101
|
Chen XY, Dong YC, Yu YY, Jiang M, Bu WJ, Li P, Sun ZJ, Dong DL. Anthelmintic nitazoxanide protects against experimental pulmonary fibrosis. Br J Pharmacol 2023; 180:3008-3023. [PMID: 37428102 DOI: 10.1111/bph.16190] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 06/02/2023] [Accepted: 07/03/2023] [Indexed: 07/11/2023] Open
Abstract
BACKGROUND AND PURPOSE Nitazoxanide is a therapeutic anthelmintic drug. Our previous studies found that nitazoxanide and its metabolite tizoxanide activated adenosine 5'-monophosphate-activated protein kinase (AMPK) and inhibited signal transducer and activator of transcription 3 (STAT3) signals. As AMPK activation and/or STAT3 inhibition are targets for treating pulmonary fibrosis, we hypothesized that nitazoxanide would be effective in experimental pulmonary fibrosis. EXPERIMENTAL APPROACH The mitochondrial oxygen consumption rate of cells was measured by using the high-resolution respirometry system Oxygraph-2K. The mitochondrial membrane potential of cells was evaluated by tetramethyl rhodamine methyl ester (TMRM) staining. The target protein levels were measured by using western blotting. The mice pulmonary fibrosis model was established through intratracheal instillation of bleomycin. The examination of the lung tissues changes were carried out using haematoxylin and eosin (H&E), and Masson staining. KEY RESULTS Nitazoxanide and tizoxanide activated AMPK and inhibited STAT3 signalling in human lung fibroblast cells (MRC-5 cells). Nitazoxanide and tizoxanide inhibited transforming growth factor-β1 (TGF-β1)-induced proliferation and migration of MRC-5 cells, collagen-I and α-smooth muscle cell actin (α-SMA) expression, and collagen-I secretion from MRC-5 cells. Nitazoxanide and tizoxanide inhibited epithelial-mesenchymal transition (EMT) and inhibited TGF-β1-induced Smad2/3 activation in mouse lung epithelial cells (MLE-12 cells). Oral administration of nitazoxanide reduced the bleomycin-induced mice pulmonary fibrosis and, in the established bleomycin-induced mice, pulmonary fibrosis. Delayed nitazoxanide treatment attenuated the fibrosis progression. CONCLUSIONS AND IMPLICATIONS Nitazoxanide improves the bleomycin-induced pulmonary fibrosis in mice, suggesting a potential application of nitazoxanide for pulmonary fibrosis treatment in the clinic.
Collapse
Affiliation(s)
- Xu-Yang Chen
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Yan-Chao Dong
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Yuan-Yuan Yu
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Man Jiang
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Wen-Jie Bu
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Ping Li
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Zhi-Jie Sun
- Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - De-Li Dong
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
- Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
102
|
Andresen H, Pérez‐Ternero C, Robinson J, Dickey DM, Hobbs AJ, Potter LR, Levy FO, Cataliotti A, Moltzau LR. Novel enhancers of guanylyl cyclase-A activity acting via allosteric modulation. Br J Pharmacol 2023; 180:3254-3270. [PMID: 37522273 PMCID: PMC10952227 DOI: 10.1111/bph.16203] [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: 08/26/2022] [Revised: 06/30/2023] [Accepted: 07/11/2023] [Indexed: 08/01/2023] Open
Abstract
BACKGROUND AND PURPOSE Guanylyl cyclase-A (GC-A), activated by endogenous atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP), plays an important role in the regulation of cardiovascular and renal homeostasis and is an attractive drug target. Even though small molecule modulators allow oral administration and longer half-life, drug targeting of GC-A has so far been limited to peptides. Thus, in this study we aimed to develop small molecular activators of GC-A. EXPERIMENTAL APPROACH Hits were identified through high-throughput screening and optimized by in silico design. Cyclic GMP was measured in QBIHEK293A cells expressing GC-A, GC-B or chimerae of the two receptors using AlphaScreen technology. Binding assays were performed in membrane preparations or whole cells using 125 I-ANP. Vasorelaxation was measured in aortic rings isolated from Wistar rats. KEY RESULTS We have identified small molecular allosteric enhancers of GC-A, which enhanced ANP or BNP effects in cellular systems and ANP-induced vasorelaxation in rat aortic rings. The mechanism of action appears novel and not mediated through previously described allosteric binding sites. In addition, the selectivity and activity depend on a single amino acid residue that differs between the two similar receptors GC-A and GC-B. CONCLUSION AND IMPLICATIONS We describe a novel allosteric binding site on GC-A, which can be targeted by small molecules to enhance ANP and BNP effects. These compounds will be valuable tools in further development and proof-of-concept of GC-A enhancement for the potential use in cardiovascular therapy.
Collapse
Affiliation(s)
- Henriette Andresen
- Department of Pharmacology, Institute of Clinical MedicineUniversity of Oslo and Oslo University HospitalOsloNorway
- Institute for Experimental Medical ResearchUniversity of Oslo and Oslo University HospitalOsloNorway
| | - Cristina Pérez‐Ternero
- William Harvey Research Institute, Barts & The London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | - Jerid Robinson
- Department of Biochemistry, Molecular Biology, and BiophysicsUniversity of Minnesota Medical SchoolMinneapolisMinnesotaUSA
| | - Deborah M. Dickey
- Department of Biochemistry, Molecular Biology, and BiophysicsUniversity of Minnesota Medical SchoolMinneapolisMinnesotaUSA
| | - Adrian J. Hobbs
- William Harvey Research Institute, Barts & The London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | - Lincoln R. Potter
- Department of Biochemistry, Molecular Biology, and BiophysicsUniversity of Minnesota Medical SchoolMinneapolisMinnesotaUSA
| | - Finn Olav Levy
- Department of Pharmacology, Institute of Clinical MedicineUniversity of Oslo and Oslo University HospitalOsloNorway
| | - Alessandro Cataliotti
- Institute for Experimental Medical ResearchUniversity of Oslo and Oslo University HospitalOsloNorway
| | - Lise Román Moltzau
- Department of Pharmacology, Institute of Clinical MedicineUniversity of Oslo and Oslo University HospitalOsloNorway
| |
Collapse
|
103
|
Vozella V, Cruz B, Feldman HC, Bullard R, Bianchi PC, Natividad LA, Cravatt BF, Zorrilla EP, Ciccocioppo R, Roberto M. Sexually dimorphic effects of monoacylglycerol lipase inhibitor MJN110 on stress-related behaviour and drinking in Marchigian Sardinian alcohol-preferring rats. Br J Pharmacol 2023; 180:3130-3145. [PMID: 37488777 PMCID: PMC10805956 DOI: 10.1111/bph.16197] [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: 03/25/2023] [Revised: 07/10/2023] [Accepted: 07/11/2023] [Indexed: 07/26/2023] Open
Abstract
BACKGROUND AND PURPOSE The endocannabinoid (eCB) system plays an important homeostatic role in the regulation of stress circuits and has emerged as a therapeutic target to treat stress disorders and alcohol use disorder (AUD). Extensive research has elucidated a role for the eCB anandamide (AEA), but less is known about 2-arachidonoylglycerol (2-AG) mediated signalling. EXPERIMENTAL APPROACH We pharmacologically enhanced eCB signalling by inhibiting the 2-AG metabolizing enzyme, monoacylglycerol lipase (MAGL), in male and female Marchigian Sardinian alcohol-preferring (msP) rats, a model of innate alcohol preference and stress hypersensitivity, and in control Wistar rats. We tested the acute effect of the selective MAGL inhibitor MJN110 in alleviating symptoms of alcohol drinking, anxiety, irritability and fear. KEY RESULTS A single systemic administration of MJN110 increased 2-AG levels in the central amygdala, prelimbic and infralimbic cortex but did not acutely alter alcohol drinking. MAGL inhibition reduced aggressive behaviours in female msPs, and increased defensive behaviours in male msPs, during the irritability test. Moreover, in the novelty-induced hypophagia test, MJN110 selectively enhanced palatable food consumption in females, mitigating stress-induced food suppression. Lastly, msP rats showed increased conditioned fear behaviour compared with Wistar rats, and MJN110 reduced context-associated conditioned fear responses, but not cue-probed fear expression, in male msPs. CONCLUSIONS AND IMPLICATIONS Acute inhibition of MAGL attenuated some stress-related responses in msP rats but not voluntary alcohol drinking. Our results provide new insights into the sex dimorphism documented in stress-induced responses. Sex-specific eCB-based approaches should be considered in the clinical development of therapeutics.
Collapse
Affiliation(s)
- Valentina Vozella
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | - Bryan Cruz
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | - Hannah C. Feldman
- Department of Chemistry, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | - Ryan Bullard
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | - Paula C. Bianchi
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA 92037, USA
- Department of Pharmacology, Universidade Federal de São Paulo - UNIFESP, São Paulo, SP 04023-062, Brazil
| | - Luis A. Natividad
- College of Pharmacy, Division of Pharmacology and Toxicology, The University of Texas at Austin, 107 W. Dean Keeton Street, Austin, TX 78712, USA
| | - Benjamin F. Cravatt
- Department of Chemistry, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | - Eric P. Zorrilla
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | - Roberto Ciccocioppo
- School of Pharmacy, Pharmacology Unit, University of Camerino, Via Madonna delle Carceri 9, Camerino, 62032 Italy
| | - Marisa Roberto
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA 92037, USA
| |
Collapse
|
104
|
Rahmanian-Devin P, Askari VR, Sanei-Far Z, Baradaran Rahimi V, Kamali H, Jaafari MR, Golmohammadzadeh S. Preparation and characterization of solid lipid nanoparticles encapsulated noscapine and evaluation of its protective effects against imiquimod-induced psoriasis-like skin lesions. Biomed Pharmacother 2023; 168:115823. [PMID: 37924792 DOI: 10.1016/j.biopha.2023.115823] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 10/23/2023] [Accepted: 10/31/2023] [Indexed: 11/06/2023] Open
Abstract
Psoriasis is a chronic inflammatory skin disease characterized by thickening the epidermis with erythema, scaling, and proliferation. Noscapine (NOS) has several anti-inflammatory, anti-angiogenic, and anti-fibrotic effects, but its low solubility and large size results in its lower efficacy in the clinic. In this regard, solid lipid nanoparticles (SLN) encapsulated NOS (SLN-NOS) were fabricated using the well-known response surface method based on the central composite design and modified high-shear homogenization and ultrasound method. As a result, Precirol® was selected as the best lipid base for the SLN formulation based on Hildebrand-Hansen solubility parameters, in which SLN-NOS 1 % had the best zeta potential (-35.74 ± 2.59 mV), average particle size (245.66 ± 17 nm), polydispersity index (PDI, 0.226 ± 0.09), high entrapment efficiency (89.77 %), and ICH-based stability results. After 72 h, the SLN-NOS 1 % released 83.23 % and 58.49 % of the NOS at pH 5.8 and 7.4, respectively. Moreover, Franz diffusion cell's results indicated that the skin levels of NOS for SLN and cream formulations were 46.88 % and 13.5 % of the total amount, respectively. Our pharmacological assessments revealed that treatment with SLN-NOS 1 % significantly attenuated clinical parameters, namely ear thickness, length, and psoriasis area and severity index, compared to the IMQ group. Interestingly, SLN-NOS 1 % reduced the levels of interleukin (IL)-17, tumor necrosis factor-α, and transforming growth factor-β, while elevating IL-10, compared to the IMQ group. Histology studies also showed that topical application of SLN-NOS 1 % significantly decreased parakeratosis, hyperkeratosis, acanthosis, and inflammation compared to the IMQ group. Taken together, SLN-NOS 1 % showed a high potential to attenuate skin inflammation.
Collapse
Affiliation(s)
- Pouria Rahmanian-Devin
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vahid Reza Askari
- International UNESCO Center for Health-Related Basic Sciences and Human Nutrition, Mashhad University of Medical Sciences, Mashhad, Iran; Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Zahra Sanei-Far
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vafa Baradaran Rahimi
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Cardiovascular Diseases, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Hossein Kamali
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Shiva Golmohammadzadeh
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
105
|
Sharma H, Reeta KH, Sharma U, Suri V, Singh S. AMPA receptor modulation through sequential treatment with perampanel and aniracetam mitigates post-stroke damage in experimental model of ischemic stroke. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:3529-3545. [PMID: 37231168 DOI: 10.1007/s00210-023-02544-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 05/18/2023] [Indexed: 05/27/2023]
Abstract
The present study evaluates the effect of modulating α-amino-3-hydroxy-5-methyl-4-isoxazole propionate receptor (AMPAR) by inhibiting them in the acute phase and activating them in the sub-acute phase on post-stroke recovery in middle cerebral artery occlusion (MCAo) model of stroke in rats. After 90 min of MCAo, perampanel (an AMPAR antagonist, 1.5 mg/kg i.p) and aniracetam (an AMPA agonist, 50 mg/kg i.p.) were administered for different durations after MCAo. Later, after obtaining the best time point for the antagonist and the agonist treatment protocols, sequential treatment with perampanel and aniracetam were given, and the effect on neurological damage and post stroke recovery were assessed. Perampanel and aniracetam significantly protected MCAo-induced neurological damage and diminished the infarct percentage. Furthermore, treatment with these study drugs improved the motor coordination and grip strength. Sequential treatment with perampanel and aniracetam reduced the infarct percentage as assessed by MRI. Moreover, these compounds diminished the inflammation via reducing the levels of pro-inflammatory cytokines (TNF-α, IL-1β) and increasing the levels of anti-inflammatory cytokine (IL-10) along with reductions in GFAP expression. Moreover, the neuroprotective markers (BDNF and TrkB) were found to be significantly increased. Levels of apoptotic markers (Bax, cleaved-caspase-3; Bcl2 and TUNEL positive cells) and neuronal damage (MAP-2) were normalized with the AMPA antagonist and agonist treatment. Expressions of GluR1 and GluR2 subunits of AMPAR were significantly enhanced with sequential treatment. The present study thus showed that modulation of AMPAR improves neurobehavioral deficits and reduces the infarct percentage through anti-inflammatory, neuroprotective and anti-apoptotic effects.
