accepted: 06/23/2022] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE Atopic dermatitis (AD) is one of the most common chronic inflammatory cutaneous diseases with unmet clinical needs. As a common ingredient found in several medicinal herbs with efficacy on cutaneous inflammatory diseases, Scutellarein (Scu) has been shown to possess anti-inflammatory and anti-proliferative activities. We aimed to evaluate the therapeutic efficacy of Scu against AD and its underlying molecular mechanism. EXPERIMENTAL APPROACH Efficacy of Scu on AD was evaluated in 2,4-dinitrofluorobenzene (DNFB) and carvacrol-induced dermatitis mouse models. Cytokine mRNA and serum IgE levels were examined using qPCR and ELISA, respectively. Voltage clamp recordings were used to measure currents mediated by transient receptor potential (TRP) channels. In silico docking, site-direct mutagenesis, and covalent modification were used to explore the binding pocket of Scu on TRPV3. KEY RESULTS Subcutaneous administration of Scu efficaciously suppresses DNFB and carvacrol-induced pruritus, epidermal hyperplasia and skin inflammation in wild type mice but has no additional benefit in Trpv3 knockout mice in the carvacrol model. Scu is a potent and selective TRPV3 channel allosteric negative modulator with an apparent affinity of 1.18 μM. Molecular docking coupled with site-direct mutagenesis and covalent modification of incorporated cysteine residues demonstrate that Scu targets the cavity formed between the pore helix and transmembrane helix S6. Moreover, Scu attenuates endogenous TRPV3 activity in human keratinocytes and inhibits carvacrol-induced proliferative and proinflammatory responses. CONCLUSIONS AND IMPLICATIONS Collectively, these data demonstrate that Scu ameliorates carvacrol-induced skin inflammation by directly inhibiting TRPV3, and TRPV3 represents a viable therapeutic target for AD treatment.
Collapse
Affiliation(s)
- Yujing Wang
- State Key Laboratory of Natural Medicines and Department of TCM Pharmacology, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Liaoxi Tan
- State Key Laboratory of Natural Medicines and Department of TCM Pharmacology, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Kejun Jiao
- State Key Laboratory of Natural Medicines and Department of TCM Pharmacology, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Chu Xue
- State Key Laboratory of Natural Medicines and Department of TCM Pharmacology, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Qinglian Tang
- State Key Laboratory of Natural Medicines and Department of TCM Pharmacology, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Shan Jiang
- State Key Laboratory of Natural Medicines and Department of TCM Pharmacology, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Younan Ren
- State Key Laboratory of Natural Medicines and Department of TCM Pharmacology, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Hao Chen
- Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| | | | - Khalid N M Abdelazeem
- Radiation Biology Research Department, National Centre for Radiation Research and Technology, Atomic Energy Authority, Cairo, Egypt
| | - Ye Yu
- Department of Basic Medicine, School of Basic Medicine and Clinic Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Fang Zhao
- State Key Laboratory of Natural Medicines and Department of TCM Pharmacology, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Michael X Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Zhengyu Cao
- State Key Laboratory of Natural Medicines and Department of TCM Pharmacology, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
248
|
Su H, Mei Y, Luo S, Wu H, He Y, Shiraishi Y, Hu P, Cohen RA, Tong X. Substitution of the SERCA2 Cys 674 reactive thiol accelerates atherosclerosis by inducing endoplasmic reticulum stress and inflammation. Br J Pharmacol 2022; 179:4778-4791. [PMID: 35763220 DOI: 10.1111/bph.15912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 05/31/2022] [Accepted: 06/03/2022] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND AND PURPOSE The cysteine674 (C674) thiol of sarcoplasmic/endoplasmic reticulum Ca2+ ATPase 2 is easily and irreversibly oxidized under atherosclerotic conditions. However, the contribution of the C674 thiol redox status in the development of atherosclerosis remains unclear. Our goal was to elucidate the possible mechanism involved. EXPERIMENTAL APPROACH Heterozygous SERCA2 C674S knock-in mice in which half of the C674 was substituted by serine674 (S674) were used to mimic the removal of the reactive C674 thiol which occurs under pathological conditions. Bone marrow-derived macrophages (BMDMs) and cardiac endothelial cells (ECs) were used for intracellular Ca2+ , macrophage adhesion, and protein expression analysis. The whole aorta and aortic root were isolated for histological analysis. KEY RESULTS Cell culture studies suggest the partial substitution of SERCA2 C674 increased intracellular Ca2+ levels and induced ER stress in both BMDMs and ECs. The release of pro-inflammatory factors and macrophage adhesion increased in SKI BMDMs. In ECs, the overexpression of S674 induced endothelial inflammation and promoted macrophage recruitment. SKI mice developed more severe atherosclerotic plaque and macrophage accumulation. Additionally, 4-phenyl butyric acid (PBA), an ER stress inhibitor, suppressed ER stress and inflammatory responses in BMDMs and ECs, and alleviate atherosclerosis in SKI mice. CONCLUSIONS AND IMPLICATIONS The substitution of SERCA2 C674 thiol accelerates the development of atherosclerosis by inducing ER stress and inflammation. Our findings highlight the importance of SERCA2 C674 redox state in the context of atherosclerosis and open up a novel therapeutic strategy to combat atherosclerosis.