Collapse
Affiliation(s)
- Himanshu Sharma
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India
| | - K H Reeta
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India.
| | - Uma Sharma
- Department of NMR, All India Institute of Medical Sciences, New Delhi, India
| | - Vaishali Suri
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Surender Singh
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
106
|
Cheng LZ, Huang DL, Tang ZR, Zhang JH, Xiong T, Zhou C, Zhang NX, Fu R, Cheng YX, Wu ZQ. Pharmacological targeting of Axin2 suppresses cell growth and metastasis in colorectal cancer. Br J Pharmacol 2023; 180:3071-3091. [PMID: 37461816 DOI: 10.1111/bph.16193] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 04/26/2023] [Accepted: 07/02/2023] [Indexed: 08/20/2023] Open
Abstract
BACKGROUND AND PURPOSE The scaffold molecule Axin2 is constitutively activated in colorectal cancer (CRC) and functions as a potent promoter of CRC behaviour. Pharmacological targeting of Axin2 may therefore exert a therapeutic effect in patients with CRC. Here, we discovered a potent small-molecule inhibitor of Axin2, based on the mechanism by which Axin2 is regulated post-translationally, and investigated its antitumour effects. EXPERIMENTAL APPROACH Compound discovery and its inhibitory action on Axin2 protein were revealed by microscale thermophoresis, in vitro kinase assay, quantitative kinetic assay, immunoblotting/immunoprecipitation, RT-qPCR and cycloheximide pulse-chase assay. Compound antitumour effects and the underlying mechanisms were evaluated in multiple cell-based assays and mouse models. KEY RESULTS We discovered that glycogen synthase kinase 3β (GSK3β) phosphorylates Axin2 at two consensus motifs and coupled Axin2 phosphorylation to its ubiquitination (mediated by the E3 ligase β-Trcp2) and proteasomal degradation. The binding of Axin2 to GSK3β in CRC cells is faint, which enables most of the Axin2 protein to maintain an unphosphorylated status and thereby permits the cells to preserve high levels of Axin2. Importantly, we identified a small-molecule compound CW85319 that enhances Axin2's interaction with GSK3β via forming a high affinity for Axin2. Treatment of CRC cells with CW85319 enhanced Axin2 binding with GSK3β, thereby promoting Axin2 phosphorylation, subsequent ubiquitination, and degradation. Furthermore, we demonstrated that CW85319 efficiently suppressed Axin2-driven CRC growth and metastasis, without eliciting side toxicity. CONCLUSIONS AND IMPLICATIONS These findings suggest that pharmacological targeting of Axin2 by CW85319 may provide therapeutic benefits against certain human cancers, especially CRC.
Collapse
Affiliation(s)
- Li-Zhi Cheng
- State Key Laboratory of Natural Medicines, Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Dan-Ling Huang
- Institute for Inheritance-Based Innovation of Chinese Medicine, School of Pharmaceutical Sciences, Shenzhen University Health Science Center, Shenzhen, China
| | - Zhang-Rui Tang
- State Key Laboratory of Natural Medicines, Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Jia-Hao Zhang
- State Key Laboratory of Natural Medicines, Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Ting Xiong
- State Key Laboratory of Natural Medicines, Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Chen Zhou
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Nai-Xia Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Rong Fu
- State Key Laboratory of Natural Medicines, Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yong-Xian Cheng
- Institute for Inheritance-Based Innovation of Chinese Medicine, School of Pharmaceutical Sciences, Shenzhen University Health Science Center, Shenzhen, China
| | - Zhao-Qiu Wu
- State Key Laboratory of Natural Medicines, Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
107
|
Gawrys O, Husková Z, Škaroupková P, Honetschlägerová Z, Vaňourková Z, Kikerlová S, Melenovský V, Bačová BS, Sykora M, Táborský M, Červenka L. The treatment with sGC stimulator improves survival of hypertensive rats in response to volume-overload induced by aorto-caval fistula. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:3757-3773. [PMID: 37338578 PMCID: PMC10643302 DOI: 10.1007/s00210-023-02561-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 05/30/2023] [Indexed: 06/21/2023]
Abstract
Heart failure (HF) has been declared as global pandemic and current therapies are still ineffective, especially in patients that develop concurrent cardio-renal syndrome. Considerable attention has been focused on the nitric oxide (NO)/soluble guanylyl cyclase (sGC)/cyclic guanosine monophosphate (cGMP) pathway. In the current study, we aimed to investigate the effectiveness of sGC stimulator (BAY41-8543) with the same mode of action as vericiguat, for the treatment of heart failure (HF) with cardio-renal syndrome. As a model, we chose heterozygous Ren-2 transgenic rats (TGR), with high-output heart failure, induced by aorto-caval fistula (ACF). The rats were subjected into three experimental protocols to evaluate short-term effects of the treatment, impact on blood pressure, and finally the long-term survival lasting 210 days. As control groups, we used hypertensive sham TGR and normotensive sham HanSD rats. We have shown that the sGC stimulator effectively increased the survival of rats with HF in comparison to untreated animals. After 60 days of sGC stimulator treatment, the survival was still 50% compared to 8% in the untreated rats. One-week treatment with sGC stimulator increased the excretion of cGMP in ACF TGR (109 ± 28 nnmol/12 h), but the ACE inhibitor decreased it (-63 ± 21 nnmol/12 h). Moreover, sGC stimulator caused a decrease in SBP, but this effect was only temporary (day 0: 117 ± 3; day 2: 108 ± 1; day 14: 124 ± 2 mmHg). These results support the concept that sGC stimulators might represent a valuable class of drugs to battle heart failure especially with cardio-renal syndrome, but further studies are necessary.
Collapse
Affiliation(s)
- Olga Gawrys
- Experimental Medicine Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic.
| | - Zuzana Husková
- Experimental Medicine Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Petra Škaroupková
- Experimental Medicine Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Zuzana Honetschlägerová
- Experimental Medicine Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Zdeňka Vaňourková
- Experimental Medicine Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Soňa Kikerlová
- Experimental Medicine Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Vojtěch Melenovský
- Department of Cardiology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Barbara Szeiffová Bačová
- Centre of Experimental Medicine, Slovak Academy of Sciences, Institute for Heart Research, Bratislava, Slovakia
| | - Matúš Sykora
- Centre of Experimental Medicine, Slovak Academy of Sciences, Institute for Heart Research, Bratislava, Slovakia
| | - Miloš Táborský
- Department of Internal Medicine I, Cardiology, University Hospital Olomouc and Palacký University, Olomouc, Czech Republic
| | - Luděk Červenka
- Experimental Medicine Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
- Department of Internal Medicine I, Cardiology, University Hospital Olomouc and Palacký University, Olomouc, Czech Republic
| |
Collapse
|
108
|
Patel M, Grimsey NL, Banister SD, Finlay DB, Glass M. Evaluating signaling bias for synthetic cannabinoid receptor agonists at the cannabinoid CB 2 receptor. Pharmacol Res Perspect 2023; 11:e01157. [PMID: 38018694 PMCID: PMC10685394 DOI: 10.1002/prp2.1157] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 11/10/2023] [Indexed: 11/30/2023] Open
Abstract
The rapid structural evolution and emergence of novel synthetic cannabinoid receptor agonists (SCRAs) in the recreational market remains a key public health concern. Despite representing one of the largest classes of new psychoactive substances, pharmacological data on new SCRAs is limited, particularly at the cannabinoid CB2 receptor (CB2 ). Hence, the current study aimed to characterize the molecular pharmacology of a structurally diverse panel of SCRAs at CB2 , including 4-cyano MPP-BUT7AICA, 4F-MDMB-BUTINACA, AMB-FUBINACA, JWH-018, MDMB-4en-PINACA, and XLR-11. The activity of SCRAs was assessed in a battery of in vitro assays in CB2 -expressing HEK 293 cells: G protein activation (Gαi3 and GαoB ), phosphorylation of ERK1/2, and β-arrestin 1/2 translocation. The activity profiles of the ligands were further evaluated using the operational analysis to identify ligand bias. All SCRAs activated the CB2 signaling pathways in a concentration-dependent manner, although with varying potencies and efficacies. Despite the detection of numerous instances of statistically significant bias, compound activities generally appeared only subtly distinct in comparison with the reference ligand, CP55940. In contrast, the phytocannabinoid THC exhibited an activity profile distinct from the SCRAs; most notably in the translocation of β-arrestins. These findings demonstrate that CB2 is able to accommodate a structurally diverse array of SCRAs to generate canonical agonist activity. Further research is required to elucidate whether the activation of CB2 contributes to the toxicity of these compounds.
Collapse
Affiliation(s)
- Monica Patel
- Department of Pharmacology and ToxicologyUniversity of OtagoDunedinNew Zealand
| | - Natasha L. Grimsey
- Department of Pharmacology and Clinical Pharmacology, School of Medical Sciences, Faculty of Medical and Health SciencesUniversity of AucklandAucklandNew Zealand
| | - Samuel D. Banister
- Lambert Initiative for Cannabinoid Therapeutics, Brain and Mind CentreUniversity of SydneyNew South WalesAustralia
- School of Chemistry, Faculty of ScienceUniversity of SydneyNew South WalesAustralia
| | - David B. Finlay
- Department of Pharmacology and ToxicologyUniversity of OtagoDunedinNew Zealand
| | - Michelle Glass
- Department of Pharmacology and ToxicologyUniversity of OtagoDunedinNew Zealand
| |
Collapse
|
109
|
da Silva ALM, Nascimento CP, Azevedo JEC, Vieira LR, Hamoy AO, Tiago ACDS, Martins Rodrigues JC, de Araujo DB, Favacho Lopes DC, de Mello VJ, Hamoy M. Unmasking hidden risks: The surprising link between PDE5 inhibitors and seizure susceptibility. PLoS One 2023; 18:e0294754. [PMID: 38033148 PMCID: PMC10688920 DOI: 10.1371/journal.pone.0294754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 11/08/2023] [Indexed: 12/02/2023] Open
Abstract
BACKGROUND Phosphodiesterase 5 inhibitors (PDE5i) are the first line treatment for erectile dysfunction; however, several articles and case reports have shown central nervous system effects, that can cause seizures in susceptible patients. This study aims to describe the changes caused by the use of Sildenafil and Tadalafil through the analysis of abnormalities expressed in the electrocorticogram (ECoG) of rats and evaluate the seizure threshold response and treatment of seizures with anticonvulsants. MATERIALS AND METHODS The study used 108 rats (Wistar). Before surgery for electrode placement in dura mater, the animals were randomly separated into 3 experiments for electrocorticogram analysis. Experiment 1: ECoG response to using PD5i (Sildenafil 20mg/kg and Tadalafil 2.6mg/kg p.o.). Experiment 2: ECoG response to the use of PD5i in association with Pentylenetetrazole (PTZ-30 mg/kg i.p.), a convulsive model. Experiment 3: ECoG response to anticonvulsant treatment (Phenytoin, Phenobarbital and Diazepam) of seizures induced by association IPDE5 + PTZ. All recordings were made thirty minutes after administration of the medication and analyzed for ten minutes, only once. We considered statistical significance level of *p<0.05, **p<0.01 and ***p < 0.001. RESULTS After administration of Sildenafil and Tadalafil, there were increases in the power of recordings in the frequency bands in oscillations in alpha (p = 0.0920) and beta (p = 0.602) when compared to the control group (p<0.001). After the use of Sildenafil and Tadalafil associated with PTZ, greater potency was observed in the recordings during seizures (p<0.001), however, the Sildenafil group showed greater potency when compared to Tadalafil (p<0.05). Phenobarbital and Diazepam showed a better response in controlling discharges triggered by the association between proconvulsant drugs. CONCLUSIONS PDE5i altered the ECoG recordings in the rats' motor cortexes, demonstrating cerebral asynchrony and potentiating the action of PTZ. These findings demonstrate that PDE5i can lower the seizure threshold.
Collapse
Affiliation(s)
- Alex Luiz Menezes da Silva
- Laboratory of Pharmacology and Toxicology of Natural Products, Institute of Biological Sciences, Federal University of Pará, UFPA, Belém, Pará, Brazil
| | - Chirlene Pinheiro Nascimento
- Laboratory of Pharmacology and Toxicology of Natural Products, Institute of Biological Sciences, Federal University of Pará, UFPA, Belém, Pará, Brazil
| | - Julianne Elba Cunha Azevedo
- Laboratory of Pharmacology and Toxicology of Natural Products, Institute of Biological Sciences, Federal University of Pará, UFPA, Belém, Pará, Brazil
| | - Luana Rodrigues Vieira
- Laboratory of Pharmacology and Toxicology of Natural Products, Institute of Biological Sciences, Federal University of Pará, UFPA, Belém, Pará, Brazil
| | - Akira Otake Hamoy
- Laboratory of Pharmacology and Toxicology of Natural Products, Institute of Biological Sciences, Federal University of Pará, UFPA, Belém, Pará, Brazil
| | - Allan Carlos da Silva Tiago
- Laboratory of Pharmacology and Toxicology of Natural Products, Institute of Biological Sciences, Federal University of Pará, UFPA, Belém, Pará, Brazil
| | - João Cleiton Martins Rodrigues
- Laboratory of Experimental Neuropathology, Institute of Biological Sciences, Federal University of Pará, UFPA, Belém, Pará, Brazil
| | - Daniella Bastos de Araujo
- Laboratory of Pharmacology and Toxicology of Natural Products, Institute of Biological Sciences, Federal University of Pará, UFPA, Belém, Pará, Brazil
| | - Dielly Catrina Favacho Lopes
- Laboratory of Experimental Neuropathology, Institute of Biological Sciences, Federal University of Pará, UFPA, Belém, Pará, Brazil
| | - Vanessa Jóia de Mello
- Laboratory of Pharmacology and Toxicology of Natural Products, Institute of Biological Sciences, Federal University of Pará, UFPA, Belém, Pará, Brazil
| | - Moisés Hamoy
- Laboratory of Pharmacology and Toxicology of Natural Products, Institute of Biological Sciences, Federal University of Pará, UFPA, Belém, Pará, Brazil
| |
Collapse
|
110
|
Nazabal A, Mendiguren A, Pineda J. Inhibition of rat locus coeruleus neurons by prostaglandin E 2 EP3 receptors: pharmacological characterization ex vivo. Front Pharmacol 2023; 14:1290605. [PMID: 38035000 PMCID: PMC10684765 DOI: 10.3389/fphar.2023.1290605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 10/30/2023] [Indexed: 12/02/2023] Open
Abstract
Prostaglandin E2 (PGE2) is an inflammatory mediator synthesized by the brain constitutive cyclooxygenase enzyme. PGE2 binds to G protein-coupled EP1-4 receptors (EP1 to Gq, EP2,4 to Gs, and EP3 to Gi/o). EP2, EP3 and EP4 receptors are expressed in the locus coeruleus (LC), the main noradrenergic nucleus in the brain. EP3 receptors have been explored in the central nervous system, although its role regulating the locus coeruleus neuron activity has not been pharmacologically defined. Our aim was to characterize the function of EP3 receptors in neurons of the LC. Thus, we studied the effect of EP3 receptor agonists on the firing activity of LC cells in rat brain slices by single-unit extracellular electrophysiological techniques. The EP3 receptor agonist sulprostone (0.15 nM-1.28 µM), PGE2 (0.31 nM-10.2 µM) and the PGE1 analogue misoprostol (0.31 nM-2.56 µM) inhibited the firing rate of LC neurons in a concentration-dependent manner (EC50 = 15 nM, 110 nM, and 51 nM, respectively). The EP3 receptor antagonist L-798,106 (3-10 µM), but not the EP2 (PF-04418948, 3-10 µM) or EP4 (L-161,982, 3-10 µM) receptor antagonists, caused rightward shifts in the concentration-effect curves for the EP3 receptor agonists. Sulprostone-induced effect was attenuated by the Gi/o protein blocker pertussis toxin (pertussis toxin, 500 ng ml-1) and the inhibitors of inwardly rectifying potassium channels (GIRK) BaCl2 (300 µM) and SCH-23390 (15 µM). In conclusion, LC neuron firing activity is regulated by EP3 receptors, presumably by an inhibitory Gi/o protein- and GIRK-mediated mechanism.
Collapse
|
111
|
Bensemmane L, Milliat F, Treton X, Linard C. Systemically delivered adipose stromal vascular fraction mitigates radiation-induced gastrointestinal syndrome by immunomodulating the inflammatory response through a CD11b + cell-dependent mechanism. Stem Cell Res Ther 2023; 14:325. [PMID: 37953266 PMCID: PMC10641938 DOI: 10.1186/s13287-023-03562-7] [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: 03/01/2023] [Accepted: 11/07/2023] [Indexed: 11/14/2023] Open
Abstract
BACKGROUND Stromal vascular fraction (SVF) treatment promoted the regeneration of the intestinal epithelium, limiting lethality in a mouse model of radiation-induced gastrointestinal syndrome (GIS). The SVF has a heterogeneous cell composition; the effects between SVF and the host intestinal immunity are still unknown. The specific role of the different cells contained in the SVF needs to be clarified. Monocytes-macrophages have a crucial role in repair and monocyte recruitment and activation are orchestrated by the chemokine receptors CX3CR1 and CCR2. METHODS Mice exposed to abdominal radiation (18 Gy) received a single intravenous injection of SVF (2.5 × 106 cells), obtained by enzymatic digestion of inguinal fat tissue, on the day of irradiation. Intestinal immunity and regeneration were evaluated by flow cytometry, RT-PCR and histological analyses. RESULTS Using flow cytometry, we showed that SVF treatment modulated intestinal monocyte differentiation at 7 days post-irradiation by very early increasing the CD11b+Ly6C+CCR2+ population in the intestine ileal mucosa and accelerating the phenotype modification to acquire CX3CR1 in order to finally restore the F4/80+CX3CR1+ macrophage population. In CX3CR1-depleted mice, SVF treatment fails to mature the Ly6C-MCHII+CX3CR1+ population, leading to a macrophage population deficit associated with proinflammatory environment maintenance and defective intestinal repair; this impaired SVF efficiency on survival. Consistent with a CD11b+ being involved in SVF-induced intestinal repair, we showed that SVF-depleted CD11b+ treatment impaired F4/80+CX3CR1+macrophage pool restoration and caused loss of anti-inflammatory properties, abrogating stem cell compartment repair and survival. CONCLUSIONS These data showed that SVF treatment mitigates the GIS-involving immunomodulatory effect. Cooperation between the monocyte in SVF and the host monocyte defining the therapeutic properties of the SVF is necessary to guarantee the effective action of the SVF on the GIS.
Collapse
Affiliation(s)
- Lydia Bensemmane
- PSE-SANTE/SERAMED/LRMed, Institut de Radioprotection et de Sûreté Nucléaire (IRSN), 92260, Fontenay-Aux-Roses, France
| | - Fabien Milliat
- PSE-SANTE/SERAMED/LRMed, Institut de Radioprotection et de Sûreté Nucléaire (IRSN), 92260, Fontenay-Aux-Roses, France
| | | | - Christine Linard
- PSE-SANTE/SERAMED/LRMed, Institut de Radioprotection et de Sûreté Nucléaire (IRSN), 92260, Fontenay-Aux-Roses, France.
| |
Collapse
|
112
|
Ledesma-Corvi S, García-Fuster MJ. Electroconvulsive seizures regulate various stages of hippocampal cell genesis and mBDNF at different times after treatment in adolescent and adult rats of both sexes. Front Mol Neurosci 2023; 16:1275783. [PMID: 37965039 PMCID: PMC10642262 DOI: 10.3389/fnmol.2023.1275783] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/16/2023] [Indexed: 11/16/2023] Open
Abstract
Electroconvulsive therapy, a fast-acting option for treatment-resistant depression, is modeled at the preclinical level through the induction of electroconvulsive seizures (ECS) in rodents. Recent studies from our group proved sex- and age-differences in the antidepressant-like response elicited by ECS in rats; while an antidepressant-like response was observed in male adolescent and adult rats (although with greater efficacy in adulthood), the same parameters rendered inefficacious in females of any age. To better understand the potential sex differences taking place at the molecular level that might be mediating these behavioral disparities, we evaluated the impact of a repeated treatment with ECS (95 mA for 0.6 s, 100 Hz, 0.6 ms) in adolescent and adult rats of both sexes. Several hippocampal markers of neuroplasticity, commonly regulated by most antidepressants, such as those of neurogenesis (cell proliferation, neurogenic differentiation, long-term cell survival) or mBDNF and associated signaling (e.g., mTOR and ERK1/2) were evaluated at different time-points after treatment (1-, 8-, 15- and up to 30-days post-treatment). The main results demonstrated that ECS improved the survival rate of new cells born in the dentate gryus before treatment. Moreover, ECS increased cell proliferation and neurogenic differentiation at different times post-treatment, paired with persistent increases in mBDNF, observed long after treatment. In general, effects were different for each sex and varied with the age of the animal (adolescent vs. adulthood). The present study is the first-one to demonstrate that such persistent molecular changes induced by ECS in hippocampus, some of them observed up to 30-days post-treatment, also occurred in female rats and adolescence. Although these molecular changes could not justify the lack of ECS efficacy described by these same parameters of ECS in female rats (vs. male rats), they proposed certain beneficial effects common to both sexes, and age periods studied, opening the avenue for further studies. Based on these neurochemical effects, ECS should have displayed similar efficacies for both biological sexes. Therefore, the reason behind these disparities should be further explored to better translate efficacious treatments specific and/or personalized for each sex to the clinic.
Collapse
Affiliation(s)
- Sandra Ledesma-Corvi
- IUNICS, University of the Balearic Islands, Palma, Spain
- Health Research Institute of Balearic Islands (IdISBa), Palma, Spain
| | - M. Julia García-Fuster
- IUNICS, University of the Balearic Islands, Palma, Spain
- Health Research Institute of Balearic Islands (IdISBa), Palma, Spain
- Department of Medicine, University of the Balearic Islands, Palma, Spain
| |
Collapse
|
113
|
Ledesma-Corvi S, Jornet-Plaza J, García-Fuster MJ. Aromatase inhibition and ketamine in rats: sex-differences in antidepressant-like efficacy. Biol Sex Differ 2023; 14:73. [PMID: 37876000 PMCID: PMC10599051 DOI: 10.1186/s13293-023-00560-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 10/17/2023] [Indexed: 10/26/2023] Open
Abstract
BACKGROUND Ketamine has been recently approved to treat resistant depression; however preclinical studies showed sex differences in its efficacy. Sex steroids, such as estrogens and testosterone, both in the periphery and locally in the brain, are regarded as important modulators of these sex differences. Therefore, the present study evaluated how inhibiting the biosynthesis of estrogens with letrozole (an aromatase inhibitor) could affect the observed sex differences in ketamine's antidepressant-like-response. METHODS We performed several consecutive studies in adult Sprague-Dawley rats to evaluate potential sex differences in the antidepressant-like effects of ketamine (5 mg/kg, 7 days, i.p.), letrozole (1 mg/kg, 8 days, i.p.) and their combination (letrozole pre-treatment 3 h before ketamine). Acute and repeated antidepressant-like responses were ascertained in a series of behavioral tests (forced-swim, novelty-suppressed feeding, two-bottle choice for sucrose preference). RESULTS The main results proved clear sex differences in the antidepressant-like response induced by ketamine, which was observed following a repeated paradigm in adult male rats, but rendered inefficacious in female rats. Moreover, decreasing estrogens production with letrozole induced on itself an antidepressant-like response in female rats, while also increased ketamine's response in male rats (i.e., quicker response observed after only a single dose). Interestingly, both the antidepressant-like effects induced by ketamine in male rats or letrozole in female rats persisted over time up to 65 days post-treatment, suggesting long-term sex-directed benefits for these drugs. CONCLUSIONS The present results demonstrated a sex-specific role for aromatase inhibition with letrozole in the antidepressant-like response induced by ketamine in male rats. Moreover, letrozole itself presented as a potential antidepressant for females with persistent effects over time. Clearly, the production of estrogens is key in modulating, in a sex-specific manner, affective-like responses and thus deserve further studies.
Collapse
Affiliation(s)
- Sandra Ledesma-Corvi
- IUNICS, University of the Balearic Islands, Cra. de Valldemossa Km 7.5, 07122, Palma, Spain
- Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - Jordi Jornet-Plaza
- IUNICS, University of the Balearic Islands, Cra. de Valldemossa Km 7.5, 07122, Palma, Spain
- Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - M Julia García-Fuster
- IUNICS, University of the Balearic Islands, Cra. de Valldemossa Km 7.5, 07122, Palma, Spain.
- Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain.
- Department of Medicine, University of the Balearic Islands, Palma, Spain.
| |
Collapse
|
114
|
Manhas A, Tripathi D, Jagavelu K. Involvement of HIF1α/Reg protein in the regulation of HMGB3 in myocardial infarction. Vascul Pharmacol 2023; 152:107197. [PMID: 37467910 DOI: 10.1016/j.vph.2023.107197] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 04/27/2023] [Accepted: 07/16/2023] [Indexed: 07/21/2023]
Abstract
AIMS Myocardial ischemia and infarction are the number one cause of cardiovascular disease associated mortality. Cardiomyocyte death during ischemia leads to the loss of cardiac tissue and initiates a signaling cascade between the infarct zone and the area at risk of the myocardium. Here, we sought to determine the involvement of one of the damage-associated molecular patterns HMGB3 in myocardial ischemia and infarction. METHODS AND RESULTS We used the left anterior descending coronary artery ligation model to study the involvement of HMGB3 in myocardial ischemia and infarction. Our results indicated the presence of HMGB3 at a low level under normal conditions, while myocardial injury caused a robust increase in HMGB3 levels in the heart. Further, intra-cardiac injection of mabHMGB3 had improved cardiac function at day 3 by downregulating HMGB3 levels. In contrast, injection of recombinant rat HMGB3 for 7 days during the adaptation phase of myocardial ischemia improved cardiac functional parameters by increasing regenerative protein family expression. Further, to mimic the disease condition, neonatal rat ventricle cardiomyocytes and fibroblasts were exposed to hypoxia; we observed a significant upregulation in the HMGB3, HIF1α, and Reg1α levels. Endothelial cells exposed to recombinant HMGB3 increased the tubule length. Further, the mitochondrial oxygen consumption rate was reduced with the acute induction of recombinant HMGB3 on cardiomyocytes and fibroblasts. CONCLUSION HMGB3 plays a dual role during the progression of myocardial ischemia and infarction. Clinically, post-myocardial infarction HMGB3-induced sterile inflammation needs to be tightly controlled, as it plays both a pro-inflammatory role and improves cardiac function during the cardiac remodeling phase.