Collapse
Affiliation(s)
- Hang Su
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Yu Mei
- Vascular Biology Section, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Shuangxue Luo
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Haixia Wu
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Yan He
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Yasumasa Shiraishi
- Department of Internal Medicine, Division of Cardiovascular Medicine, National Defense Medical College, Saitama, Japan
| | - Pingping Hu
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, China.,College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Richard A Cohen
- Vascular Biology Section, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Xiaoyong Tong
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| |
Collapse
|
249
|
Li H, Li X, Hao Y, Wu C, Fu Y, Su N, Chen H, Ying B, Wang H, Su L, Cai H, He Q, Cai M, Sun J, Lin J, Scott A, Smith F, Huang X, Jin S. Maresin 1 intervention Reverses Experimental Pulmonary Arterial Hypertension in mice. Br J Pharmacol 2022; 179:5132-5147. [PMID: 35764296 DOI: 10.1111/bph.15906] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 05/24/2022] [Accepted: 06/06/2022] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE Pulmonary arterial hypertension (PAH) is a pulmonary vasculature obstructive disease that leads to right heart failure and death. Maresin 1 is an endogenous lipid mediator known to promote inflammation resolution. However, the effect of Maresin 1 on PAH remains unclear. EXPERIMENTAL APPROACH The serum Maresin 1 concentration was assessed using UPLC. A mouse model of PAH was established by combining the Sugen 5416 injection and hypoxia exposure (SuHx). After treatment with Maresin 1, the right ventricular systolic pressure (RVSP) and right ventricular function were measured by hemodynamic measurement and echocardiography, respectively. Vascular remodeling was evaluated by histological staining. Confocal and western blot were used to test related protein expression. In vitro, cell migration, proliferation and apoptosis assays were performed in primary rat pulmonary artery smooth muscle cells (PASMCs). Western blotting and siRNA transfection were used to clarify the mechanism of Maresin 1. KEY RESULTS Endogenous serum Maresin 1 was decreased in PAH patients and mice. Maresin 1 treatment decreased RVSP and attenuated the right ventricular dysfunction (RVD) in murine PAH model. Maresin 1 reversed abnormal changes in pulmonary vascular remodeling, attenuating endothelial to mesenchymal transformation (EndoMT) and enhancing apoptosis of α-SMA positive cells. Furthermore, Maresin 1 inhibited PASMC proliferation and promoted apoptosis by inhibiting STAT, AKT, ERK and FoxO1 phosphorylation via LGR6. CONCLUSION AND IMPLICATIONS Maresin 1 improved abnormal pulmonary vascular remodeling and right ventricular dysfunction in PAH mice, targeting aberrant PASMC proliferation. This suggests Maresin 1 may have a potent therapeutic effect in vascular disease.