Collapse
Affiliation(s)
- Amit Manhas
- Department of Pharmacology, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Science and Innovation Research, New Delhi, India
| | - Dipti Tripathi
- Department of Pharmacology, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Science and Innovation Research, New Delhi, India
| | - Kumaravelu Jagavelu
- Department of Pharmacology, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Science and Innovation Research, New Delhi, India.
| |
Collapse
|
115
|
Dong T, Fan X, Zheng N, Yan K, Hou T, Peng L, Ci X. Activation of Nrf2 signalling pathway by tectoridin protects against ferroptosis in particulate matter-induced lung injury. Br J Pharmacol 2023; 180:2532-2549. [PMID: 37005797 DOI: 10.1111/bph.16085] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 03/01/2023] [Accepted: 03/21/2023] [Indexed: 04/04/2023] Open
Abstract
BACKGROUND AND PURPOSE Our previous research showed that ferroptosis plays a crucial role in the pathophysiology of PM2.5-induced lung injury. The present study aimed to investigate the protective role of the Nrf2 signalling pathway and its bioactive molecule tectoridin in PM2.5-induced lung injury by regulating ferroptosis. EXPERIMENTAL APPROACH We examined the regulatory effect of Nrf2 on ferroptosis in PM2.5-induced lung injury and Beas-2b cells using Nrf2-knockout (KO) mice and Nrf2 siRNA transfection. The effects and underlying mechanisms of tectoridin on PM2.5-induced lung injury were evaluated in vitro and in vivo. KEY RESULTS Nrf2 deletion increased iron accumulation and ferroptosis-related protein expression in vivo and vitro, further exacerbating lung injury and cell death in response to PM2.5 exposure. Tectoridin activated Nrf2 target genes and ameliorated cell death caused by PM2.5. In addition, tectoridin prevented lipid peroxidation, iron accumulation and ferroptosis in vitro, but in siNrf2-treated cells, these effects almost disappeared. In addition, tectoridin effectively mitigated PM2.5-induced respiratory system damage, as evaluated by HE, PAS, and inflammatory factors. Tectoridin also augmented the antioxidative Nrf2 signalling pathway and prevented changes in ferroptosis-related morphological and biochemical indicators, including MDA levels, GSH depletion and GPX4 and xCT downregulation, in PM2.5-induced lung injury. However, the effects of tectoridin on ferroptosis and respiratory injury were almost abolished in Nrf2-KO mice. CONCLUSION AND IMPLICATIONS Our data proposed the protective effect of Nrf2 activation on PM2.5-induced lung injury by inhibiting ferroptosis-mediated lipid peroxidation and highlight the potential of tectoridin as a PM2.5-induced lung injury treatment.
Collapse
Affiliation(s)
- Tingting Dong
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, China
| | - Xiaoye Fan
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, China
| | - Nan Zheng
- Department of Pharmacy, the Second Hospital of Jilin University, Changchun, China
| | - Kun Yan
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, China
| | - Tianhua Hou
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, China
| | - Liping Peng
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, China
| | - Xinxin Ci
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, China
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
116
|
Chen CM, Gung PY, Ho YC, Hamdin CD, Yet SF. Probucol treatment after traumatic brain injury activates BDNF/TrkB pathway, promotes neuroregeneration and ameliorates functional deficits in mice. Br J Pharmacol 2023; 180:2605-2622. [PMID: 37263748 DOI: 10.1111/bph.16157] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 04/11/2023] [Accepted: 05/22/2023] [Indexed: 06/03/2023] Open
Abstract
BACKGROUND AND PURPOSE Traumatic brain injury (TBI) is a major cause of mortality and morbidity worldwide, yet pharmacotherapies for TBI are currently lacking. Neuroregeneration is important in brain repair and functional recovery. In this study, probucol, a cholesterol-lowering drug with established safety profiles, was examined for its therapeutic effects and neuroregenerative actions in TBI. EXPERIMENTAL APPROACH Male mice were subjected to the controlled cortical impact model of TBI, followed by daily administration of probucol. Neurological and cognitive functions were evaluated. Histological analyses of the neocortex and hippocampus were performed to detect the lesion, dendritic degeneration (microtubule-associated protein 2), synaptic density (synaptophysin), neurogenesis (doublecortin), brain-derived neurotrophic factor (BDNF) and tropomyosin receptor kinase B (TrkB) activation. Involvement of BDNF/TrkB pathway in probucol-mediated effects was examined in primary cultures of cortical neurons. KEY RESULTS Probucol reduced brain lesion volume, enhanced the recovery of body symmetry, improved motor function and attenuated memory dysfunction after TBI. Meanwhile, probucol promoted post-injury dendritic growth and synaptogenesis and increased hippocampal proliferating neuronal progenitor cells, along with the formation as well as the survival of newborn neurons. Moreover, probucol enhances BDNF expression and TrkB activation. In vitro, probucol promoted neurite outgrowth, which was inhibited by a selective TrkB antagonist ANA-12. CONCLUSIONS AND IMPLICATIONS Probucol enhanced functional restoration and ameliorated cognitive impairment after TBI by promoting post-injury neuronal remodelling and neurogenesis. Increased activation of BDNF/TrkB pathway by probucol, at least in part, contributed to the neuroregenerative effects of probucol. Together, it may be promising to repurpose probucol for TBI.
Collapse
Affiliation(s)
- Chen-Mei Chen
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan
| | - Pei-Yu Gung
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan
| | - Yen-Chun Ho
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan
- Cardiovascular Biology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Candra D Hamdin
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan
- National Health Research Institutes & Department of Life Sciences, National Central University Joint Ph.D. Program in Biomedicine, Taoyuan City, Taiwan
| | - Shaw-Fang Yet
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| |
Collapse
|
117
|
Nizamoglu M, Joglekar MM, Almeida CR, Larsson Callerfelt AK, Dupin I, Guenat OT, Henrot P, van Os L, Otero J, Elowsson L, Farre R, Burgess JK. Innovative three-dimensional models for understanding mechanisms underlying lung diseases: powerful tools for translational research. Eur Respir Rev 2023; 32:230042. [PMID: 37495250 PMCID: PMC10369168 DOI: 10.1183/16000617.0042-2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 05/04/2023] [Indexed: 07/28/2023] Open
Abstract
Chronic lung diseases result from alteration and/or destruction of lung tissue, inevitably causing decreased breathing capacity and quality of life for patients. While animal models have paved the way for our understanding of pathobiology and the development of therapeutic strategies for disease management, their translational capacity is limited. There is, therefore, a well-recognised need for innovative in vitro models to reflect chronic lung diseases, which will facilitate mechanism investigation and the advancement of new treatment strategies. In the last decades, lungs have been modelled in healthy and diseased conditions using precision-cut lung slices, organoids, extracellular matrix-derived hydrogels and lung-on-chip systems. These three-dimensional models together provide a wide spectrum of applicability and mimicry of the lung microenvironment. While each system has its own limitations, their advantages over traditional two-dimensional culture systems, or even over animal models, increases the value of in vitro models. Generating new and advanced models with increased translational capacity will not only benefit our understanding of the pathobiology of lung diseases but should also shorten the timelines required for discovery and generation of new therapeutics. This article summarises and provides an outline of the European Respiratory Society research seminar "Innovative 3D models for understanding mechanisms underlying lung diseases: powerful tools for translational research", held in Lisbon, Portugal, in April 2022. Current in vitro models developed for recapitulating healthy and diseased lungs are outlined and discussed with respect to the challenges associated with them, efforts to develop best practices for model generation, characterisation and utilisation of models and state-of-the-art translational potential.
Collapse
Affiliation(s)
- Mehmet Nizamoglu
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, The Netherlands
- Both authors contributed equally
| | - Mugdha M Joglekar
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, The Netherlands
- Both authors contributed equally
| | - Catarina R Almeida
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, Aveiro, Portugal
| | | | - Isabelle Dupin
- Centre de Recherche Cardio-thoracique de Bordeaux, Université de Bordeaux, Pessac, France
- INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, Pessac, France
| | - Olivier T Guenat
- Organs-on-Chip Technologies, ARTORG Center for Biomedical Engineering Research, University of Bern, Bern, Switzerland
- Department of Pulmonary Medicine, University Hospital of Bern, Bern, Switzerland
- Department of General Thoracic Surgery, University Hospital of Bern, Bern, Switzerland
| | - Pauline Henrot
- Centre de Recherche Cardio-thoracique de Bordeaux, Université de Bordeaux, Pessac, France
- INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, Pessac, France
- Service d'exploration fonctionnelle respiratoire, CHU de Bordeaux, Pessac, France
| | - Lisette van Os
- Organs-on-Chip Technologies, ARTORG Center for Biomedical Engineering Research, University of Bern, Bern, Switzerland
| | - Jorge Otero
- Unit of Biophysics and Bioengineering, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
- CIBER de Enfermedades Respiratorias, Madrid, Spain
| | - Linda Elowsson
- Lung Biology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Ramon Farre
- Unit of Biophysics and Bioengineering, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
- CIBER de Enfermedades Respiratorias, Madrid, Spain
- Institut Investigacions Biomediques August Pi Sunyer, Barcelona, Spain
| | - Janette K Burgess
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, W.J. Kolff Institute for Biomedical Engineering and Materials Science-FB41, Groningen, The Netherlands
| |
Collapse
|
118
|
Ledesma-Corvi S, García-Fuster MJ. Comparing the antidepressant-like effects of electroconvulsive seizures in adolescent and adult female rats: an intensity dose-response study. Biol Sex Differ 2023; 14:67. [PMID: 37777813 PMCID: PMC10541687 DOI: 10.1186/s13293-023-00552-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 09/22/2023] [Indexed: 10/02/2023] Open
Abstract
BACKGROUND The induction of electroconvulsive seizures (ECS) in rodents induces sex- and age-specific disparities in antidepressant-like responses, with females and young age being the most unresponsive ones. Since the electrical charge needed to induce an effective convulsion is also altered by these variables, our aim was to compare different dose-intensities of ECS exclusively in female rats, since there is a lack of preclinical data characterizing this particular sex, while also evaluating efficacy during distinctive age periods of treatment (adolescence vs. adulthood). METHODS Adolescent and adult female Sprague-Dawley rats were exposed to an intensity dose-response study (55, 75 or 95 mA; 0.6 s, 100 Hz, 1 session/day, 5 days). The particular characteristics of the induced convulsions (tonic, clonic, recovery times) were monitored during treatment. Antidepressant-like responses were evaluated under the stress of the forced-swim test 1-, 3-, and 7-days post-treatment (i.e., improved immobility time as an indicative of an antidepressant-like response), and brains were collected 24 h later (8 days post-treatment) to evaluate potential changes in hippocampal neurogenesis (Ki-67 and NeuroD) by immunohistochemistry. RESULTS The lowest intensities tested of ECS (55 and 75 mA) induced an antidepressant-like effect in adult female rats, but rendered insufficient in adolescence. The lack of efficacy observed in adolescent rats paralleled differences in the characteristics of the seizures induced by ECS as compared to adulthood. In line with prior results, different dose-intensities of ECS modulated hippocampal neurogenesis in a comparable fashion with age (i.e., increased survival of neural progenitors 8 days post-treatment). CONCLUSIONS In conjunction, these results reinforce the importance of fine-tuning the parameters of ECS that might render efficacious while considering sex and age as essential variables for treatment response, and suggest that other molecular mechanisms, beside the partial role of hippocampal neurogenesis, might be participating in the antidepressant-like effects induced by ECS.
Collapse
Affiliation(s)
- Sandra Ledesma-Corvi
- IUNICS, University of the Balearic Islands, Cra. de Valldemossa Km 7.5, 07122, Palma, Spain
- Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - M Julia García-Fuster
- IUNICS, University of the Balearic Islands, Cra. de Valldemossa Km 7.5, 07122, Palma, Spain.
- Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain.
- Department of Medicine, University of the Balearic Islands, Palma, Spain.
| |
Collapse
|
119
|
Ledesma-Corvi S, García-Fuster MJ. Aromatase Inhibition and Electroconvulsive Seizures in Adolescent Rats: Antidepressant and Long-Term Cognitive Sex Differences. Int J Neuropsychopharmacol 2023; 26:607-615. [PMID: 37559395 PMCID: PMC10519810 DOI: 10.1093/ijnp/pyad047] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 08/01/2023] [Indexed: 08/11/2023] Open
Abstract
BACKGROUND We recently showed sex differences in the antidepressant-like potential of electroconvulsive seizures (ECS) in adolescent rats; whereas it worked for male rats, it was inefficacious in females. Because sex steroids might be important modulators of these sex disparities, we evaluated the role of estrogens in the differential response induced by adolescent ECS. Moreover, given the literature suggesting certain cognitive sequelae from ECS exposure, we aimed at evaluating its long-term safety profile in adulthood. METHODS Adolescent Sprague-Dawley rats were pretreated with letrozole (1 mg/kg/day) or vehicle (1 mL/kg/day) for 8 days (i.p.) and treated during the last 5 days (3 hours later) with ECS (95 mA, 0.6 s, 100 Hz) or SHAM. Antidepressant-like responses were measured in the forced swim test, and long-term cognitive performance was assessed in the Barnes maze. RESULTS During adolescence, whereas ECS alone exerted an antidepressant-like response in male rats, its combination with letrozole permitted ECS to also induce efficacy in females. Moreover, adolescent ECS treatment improved cognitive performance in adulthood although exclusively in male rats. CONCLUSIONS Adolescent ECS demonstrated an antidepressant-like potential together with certain long-term beneficial cognitive effects but exclusively in male rats. For females, efficacy was restricted to a situation in which the biosynthesis of estrogens was reduced. Therefore, estrogens and/or testosterone levels play a crucial role in the sex disparities induced by ECS in Sprague-Dawley rats. Based on this study and on the literature supporting its safety, ECS should be encouraged for use in cases of treatment-resistant depression during adolescence, while adhering to sex-specific considerations.
Collapse
Affiliation(s)
- Sandra Ledesma-Corvi
- IUNICS, University of the Balearic Islands, Palma, Spain
- Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - M Julia García-Fuster
- IUNICS, University of the Balearic Islands, Palma, Spain
- Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| |
Collapse
|
120
|
Olivencia MA, Gil de Biedma-Elduayen L, Giménez-Gómez P, Barreira B, Fernández A, Angulo J, Colado MI, O'Shea E, Perez-Vizcaino F. Oxidized soluble guanylyl cyclase causes erectile dysfunction in alcoholic mice. Br J Pharmacol 2023; 180:2361-2376. [PMID: 37021655 DOI: 10.1111/bph.16087] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 03/13/2023] [Accepted: 04/03/2023] [Indexed: 04/07/2023] Open
Abstract
BACKGROUND AND PURPOSE Alcohol abuse has been associated with erectile dysfunction (ED), but the implicated molecular mechanisms are unresolved. This study analyses the role of alterations in soluble guanylyl cyclase (sGC) in ED. EXPERIMENTAL APPROACH ED was analysed in adult male C57BL/6J mice subjected to the Chronic Intermittent Ethanol (CIE) paradigm. Erectile function was assessed in anaesthetised mice in vivo by evaluating intracavernosal pressure (ICP) and in vitro in isolated mice corpora cavernosa (CC) mounted in a myograph. Protein expression and reactive oxygen species were analysed by western blot and dihydroethidium staining, respectively. KEY RESULTS In CIE mice, we observed a significant decrease in the relaxant response of the CC to stimulation of NO release from nitrergic nerves by electrical field stimulation, to NO release from endothelial cells by acetylcholine, to the PDE5 inhibitor sildenafil, and to the sGC stimulator riociguat. Conversely, the response to the sGC activator cinaciguat, whose action is independent of the oxidation state of sGC, was significantly enhanced in these CC. The responses to adenylyl cyclase stimulation with forskolin were unchanged. We found an increase in reactive oxygen species in the CC from CIE mice as well as an increase in CYP2E1 and NOX2 protein expression. In vivo pre-treatment with tempol prevented alcohol-induced erectile dysfunction. CONCLUSIONS AND IMPLICATIONS Our results demonstrate that alcoholic mice show ED in vitro and in vivo due to an alteration in the redox state of sGC and suggest that sGC activators may be effective in ED associated with alcoholism.
Collapse
Affiliation(s)
- Miguel A Olivencia
- Departamento de Farmacologia y Toxicologia, Facultad de Medicina, Universidad Complutense, Madrid, Spain
- CIBER Enfermedades Respiratorias, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Leticia Gil de Biedma-Elduayen
- Departamento de Farmacologia y Toxicologia, Facultad de Medicina, Universidad Complutense, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain
- Red de Investigación en Atención Primaria de Adicciones del Instituto de Salud Carlos III, Madrid, Spain
- Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Pablo Giménez-Gómez
- Departamento de Farmacologia y Toxicologia, Facultad de Medicina, Universidad Complutense, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain
- Red de Investigación en Atención Primaria de Adicciones del Instituto de Salud Carlos III, Madrid, Spain
- Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Bianca Barreira
- Departamento de Farmacologia y Toxicologia, Facultad de Medicina, Universidad Complutense, Madrid, Spain
- CIBER Enfermedades Respiratorias, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Argentina Fernández
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
- Servicio de Histología-Investigación, Unidad de Investigación Traslacional en Cardiología (IRYCIS-UFV), Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Javier Angulo
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
- Servicio de Histología-Investigación, Unidad de Investigación Traslacional en Cardiología (IRYCIS-UFV), Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Maria Isabel Colado
- Departamento de Farmacologia y Toxicologia, Facultad de Medicina, Universidad Complutense, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain
- Red de Investigación en Atención Primaria de Adicciones del Instituto de Salud Carlos III, Madrid, Spain
- Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Esther O'Shea
- Departamento de Farmacologia y Toxicologia, Facultad de Medicina, Universidad Complutense, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre, Madrid, Spain
- Red de Investigación en Atención Primaria de Adicciones del Instituto de Salud Carlos III, Madrid, Spain
- Instituto Universitario de Investigación Neuroquímica (IUIN), Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Francisco Perez-Vizcaino
- Departamento de Farmacologia y Toxicologia, Facultad de Medicina, Universidad Complutense, Madrid, Spain
- CIBER Enfermedades Respiratorias, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| |
Collapse
|
121
|
Yao X, Kong X, Ren J, Cui Y, Chen S, Cheng J, Gao J, Sun J, Xu X, Hu W, Li H, Che F, Wan Q. Transcranial direct-current stimulation confers neuroprotection by regulating isoleucine-dependent signalling after rat cerebral ischemia-reperfusion injury. Eur J Neurosci 2023; 58:3330-3346. [PMID: 37452630 DOI: 10.1111/ejn.16091] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 06/25/2023] [Accepted: 06/28/2023] [Indexed: 07/18/2023]
Abstract
Isoleucine is a branched chain amino acid. The role of isoleucine in cerebral ischemia-reperfusion injury remains unclear. Here, we show that the concentration of isoleucine is decreased in cerebrospinal fluid in a rat model of cerebral ischemia-reperfusion injury, the rat middle cerebral artery occlusion (MCAO). To our surprise, the level of intraneuronal isoleucine is increased in an in vitro model of cerebral ischemia injury, the oxygen-glucose deprivation (OGD). We found that the increased activity of LAT1, an L-type amino acid transporter 1, leads to the elevation of intraneuronal isoleucine after OGD insult. Reducing the level of intraneuronal isoleucine promotes cell survival after cerebral ischemia-reperfusion injury, but supplementing isoleucine aggravates the neuronal damage. To understand how isoleucine promotes ischemia-induced neuronal death, we reveal that isoleucine acts upstream to reduce the expression of CBFB (core binding factor β, a transcript factor involved in cell development and growth) and that the phosphatase PTEN acts downstream of CBFB to mediate isoleucine-induced neuronal damage after OGD insult. Interestingly, we demonstrate that direct-current stimulation reduces the level of intraneuronal isoleucine in cortical cultures subjected to OGD and that transcranial direct-current stimulation (tDCS) decreases the cerebral infarct volume of MCAO rat through reducing LAT1-depencent increase of intraneuronal isoleucine. Together, these results lead us to conclude that LAT1 over activation-dependent isoleucine-CBFB-PTEN signal transduction pathway may mediate ischemic neuronal injury and that tDCS exerts its neuroprotective effect by suppressing LAT1 over activation-dependent signalling after cerebral ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Xujin Yao
- Institute of Neuroregeneration and Neurorehabilitation, Department of Pathophysiology, Qingdao University, Qingdao, China
| | - Xiangyi Kong
- Institute of Neuroregeneration and Neurorehabilitation, Department of Pathophysiology, Qingdao University, Qingdao, China
| | - Jinyang Ren
- Institute of Neuroregeneration and Neurorehabilitation, Department of Pathophysiology, Qingdao University, Qingdao, China
| | - Yu Cui
- Institute of Neuroregeneration and Neurorehabilitation, Department of Pathophysiology, Qingdao University, Qingdao, China
| | - Songfeng Chen
- Department of Physiology, School of Medicine, Wuhan University, Wuhan, China
| | - Jing Cheng
- Department of Physiology, School of Medicine, Wuhan University, Wuhan, China
| | - Jingchen Gao
- Institute of Neuroregeneration and Neurorehabilitation, Department of Pathophysiology, Qingdao University, Qingdao, China
| | - Jiangdong Sun
- Institute of Neuroregeneration and Neurorehabilitation, Department of Pathophysiology, Qingdao University, Qingdao, China
| | - Xiangyu Xu
- Department of Rehabilitation, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wenjie Hu
- Institute of Neuroregeneration and Neurorehabilitation, Department of Pathophysiology, Qingdao University, Qingdao, China
| | - Huanting Li
- Institute of Neuroregeneration and Neurorehabilitation, Department of Pathophysiology, Qingdao University, Qingdao, China
| | - Fengyuan Che
- Central Laboratory, Department of Neurology, Linyi People's Hospital, Qingdao University, Linyi, Shandong, China
| | - Qi Wan
- Institute of Neuroregeneration and Neurorehabilitation, Department of Pathophysiology, Qingdao University, Qingdao, China
- Qingdao Gui-Hong Intelligent Medical Technology Co. Ltd, Qingdao, China
| |
Collapse
|
122
|
Borgonetti V, Roberts AJ, Bajo M, Galeotti N, Roberto M. Chronic alcohol induced mechanical allodynia by promoting neuroinflammation: A mouse model of alcohol-evoked neuropathic pain. Br J Pharmacol 2023; 180:2377-2392. [PMID: 37050867 PMCID: PMC10898491 DOI: 10.1111/bph.16091] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 04/05/2023] [Accepted: 04/06/2023] [Indexed: 04/14/2023] Open
Abstract
BACKGROUND AND PURPOSE Chronic pain is considered a key factor contributing to alcohol use disorder (AUD). The mechanisms responsible for chronic pain associated with chronic alcohol consumption are unknown. We evaluated the development of chronic pain in a mouse model of alcohol dependence and investigate the role of neuroinflammation. EXPERIMENTAL APPROACH The chronic-intermittent ethanol two-bottle choice CIE-2BC paradigm generates three groups: alcohol-dependent with escalating alcohol intake, nondependent (moderate drinking) and alcohol-naïve control male and female mice. We measured mechanical allodynia during withdrawal and after the last voluntary drinking. Immunoblotting was used to evaluate the protein levels of IBA-1, CSFR, IL-6, p38 and ERK2/1 in spinal cord tissue of dependent and non-dependent animals. KEY RESULTS We found significant escalation of drinking in the dependent group in male and female compared with the non-dependent group. The dependent group developed mechanical allodynia during 72 h of withdrawal, which was completely reversed after voluntary drinking. We observed an increased pain hypersensitivity compared with the naïve in 50% of non-dependent group. Increased IBA-1 and CSFR expression was observed in spinal cord tissue of both hypersensitivity-abstinence related and neuropathy-alcohol mice, and increased IL-6 expression and ERK1/2 activation in mice with hypersensitivity-related to abstinence, but not in mice with alcohol-evoked neuropathic pain. CONCLUSIONS AND IMPLICATIONS The CIE-2BC model induces two distinct pain conditions specific to the type of ethanol exposure: abstinence-related hypersensitivity in dependent mice and alcohol-evoked neuropathic pain in about a half of the non-dependent mice.
Collapse
Affiliation(s)
- Vittoria Borgonetti
- Department of Neuroscience, Psychology, Drug Research, and Child Health (NEUROFARBA), Section of Pharmacology, University of Florence, Viale G. Pieraccini 6, Florence, 50139, Italy
- Department of Molecular Medicine and Neuroscience, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Amanda J. Roberts
- Animal Models Core, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Michal Bajo
- Department of Molecular Medicine and Neuroscience, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Nicoletta Galeotti
- Department of Neuroscience, Psychology, Drug Research, and Child Health (NEUROFARBA), Section of Pharmacology, University of Florence, Viale G. Pieraccini 6, Florence, 50139, Italy
| | - Marisa Roberto
- Department of Molecular Medicine and Neuroscience, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, USA
| |
Collapse
|
123
|
Somalo-Barranco G, Pagano Zottola AC, Abdulrahman AO, El Zein RM, Cannich A, Muñoz L, Serra C, Oishi A, Marsicano G, Masri B, Bellocchio L, Llebaria A, Jockers R. Mitochondria-targeted melatonin photorelease supports the presence of melatonin MT1 receptors in mitochondria inhibiting respiration. Cell Chem Biol 2023; 30:920-932.e7. [PMID: 37572668 DOI: 10.1016/j.chembiol.2023.07.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 04/16/2023] [Accepted: 07/20/2023] [Indexed: 08/14/2023]
Abstract
The presence of signaling-competent G protein-coupled receptors in intracellular compartments is increasingly recognized. Recently, the presence of Gi/o protein-coupled melatonin MT1 receptors in mitochondria has been revealed, in addition to the plasma membrane. Melatonin is highly cell permeant, activating plasma membrane and mitochondrial receptors equally. Here, we present MCS-1145, a melatonin derivative bearing a triphenylphosphonium cation for specific mitochondrial targeting and a photocleavable o-nitrobenzyl group releasing melatonin upon illumination. MCS-1145 displayed low affinity for MT1 and MT2 but spontaneously accumulated in mitochondria, where it was resistant to washout. Uncaged MCS-1145 and exogenous melatonin recruited β-arrestin 2 to MT1 in mitochondria and inhibited oxygen consumption in mitochondria isolated from HEK293 cells only when expressing MT1 and from mouse cerebellum of WT mice but not from MT1-knockout mice. Overall, we developed the first mitochondria-targeted photoactivatable melatonin ligand and demonstrate that melatonin inhibits mitochondrial respiration through mitochondrial MT1 receptors.