Collapse
Affiliation(s)
- Hui Li
- Department of Anaesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xinyu Li
- Department of Anaesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yu Hao
- Department of Anaesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chenghua Wu
- Department of Anaesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuhao Fu
- Department of Anaesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Nana Su
- Department of Anaesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Houlin Chen
- Department of Anaesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Binyu Ying
- Department of Anaesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Haixing Wang
- Department of Anaesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lihuang Su
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang, China
| | - Haijian Cai
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang, China
| | - Qinlian He
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang, China
| | - Mengsi Cai
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang, China
| | - Junwei Sun
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang, China
| | - Jing Lin
- Department of Anaesthesia, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Aaron Scott
- The Birmingham Acute Care Research (BACR) Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom.,Academic Department of Anaesthesia, Critical Care, Pain and Resuscitation, Birmingham Heartlands Hospital, Heart of England National Health Service Foundation Trust, Birmingham, United Kingdom
| | - Fanggao Smith
- Department of Anaesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.,The Birmingham Acute Care Research (BACR) Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom.,Academic Department of Anaesthesia, Critical Care, Pain and Resuscitation, Birmingham Heartlands Hospital, Heart of England National Health Service Foundation Trust, Birmingham, United Kingdom
| | - Xiaoying Huang
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang, China
| | - Shengwei Jin
- Department of Anaesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
250
|
Gratal P, Mediero A, Lamuedra A, Matamoros-Recio A, Herencia C, Herrero-Beaumont G, Martín-Santamaría S, Largo R. 6-shogaol treatment improves experimental knee OA exerting a pleiotropic effect over immune innate signaling response in chondrocytes. Br J Pharmacol 2022; 179:5089-5108. [PMID: 35760458 DOI: 10.1111/bph.15908] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 04/09/2022] [Accepted: 06/14/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE The pathogenesis of osteoarthritis (OA) implicates a low-grade inflammation associated to the activation of the innate immune system. Toll like receptor (TLR) stimulation triggers the release of inflammatory mediators, which aggravate OA severity. The aim was to study the preventive effect of 6-shogaol (6S), a potential TLR4 inhibitor, on the treatment of experimental knee OA. EXPERIMENTAL APPROACH OA was induced in C57BL6 mice by surgical section of the medial meniscotibial ligament, which received 6S for eight weeks. Cartilage damage, inflammatory mediator presence, and disease markers were assessed in the joint tissues by immunohistochemistry. Computational modelling was used to predict binding modes of 6S into the TLR4/MD2 receptor and its permeability across cellular membranes. Employing LPS-stimulated chondrocytes and MAPK assay, we clarified 6S action mechanisms. KEY RESULTS 6S treatment was able to prevent articular cartilage lesions, synovitis, and the presence of pro-inflammatory mediators and disease markers in OA animals. Molecular modelling studies predicted 6S interaction with the TLR4/MD-2 heterodimer in an antagonist conformation through its binding into the MD-2 pocket. In cell culture, we confirmed that 6S reduced LPS-induced TLR4 inflammatory signaling pathways. Besides, MAPK assay demonstrated that 6S directly inhibits the ERK1/2 phosphorylation activity. CONCLUSION AND IMPLICATIONS 6S evoked a preventive action on cartilage and synovial inflammation in OA mice. 6S effect may take place not only by hindering the interaction between TLR4 ligands and the TLR4/MD-2 complex in chondrocytes, but also through inhibition of ERK phosphorylation, implying a pleiotropic effect on different mediators activated during OA, which proposes it as an attractive drug for OA treatment.
Collapse
Affiliation(s)
- Paula Gratal
- Bone and Joint Research Unit, Service of Rheumatology, IIS-Fundación Jiménez Díaz, Autonomous University of Madrid, Madrid, Spain
| | - Aránzazu Mediero
- Bone and Joint Research Unit, Service of Rheumatology, IIS-Fundación Jiménez Díaz, Autonomous University of Madrid, Madrid, Spain
| | - Ana Lamuedra
- Bone and Joint Research Unit, Service of Rheumatology, IIS-Fundación Jiménez Díaz, Autonomous University of Madrid, Madrid, Spain
| | - Alejandra Matamoros-Recio
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas Margarita Salas, CIB-CSIC, Madrid, Spain
| | - Carmen Herencia
- Bone and Joint Research Unit, Service of Rheumatology, IIS-Fundación Jiménez Díaz, Autonomous University of Madrid, Madrid, Spain
| | - Gabriel Herrero-Beaumont
- Bone and Joint Research Unit, Service of Rheumatology, IIS-Fundación Jiménez Díaz, Autonomous University of Madrid, Madrid, Spain
| | - Sonsoles Martín-Santamaría
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas Margarita Salas, CIB-CSIC, Madrid, Spain
| | - Raquel Largo
- Bone and Joint Research Unit, Service of Rheumatology, IIS-Fundación Jiménez Díaz, Autonomous University of Madrid, Madrid, Spain
| |
Collapse
|