Collapse
Affiliation(s)
- Gloria Somalo-Barranco
- Université Paris Cité, Institut Cochin, INSERM, CNRS, 75014 PARIS, France; MCS, Laboratory of Medicinal Chemistry & Synthesis, Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona, Spain
| | | | | | - Rami M El Zein
- Université Paris Cité, Institut Cochin, INSERM, CNRS, 75014 PARIS, France
| | - Astrid Cannich
- INSERM, U1215 NeuroCentre Magendie, Endocannabinoids and Neuroadaptation, Bordeaux, France
| | - Lourdes Muñoz
- MCS, Laboratory of Medicinal Chemistry & Synthesis, Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona, Spain; SIMChem, Synthesis of High Added Value Molecules, Institute of Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona, Spain
| | - Carme Serra
- MCS, Laboratory of Medicinal Chemistry & Synthesis, Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona, Spain; SIMChem, Synthesis of High Added Value Molecules, Institute of Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona, Spain
| | - Atsuro Oishi
- Université Paris Cité, Institut Cochin, INSERM, CNRS, 75014 PARIS, France
| | - Giovanni Marsicano
- INSERM, U1215 NeuroCentre Magendie, Endocannabinoids and Neuroadaptation, Bordeaux, France
| | - Bernard Masri
- Université Paris Cité, Institut Cochin, INSERM, CNRS, 75014 PARIS, France
| | - Luigi Bellocchio
- INSERM, U1215 NeuroCentre Magendie, Endocannabinoids and Neuroadaptation, Bordeaux, France
| | - Amadeu Llebaria
- MCS, Laboratory of Medicinal Chemistry & Synthesis, Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona, Spain.
| | - Ralf Jockers
- Université Paris Cité, Institut Cochin, INSERM, CNRS, 75014 PARIS, France.
| |
Collapse
|
124
|
Tsai YF, Chen CY, Yang SC, Syu YT, Hwang TL. Apremilast ameliorates acute respiratory distress syndrome by inhibiting neutrophil-induced oxidative stress. Biomed J 2023; 46:100560. [PMID: 36103985 PMCID: PMC10345255 DOI: 10.1016/j.bj.2022.09.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 08/31/2022] [Accepted: 09/07/2022] [Indexed: 11/02/2022] Open
Abstract
BACKGROUND The pathogenesis of acute respiratory distress syndrome (ARDS) is attributed to the dysregulation of oxidative stress and neutrophil recruitment. We aimed to investigate the anti-inflammatory effects of apremilast on human neutrophils and assess its efficacy for treating ARDS. METHODS We analysed superoxide anion generation, integrin expression, and adhesion in activated human neutrophils using spectrophotometry, flow cytometry, and immunofluorescence microscopy. Phosphorylation of extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK) was determined using immunoblotting. A murine lipopolysaccharide (LPS)-induced ARDS model was used to evaluate the therapeutic effects of apremilast. RESULTS Apremilast significantly decreased superoxide anion production, reactive oxygen species (ROS) generation, cluster of differentiation (CD)11 b expression, and neutrophil adhesion in formyl-l-methionyl-l-leucyl-l-phenylalanine activated human neutrophils. Apremilast elevated cyclic 3',5'-adenosine monophosphate (cAMP) and protein kinase A (PKA) activity in activated neutrophils. It reduced cellular cAMP-specific phosphodiesterase (PDE) activity and selectively inhibited enzymatic PDE4 activity. The activated cAMP/PKA pathway suppressed the phosphorylation of ERK and JNK as well as Ca2+ mobilization in activated neutrophils. All inhibitory effects of apremilast on activated neutrophils were reversed by a PKA inhibitor. In vivo examinations indicated that apremilast alleviated lung neutrophil infiltration, myeloperoxidase (MPO) activity, pulmonary oedema, and alveolar damage in LPS-induced ARDS. CONCLUSION Apremilast inhibits inflammatory responses after neutrophil activation via cAMP/PKA-dependent inhibition of ERK and JNK activation. Our study revealed apremilast suppresses oxidative stress and chemotaxis by selectively inhibiting PDE4 in neutrophils and thus protects against endotoxin-induced ARDS in mice. Apremilast can be used as an alternative off-label drug in treating acute lung damage.
Collapse
Affiliation(s)
- Yung-Fong Tsai
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Anesthesiology, Chang Gung Memorial Hospital at Taoyuan, Taoyuan, Taiwan; Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chun-Yu Chen
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Anesthesiology, Chang Gung Memorial Hospital at Taoyuan, Taoyuan, Taiwan; Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Shun-Chin Yang
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Anesthesiology, Taipei Veterans General Hospital and National Yang-Ming University, Taipei, Taiwan
| | - Yu-Ting Syu
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Anesthesiology, Chang Gung Memorial Hospital at Taoyuan, Taoyuan, Taiwan
| | - Tsong-Long Hwang
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Anesthesiology, Chang Gung Memorial Hospital at Taoyuan, Taoyuan, Taiwan; Research Center for Chinese Herbal Medicine and Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan; Department of Chemical Engineering, Ming Chi University of Technology, New Taipei, Taiwan.
| |
Collapse
|
125
|
Chan SMH, Brassington K, Almerdasi SA, Dobric A, De Luca SN, Coward‐Smith M, Wang H, Mou K, Akhtar A, Alateeq RA, Wang W, Seow HJ, Selemidis S, Bozinovski S, Vlahos R. Inhibition of oxidative stress by apocynin attenuated chronic obstructive pulmonary disease progression and vascular injury by cigarette smoke exposure. Br J Pharmacol 2023; 180:2018-2034. [PMID: 36908040 PMCID: PMC10953324 DOI: 10.1111/bph.16068] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 02/07/2023] [Accepted: 03/07/2023] [Indexed: 03/14/2023] Open
Abstract
BACKGROUND AND PURPOSE Cardiovascular disease affects up to half of the patients with chronic obstructive pulmonary disease (COPD), exerting deleterious impact on health outcomes and survivability. Vascular endothelial dysfunction marks the onset of cardiovascular disease. The present study examined the effect of a potent NADPH Oxidase (NOX) inhibitor and free-radical scavenger, apocynin, on COPD-related cardiovascular disease. EXPERIMENTAL APPROACH Male BALB/c mice were exposed to either room air (Sham) or cigarette smoke (CS) generated from 9 cigarettes·day-1 , 5 days a week for up to 24 weeks with or without apocynin treatment (5 mg·kg-1 ·day-1 , intraperitoneal injection). KEY RESULTS Eight-weeks of apocynin treatment reduced airway neutrophil infiltration (by 42%) and completely preserved endothelial function and endothelial nitric oxide synthase (eNOS) availability against the oxidative insults of cigarette smoke exposure. These preservative effects were maintained up until the 24-week time point. 24-week of apocynin treatment markedly reduced airway inflammation (reduced infiltration of macrophage, neutrophil and lymphocyte), lung function decline (hyperinflation) and prevented airway collagen deposition by cigarette smoke exposure. CONCLUSION AND IMPLICATIONS Limiting NOX activity may slow COPD progression and lower cardiovascular disease risk, particularly when signs of oxidative stress become evident.
Collapse
Affiliation(s)
- Stanley M. H. Chan
- Centre for Respiratory Science and Health, School of Health and Biomedical SciencesRMIT UniversityBundooraVictoria3083Australia
| | - Kurt Brassington
- Centre for Respiratory Science and Health, School of Health and Biomedical SciencesRMIT UniversityBundooraVictoria3083Australia
| | - Suleman Abdullah Almerdasi
- Centre for Respiratory Science and Health, School of Health and Biomedical SciencesRMIT UniversityBundooraVictoria3083Australia
| | - Aleksandar Dobric
- Centre for Respiratory Science and Health, School of Health and Biomedical SciencesRMIT UniversityBundooraVictoria3083Australia
| | - Simone N. De Luca
- Centre for Respiratory Science and Health, School of Health and Biomedical SciencesRMIT UniversityBundooraVictoria3083Australia
| | - Madison Coward‐Smith
- Centre for Respiratory Science and Health, School of Health and Biomedical SciencesRMIT UniversityBundooraVictoria3083Australia
| | - Hao Wang
- Centre for Respiratory Science and Health, School of Health and Biomedical SciencesRMIT UniversityBundooraVictoria3083Australia
| | - Kevin Mou
- Centre for Respiratory Science and Health, School of Health and Biomedical SciencesRMIT UniversityBundooraVictoria3083Australia
| | - Alina Akhtar
- Centre for Respiratory Science and Health, School of Health and Biomedical SciencesRMIT UniversityBundooraVictoria3083Australia
| | - Rana Abdullah Alateeq
- Centre for Respiratory Science and Health, School of Health and Biomedical SciencesRMIT UniversityBundooraVictoria3083Australia
| | - Wei Wang
- Centre for Respiratory Science and Health, School of Health and Biomedical SciencesRMIT UniversityBundooraVictoria3083Australia
| | - Huei Jiunn Seow
- Centre for Respiratory Science and Health, School of Health and Biomedical SciencesRMIT UniversityBundooraVictoria3083Australia
| | - Stavros Selemidis
- Centre for Respiratory Science and Health, School of Health and Biomedical SciencesRMIT UniversityBundooraVictoria3083Australia
| | - Steven Bozinovski
- Centre for Respiratory Science and Health, School of Health and Biomedical SciencesRMIT UniversityBundooraVictoria3083Australia
| | - Ross Vlahos
- Centre for Respiratory Science and Health, School of Health and Biomedical SciencesRMIT UniversityBundooraVictoria3083Australia
| |
Collapse
|
126
|
Araya A, Gallegos S, Maldonado A, Rivera-Meza M, Chandra R, Lobo MK, Aguayo LG. Overexpression of wild type glycine alpha 1 subunit rescues ethanol sensitivity in accumbal receptors and reduces binge drinking in mice. Neuropsychopharmacology 2023; 48:1367-1376. [PMID: 36175550 PMCID: PMC10353986 DOI: 10.1038/s41386-022-01459-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/12/2022] [Accepted: 09/07/2022] [Indexed: 11/09/2022]
Abstract
The nucleus accumbens (nAc) is a critical region in the brain reward system since it integrates abundant synaptic inputs contributing to the control of neuronal excitability in the circuit. The presence of inhibitory α1 glycine receptor (GlyRs) subunits, sensitive to ethanol, has been recently reported in accumbal neurons suggesting that they are protective against excessive binge consumption. In the present study, we used viral vectors (AAV) to overexpress mutant and WT α1 subunits in accumbal neurons in D1 Cre and α1 KI mice. Injection of a Cre-inducible AAV carrying an ethanol insensitive α1 subunit in D1 Cre neurons was unable to affect sensitivity to ethanol in GlyRs or affect ethanol drinking. On the other hand, using an AAV that transduced WT α1 GlyRs in GABAergic neurons in the nAc of high-ethanol consuming mice caused a reduction in ethanol intake as reflected by lowered drinking in the dark and reduced blood ethanol concentration. As expected, the AAV increased the glycine current density by 5-fold without changing the expression of GABAA receptors. Examination of the ethanol sensitivity in isolated accumbal neurons indicated that the GlyRs phenotype changed from an ethanol resistant to an ethanol sensitive type. These results support the conclusion that increased inhibition in the nAc can control excessive ethanol consumption and that selective targeting of GlyRs by pharmacotherapy might provide a mechanistic procedure to reduce ethanol binge.
Collapse
Affiliation(s)
- Anibal Araya
- Laboratory of Neurophysiology, Department of Physiology, Universidad de Concepción, Concepcion, Chile
| | - Scarlet Gallegos
- Laboratory of Neurophysiology, Department of Physiology, Universidad de Concepción, Concepcion, Chile
| | - Adolfo Maldonado
- Laboratory of Experimental Pharmacology, Department of Pharmacological and Toxicological Chemistry, Faculty of Chemical Sciences and Pharmacy, Universidad de Chile, Santiago, Chile
| | - Mario Rivera-Meza
- Laboratory of Experimental Pharmacology, Department of Pharmacological and Toxicological Chemistry, Faculty of Chemical Sciences and Pharmacy, Universidad de Chile, Santiago, Chile
| | - Ramesh Chandra
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Mary Kay Lobo
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Luis G Aguayo
- Laboratory of Neurophysiology, Department of Physiology, Universidad de Concepción, Concepcion, Chile.
| |
Collapse
|
127
|
DeBaker MC, Mitten EH, Rose TR, Marron Fernandez de Velasco E, Gao R, Lee AM, Wickman K. RGS6 negatively regulates inhibitory G protein signaling in dopamine neurons and positively regulates binge-like alcohol consumption in mice. Br J Pharmacol 2023; 180:2140-2155. [PMID: 36929333 PMCID: PMC10504421 DOI: 10.1111/bph.16071] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 02/02/2023] [Accepted: 03/13/2023] [Indexed: 03/18/2023] Open
Abstract
BACKGROUND AND PURPOSE Drugs of abuse, including alcohol, increase dopamine in the mesocorticolimbic system via actions on dopamine neurons in the ventral tegmental area (VTA). Increased dopamine transmission can activate inhibitory G protein signalling pathways in VTA dopamine neurons, including those controlled by GABAB and D2 receptors. Members of the R7 subfamily of regulator of G protein signalling (RGS) proteins can regulate inhibitory G protein signalling, but their influence on VTA dopamine neurons is unclear. Here, we investigated the influence of RGS6, an R7 RGS family memberthat has been implicated in the regulation of alcohol consumption in mice, on inhibitory G protein signalling in VTA dopamine neurons. EXPERIMENTAL APPROACH We used molecular, electrophysiological and genetic approaches to probe the impact of RGS6 on inhibitory G protein signalling in VTA dopamine neurons and on binge-like alcohol consumption in mice. KEY RESULTS RGS6 is expressed in adult mouse VTA dopamine neurons and it modulates inhibitory G protein signalling in a receptor-dependent manner, tempering D2 receptor-induced somatodendritic currents and accelerating deactivation of synaptically evoked GABAB receptor-dependent responses. RGS6-/- mice exhibit diminished binge-like alcohol consumption, a phenotype replicated in female (but not male) mice lacking RGS6 selectively in VTA dopamine neurons. CONCLUSIONS AND IMPLICATIONS RGS6 negatively regulates GABAB - and D2 receptor-dependent inhibitory G protein signalling pathways in mouse VTA dopamine neurons and exerts a sex-dependent positive influence on binge-like alcohol consumption in adult mice. As such, RGS6 may represent a new diagnostic and/or therapeutic target for alcohol use disorder.
Collapse
Affiliation(s)
- Margot C. DeBaker
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN
| | - Eric H. Mitten
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN
| | - Timothy R. Rose
- Department of Pharmacology, University of Minnesota, Minneapolis, MN
| | | | - Runbo Gao
- Department of Pharmacology, University of Minnesota, Minneapolis, MN
| | - Anna M. Lee
- Department of Pharmacology, University of Minnesota, Minneapolis, MN
| | - Kevin Wickman
- Department of Pharmacology, University of Minnesota, Minneapolis, MN
| |
Collapse
|
128
|
Borgonetti V, Galeotti N. Honokiol-Rich Magnolia officinalis Bark Extract Attenuates Trauma-Induced Neuropathic Pain. Antioxidants (Basel) 2023; 12:1518. [PMID: 37627513 PMCID: PMC10451803 DOI: 10.3390/antiox12081518] [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: 06/28/2023] [Revised: 07/23/2023] [Accepted: 07/25/2023] [Indexed: 08/27/2023] Open
Abstract
Neuropathic pain (NP) affects about 8% of the general population. Current analgesic therapies have limited efficacy, making NP one of the most difficult to treat pain conditions. Evidence indicates that excessive oxidative stress can contribute to the onset of chronic NP and several natural antioxidant compounds have shown promising efficacy in NP models. Thus, this study aimed to investigate the pain-relieving activity of honokiol (HNK)-rich standardized extract of Magnolia officinalis Rehder & E. Wilson bark (MOE), well known for its antioxidant and anti-inflammatory properties, in the spared nerve injury (SNI) model. The molecular mechanisms and efficacy toward neuroinflammation were investigated in spinal cord samples from SNI mice and LPS-stimulated BV2 microglia cells. MOE and HNK showed antioxidant activity. MOE (30 mg/kg p.o.) produced an antiallodynic effect in SNI mice in the absence of locomotor impairment, reduced spinal p-p38, p-JNK1, iNOS, p-p65, IL-1ß, and Nrf2 overexpression, increased IL-10 and MBP levels and attenuated the Notch signaling pathway by reducing Jagged1 and NEXT. These effects were prevented by the CB1 antagonist AM251. HNK reduced the proinflammatory response of LPS-stimulated BV2 and reduced Jagged1 overexpression. MOE and HNK, by modulating oxidative and proinflammatory responses, might represent interesting candidates for NP management.
Collapse
Affiliation(s)
| | - Nicoletta Galeotti
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology and Toxicology, University of Florence, Viale G. Pieraccini 6, 50139 Florence, Italy;
| |
Collapse
|
129
|
Bergen P, Munro BA, Pang DSJ. Quality of reporting of prospective in vivo and ex vivo studies published in the Journal of Veterinary Emergency and Critical Care over a 10-year period (2009-2019). J Vet Emerg Crit Care (San Antonio) 2023; 33:435-441. [PMID: 37436848 DOI: 10.1111/vec.13312] [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: 01/03/2022] [Revised: 05/29/2022] [Accepted: 05/31/2022] [Indexed: 07/14/2023]
Abstract
OBJECTIVE To evaluate the reporting of key items associated with risk of bias and weak study design over a 10-year period. DESIGN Literature survey. SETTING Not applicable. ANIMALS Not applicable. INTERVENTIONS Papers published in the Journal of Veterinary Emergency and Critical Care between 2009 and 2019 were screened for inclusion. Inclusion criteria consisted of prospective experimental studies describing in vivo or ex vivo research (or both), containing at least 2 comparison groups. Identified papers had identifying information (publication date, volume and issue, authors, affiliations) redacted by an individual not involved with paper selection or review. Two reviewers independently reviewed all papers and applied an operationalized checklist to categorize item reporting as fully reported, partially reported, not reported, or not applicable. Items assessed included randomization, blinding, data handling (inclusions and exclusions), and sample size estimation. Differences in assessment between reviewers were resolved by consensus with a third reviewer. A secondary aim was to document availability of data used to generate study results. Papers were screened for links to access data in the text and supporting information. MEASUREMENTS AND MAIN RESULTS After screening, 109 papers were included. Eleven papers were excluded during full-text review, with 98 papers included in the final analysis. Randomization was fully reported in 31.6% of papers (31/98). Blinding was fully reported in 31.6% of papers (31/98). Inclusion criteria were fully reported in all papers. Exclusion criteria were fully reported in 60.2% of papers (59/98). Sample size estimation was fully reported in 8.0% of papers (6/75). No papers (0/99) made data freely available without a requirement to contact study authors. CONCLUSIONS There is substantial room for improvement in reporting of randomization, blinding, data exclusions, and sample size estimations. Evaluation of study quality by readers is limited by the low reporting levels identified, and the risk of bias present indicates a potential for inflated effect sizes.
Collapse
Affiliation(s)
- Paige Bergen
- Department of Psychology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Brittany A Munro
- Department of Veterinary Clinical and Diagnostic Sciences, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Daniel S J Pang
- Department of Veterinary Clinical and Diagnostic Sciences, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Université de Montréal, Québec, Canada
| |
Collapse
|
130
|
Carpi S, Quarta S, Doccini S, Saviano A, Marigliano N, Polini B, Massaro M, Carluccio MA, Calabriso N, Wabitsch M, Santorelli FM, Cecchini M, Maione F, Nieri P, Scoditti E. Tanshinone IIA and Cryptotanshinone Counteract Inflammation by Regulating Gene and miRNA Expression in Human SGBS Adipocytes. Biomolecules 2023; 13:1029. [PMID: 37509065 PMCID: PMC10377153 DOI: 10.3390/biom13071029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/14/2023] [Accepted: 06/21/2023] [Indexed: 07/30/2023] Open
Abstract
Inflammation of the adipose tissue contributes to the onset and progression of several chronic obesity-related diseases. The two most important lipophilic diterpenoid compounds found in the root of Salvia milthorrhiza Bunge (also called Danshen), tanshinone IIA (TIIA) and cryptotanshinone (CRY), have many favorable pharmacological effects. However, their roles in obesity-associated adipocyte inflammation and related sub-networks have not been fully elucidated. In the present study, we investigated the gene, miRNAs and protein expression profile of prototypical obesity-associated dysfunction markers in inflamed human adipocytes treated with TIIA and CRY. The results showed that TIIA and CRY prevented tumor necrosis factor (TNF)-α induced inflammatory response in adipocytes, by counter-regulating the pattern of secreted cytokines/chemokines associated with adipocyte inflammation (CCL2/MCP-1, CXCL10/IP-10, CCL5/RANTES, CXCL1/GRO-α, IL-6, IL-8, MIF and PAI-1/Serpin E1) via the modulation of gene expression (as demonstrated for CCL2/MCP-1, CXCL10/IP-10, CCL5/RANTES, CXCL1/GRO-α, and IL-8), as well as related miRNA expression (miR-126-3p, miR-223-3p, miR-124-3p, miR-155-5p, and miR-132-3p), and by attenuating monocyte recruitment. This is the first demonstration of a beneficial effect by TIIA and CRY on adipocyte dysfunction associated with obesity development and complications, offering a new outlook for the prevention and/or treatment of metabolic diseases.
Collapse
Affiliation(s)
- Sara Carpi
- Science of Health Department, Magna Græcia University, 88100 Catanzaro, Italy
- NEST, Istituto Nanoscienze-CNR and Scuola Normale Superiore, 56100 Pisa, Italy
- Department of Pharmacy, University of Pisa, 56100 Pisa, Italy
| | - Stefano Quarta
- Department of Biological and Environmental Sciences and Technologies (DISTEBA), University of Salento, 73100 Lecce, Italy
| | - Stefano Doccini
- IRCCS Fondazione Stella Maris, Calambrone, 56128 Pisa, Italy
| | - Anella Saviano
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Noemi Marigliano
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Beatrice Polini
- Department of Pharmacy, University of Pisa, 56100 Pisa, Italy
- Department of Pathology, University of Pisa, 56100 Pisa, Italy
| | - Marika Massaro
- National Research Council (CNR), Institute of Clinical Physiology (IFC), 73100 Lecce, Italy
| | | | - Nadia Calabriso
- National Research Council (CNR), Institute of Clinical Physiology (IFC), 73100 Lecce, Italy
| | - Martin Wabitsch
- Division of Pediatric Endocrinology, Diabetes and Obesity, Department of Pediatrics and Adolescent Medicine, University of Ulm, 89075 Ulm, Germany
| | | | - Marco Cecchini
- NEST, Istituto Nanoscienze-CNR and Scuola Normale Superiore, 56100 Pisa, Italy
| | - Francesco Maione
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Paola Nieri
- Department of Pharmacy, University of Pisa, 56100 Pisa, Italy
| | - Egeria Scoditti
- National Research Council (CNR), Institute of Clinical Physiology (IFC), 73100 Lecce, Italy
| |
Collapse
|
131
|
de Oliveira Formiga R, Amaral FC, Souza CF, Mendes DAGB, Wanderley CWS, Lorenzini CB, Santos AA, Antônia J, Faria LF, Natale CC, Paula NM, Silva PCS, Fonseca FR, Aires L, Heck N, Starick MR, Queiroz‐Junior CM, Santos FRS, de Souza FRO, Costa VV, Barroso SPC, Morrot A, Van Weyenbergh J, Sordi R, Alisson‐Silva F, Cunha FQ, Rocha EL, Chollet‐Martin S, Hurtado‐Nedelec MM, Martin C, Burgel P, Mansur DS, Maurici R, Macauley MS, Báfica A, Witko‐Sarsat V, Spiller F. Neuraminidase is a host-directed approach to regulate neutrophil responses in sepsis and COVID-19. Br J Pharmacol 2023; 180:1460-1481. [PMID: 36526272 PMCID: PMC9877938 DOI: 10.1111/bph.16013] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 07/29/2022] [Accepted: 08/16/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND AND PURPOSE Neutrophil overstimulation plays a crucial role in tissue damage during severe infections. Because pathogen-derived neuraminidase (NEU) stimulates neutrophils, we investigated whether host NEU can be targeted to regulate the neutrophil dysregulation observed in severe infections. EXPERIMENTAL APPROACH The effects of NEU inhibitors on lipopolysaccharide (LPS)-stimulated neutrophils from healthy donors or COVID-19 patients were determined by evaluating the shedding of surface sialic acids, cell activation, and reactive oxygen species (ROS) production. Re-analysis of single-cell RNA sequencing of respiratory tract samples from COVID-19 patients also was carried out. The effects of oseltamivir on sepsis and betacoronavirus-induced acute lung injury were evaluated in murine models. KEY RESULTS Oseltamivir and zanamivir constrained host NEU activity, surface sialic acid release, cell activation, and ROS production by LPS-activated human neutrophils. Mechanistically, LPS increased the interaction of NEU1 with matrix metalloproteinase 9 (MMP-9). Inhibition of MMP-9 prevented LPS-induced NEU activity and neutrophil response. In vivo, treatment with oseltamivir fine-tuned neutrophil migration and improved infection control as well as host survival in peritonitis and pneumonia sepsis. NEU1 also is highly expressed in neutrophils from COVID-19 patients, and treatment of whole-blood samples from these patients with either oseltamivir or zanamivir reduced neutrophil overactivation. Oseltamivir treatment of intranasally infected mice with the mouse hepatitis coronavirus 3 (MHV-3) decreased lung neutrophil infiltration, viral load, and tissue damage. CONCLUSION AND IMPLICATIONS These findings suggest that interplay of NEU1-MMP-9 induces neutrophil overactivation. In vivo, NEU may serve as a host-directed target to dampen neutrophil dysfunction during severe infections.
Collapse
Affiliation(s)
- Rodrigo de Oliveira Formiga
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
- Université de Paris, Institut Cochin, INSERM U1016, CNRSParisFrance
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Flávia C. Amaral
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Camila F. Souza
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Daniel A. G. B. Mendes
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Carlos W. S. Wanderley
- Department of Pharmacology, School of Medicine of Ribeirão PretoUniversity of São PauloRibeirão PretoBrazil
| | - Cristina B. Lorenzini
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Adara A. Santos
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Juliana Antônia
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Lucas F. Faria
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Caio C. Natale
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Nicholas M. Paula
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Priscila C. S. Silva
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Fernanda R. Fonseca
- Department of Clinical MedicineFederal University of Santa CatarinaFlorianópolisBrazil
| | - Luan Aires
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Nicoli Heck
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Márick R. Starick
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Celso M. Queiroz‐Junior
- Department of Morphology, Institute of Biological SciencesFederal University of Minas GeraisBelo HorizonteBrazil
| | - Felipe R. S. Santos
- Department of Biochemistry and Immunology, Institute of Biological SciencesFederal University of Minas GeraisBelo HorizonteBrazil
| | - Filipe R. O. de Souza
- Department of Morphology, Institute of Biological SciencesFederal University of Minas GeraisBelo HorizonteBrazil
| | - Vivian V. Costa
- Department of Morphology, Institute of Biological SciencesFederal University of Minas GeraisBelo HorizonteBrazil
| | - Shana P. C. Barroso
- Molecular Biology Laboratory, Institute of Biomedical ResearchMarcilio Dias Naval Hospital, Navy of BrazilRio de JaneiroBrazil
| | - Alexandre Morrot
- Tuberculosis Research Laboratory, Faculty of MedicineFederal University of Rio de JaneiroRio de JaneiroBrazil
- Immunoparasitology LaboratoryOswaldo Cruz Foundation (FIOCRUZ)Rio de JaneiroBrazil
| | - Johan Van Weyenbergh
- Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory for Clinical and Epidemiological VirologyKU LeuvenLeuvenBelgium
| | - Regina Sordi
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Frederico Alisson‐Silva
- Department of Immunology, Paulo de Goes Institute of MicrobiologyFederal University of Rio de JaneiroRio de JaneiroBrazil
| | - Fernando Q. Cunha
- Department of Pharmacology, School of Medicine of Ribeirão PretoUniversity of São PauloRibeirão PretoBrazil
| | - Edroaldo L. Rocha
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Sylvie Chollet‐Martin
- INSERM UMR 996, ‘Infammation, Microbiome and Immunosurveillance’, Faculty of PharmacyUniversité Paris‐SaclayChâtenay‐MalabryFrance
| | | | - Clémence Martin
- Université de Paris, Institut Cochin, INSERM U1016, CNRSParisFrance
- Department of PneumologyAP‐HP, Hôpital CochinParisFrance
| | - Pierre‐Régis Burgel
- Université de Paris, Institut Cochin, INSERM U1016, CNRSParisFrance
- Department of PneumologyAP‐HP, Hôpital CochinParisFrance
| | - Daniel S. Mansur
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Rosemeri Maurici
- Department of Clinical MedicineFederal University of Santa CatarinaFlorianópolisBrazil
| | - Matthew S. Macauley
- Department of Chemistry, Department of Medical Microbiology and ImmunologyUniversity of AlbertaEdmontonAlbertaCanada
| | - André Báfica
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | | | - Fernando Spiller
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| |
Collapse
|
132
|
Dedoni S, Scherma M, Camoglio C, Siddi C, Fratta W, Fadda P. Anaplastic Lymphoma Kinase Receptor: Possible Involvement in Anorexia Nervosa. Nutrients 2023; 15:2205. [PMID: 37432348 DOI: 10.3390/nu15092205] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/02/2023] [Accepted: 05/04/2023] [Indexed: 07/12/2023] Open
Abstract
The pathophysiology of Anorexia Nervosa (AN) has not been fully elucidated. Anaplastic lymphoma kinase (ALK) receptor is a protein-tyrosine kinase mainly known as a key oncogenic driver. Recently, a genetic deletion of ALK in mice has been found to increase energy expenditure and confers resistance to obesity in these animals, suggesting its role in the regulation of thinness. Here, we investigated the expression of ALK and the downstream intracellular pathways in female rats subjected to the activity-based anorexia (ABA) model, which reproduces important features of human AN. In the hypothalamic lysates of ABA rats, we found a reduction in ALK receptor expression, a downregulation of Akt phosphorylation, and no change in the extracellular signal-regulated protein kinases 1 and 2 (ERK1/2) phosphorylation. After the recovery from body weight loss, ALK receptor expression returned to the control baseline values, while it was again suppressed during a second cycle of ABA induction. Overall, this evidence suggests a possible involvement of the ALK receptor in the pathophysiology of AN, that may be implicated in its stabilization, resistance, and/or its exacerbation.
Collapse
Affiliation(s)
- Simona Dedoni
- Section of Neuroscience and Clinical Pharmacology, Department of Biomedical Science, University of Cagliari, 09124 Cagliari, Italy
| | - Maria Scherma
- Section of Neuroscience and Clinical Pharmacology, Department of Biomedical Science, University of Cagliari, 09124 Cagliari, Italy
| | - Chiara Camoglio
- Section of Neuroscience and Clinical Pharmacology, Department of Biomedical Science, University of Cagliari, 09124 Cagliari, Italy
| | - Carlotta Siddi
- Section of Neuroscience and Clinical Pharmacology, Department of Biomedical Science, University of Cagliari, 09124 Cagliari, Italy
| | - Walter Fratta
- Section of Neuroscience and Clinical Pharmacology, Department of Biomedical Science, University of Cagliari, 09124 Cagliari, Italy
| | - Paola Fadda
- Section of Neuroscience and Clinical Pharmacology, Department of Biomedical Science, University of Cagliari, 09124 Cagliari, Italy
- Neuroscience Institute, Section of Cagliari, National Research Council of Italy (CNR), 09042 Cagliari, Italy
| |
Collapse
|
133
|
Borgonetti V, Galeotti N. Posttranscriptional Regulation of Gene Expression Participates in the Myelin Restoration in Mouse Models of Multiple Sclerosis: Antisense Modulation of HuR and HuD ELAV RNA Binding Protein. Mol Neurobiol 2023; 60:2661-2677. [PMID: 36696009 PMCID: PMC10039839 DOI: 10.1007/s12035-023-03236-8] [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: 07/26/2022] [Accepted: 01/13/2023] [Indexed: 01/26/2023]
Abstract
Neuropathic pain is the most difficult-to-treat pain syndrome in multiple sclerosis. Evidence relates neuropathic pain to demyelination, which often originates from unresolved neuroinflammation or altered immune response. Posttranscriptional regulation of gene expression might play a fundamental role in the regulation of these processes. The ELAV RNA-binding proteins HuR and HuD are involved in the promotion of inflammatory phenomena and in neuronal development and maintenance, respectively. Thus, the aim of this study was to investigate the role of HuR and HuD in demyelination-associated neuropathic pain in the mouse experimental autoimmune encephalomyelitis (EAE) model. HuR resulted overexpressed in the spinal cord of MOG35-55-EAE and PLP139-151-EAE mice and was detected in CD11b + cells. Conversely, HuD was largely downregulated in the MOG-EAE spinal cord, along with GAP43 and neurofilament H, while in PLP-EAE mice, HuD and neuronal markers remained unaltered. Intranasal antisense oligonucleotide (ASO) delivery to knockdown HuR, increased myelin basic protein expression, and Luxol Fast Blue staining in both EAE models, an indication of increased myelin content. These effects temporally coincided with attenuation of pain hypersensitivity. Anti-HuR ASO increased the expression of HuD in GAP43-expressing cells and promoted a HuD-mediated neuroprotective activity in MOG-EAE mice, while in PLP-EAE mice, HuR silencing dampened pro-inflammatory responses mediated by spinal microglia activation. In conclusion, anti-HuR ASO showed myelin protection at analgesic doses with multitarget mechanisms, and it deserves further consideration as an innovative agent to counteract demyelination in neuropathic pain states.
Collapse
Affiliation(s)
- Vittoria Borgonetti
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology, University of Florence, Viale G. Pieraccini 6, I-50139, Florence, Italy
| | - Nicoletta Galeotti
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology, University of Florence, Viale G. Pieraccini 6, I-50139, Florence, Italy.
| |
Collapse
|
134
|
Meng J, Abu YF, Zhang Y, Zhou Y, Xie Y, Yan Y, Tao J, Ramakrishnan S, Chen C, Roy S. Opioid-induced microbial dysbiosis disrupts irinotecan (CPT-11) metabolism and increases gastrointestinal toxicity in a murine model. Br J Pharmacol 2023; 180:1362-1378. [PMID: 36562107 PMCID: PMC10089971 DOI: 10.1111/bph.16020] [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: 01/02/2022] [Revised: 12/07/2022] [Accepted: 12/18/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND AND PURPOSE Opioids are commonly used for the management of cancer-associated pain and chemotherapy-induced diarrhoea. The chemotherapeutic irinotecan (CPT-11) causes severe gastrointestinal (GI) toxicity due to deconjugation of inactive metabolite SN-38 glucuronide (SN-38G) by bacterial β-glucuronidases to the active 7-ethyl-10-hydroxycamptothecin (SN-38). Opioids are known to cause gut microbial dysbiosis, this study evaluated whether CPT-11 anti-tumour efficacy and GI toxicity are exacerbated by opioid co-administration. EXPERIMENTAL APPROACH Eight-week-old C57BL/6 male mice were co-administration with CPT-11 ± opioid. 16S rRNA sequencing was used for gut microbiome analysis. LC-MS analyses of plasma and intestinal extracts were performed to investigate the pharmacokinetic profile of CPT-11. Histological analysis and quantitative real-time polymerase chain reaction were used to determine the severity of intestinal tissue damage. Human liver microsome In vitro assay was performed to confirm the effects of opioids on CPT-11 metabolism. KEY RESULTS Gut microbiome analysis showed that morphine treatment induced enrichment of β-glucuronidase-producing bacteria in the intestines of CPT-11-treated mice, resulting in SN-38 accumulation and exacerbation of GI toxicity in the small intestine. Oral administration of both antibiotics and glucuronidase inhibitor protected mice against GI toxicity induced with CPT-11 and morphine co-administration, implicating a microbiome-dependent mechanism. Additionally, morphine and loperamide decreased the plasma concentration of SN-38 and compromised CPT-11 anti-tumour efficacy, this seemed to be microbiome independent. CONCLUSION AND IMPLICATIONS Gut microbiota play a significant role in opioid and chemotherapeutic agent drug-drug interactions. Inhibition of gut microbial glucuronidase may also prevent adverse GI effects of CPT-11 in patients on opioids.
Collapse
Affiliation(s)
- Jingjing Meng
- Department of Surgery, University of Miami, Miami, FL 33136
| | - Yaa F. Abu
- Department of Microbiology and Immunology, University of Miami, Miami, FL 33136
| | - Yue Zhang
- Department of Surgery, University of Miami, Miami, FL 33136
| | - Yuyin Zhou
- Department of Food Science and Nutrition, University of Minnesota, MN 55108
| | - Yun Xie
- Department of Food Science and Nutrition, University of Minnesota, MN 55108
| | - Yan Yan
- Department of Surgery, University of Miami, Miami, FL 33136
| | - Junyi Tao
- Department of Surgery, University of Miami, Miami, FL 33136
| | | | - Chi Chen
- Department of Microbiology and Immunology, University of Miami, Miami, FL 33136
| | - Sabita Roy
- Department of Surgery, University of Miami, Miami, FL 33136
| |
Collapse
|
135
|
Borgonetti V, Anceschi L, Brighenti V, Corsi L, Governa P, Manetti F, Pellati F, Galeotti N. Cannabidiol-rich non-psychotropic Cannabis sativa L. oils attenuate peripheral neuropathy symptoms by regulation of CB2-mediated microglial neuroinflammation. Phytother Res 2023; 37:1924-1937. [PMID: 36583304 DOI: 10.1002/ptr.7710] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 12/02/2022] [Accepted: 12/04/2022] [Indexed: 12/31/2022]
Abstract
Neuropathic pain (NP) is a chronic disease that affects the normal quality of life of patients. To date, the therapies available are only symptomatic and they are unable to reduce the progression of the disease. Many studies reported the efficacy of Cannabis sativa L. (C. sativa) on NP, but no Δ9 -tetrahydrocannabinol (Δ9 -THC)-free extracts have been investigated in detail for this activity so far. The principal aim of this work is to investigate the potential pain-relieving effect of innovative cannabidiol-rich non-psychotropic C. sativa oils, with a high content of terpenes (K2), compared to the same extract devoid of terpenes (K1). Oral administration of K2 (25 mg kg-1 ) induced a rapid and long-lasting relief of pain hypersensitivity in a mice model of peripheral neuropathy. In spinal cord samples, K2 reduced mitogen-activated protein kinase (MAPKs) levels and neuroinflammatory factors. These effects were reverted by the administration of a CB2 antagonist (AM630), but not by a CB1 antagonist (AM251). Conversely, K1 showed a lower efficacy in the absence of CB1/CB2-mediated mechanisms. In LPS-stimulated murine microglial cells (BV2), K2 reduced microglia pro-inflammatory phenotype through the downregulation of histone deacetylase 1 (HDAC-1) and nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor (IKBα) and increased interleukin-10 (IL-10) expression, an important antiinflammatory cytokine. In conclusion, these results suggested that K2 oral administration attenuated NP symptoms by reducing spinal neuroinflammation and underline the important role of the synergism between cannabinoids and terpenes.
Collapse
Affiliation(s)
- Vittoria Borgonetti
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Lisa Anceschi
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, Modena, Italy
| | - Virginia Brighenti
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Lorenzo Corsi
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Paolo Governa
- Department of Biotechnology, Chemistry and Pharmacy - Department of Excellence 2018-2022, University of Siena, Siena, Italy
| | - Fabrizio Manetti
- Department of Biotechnology, Chemistry and Pharmacy - Department of Excellence 2018-2022, University of Siena, Siena, Italy
| | - Federica Pellati
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Nicoletta Galeotti
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology and Toxicology, University of Florence, Florence, Italy
| |
Collapse
|
136
|
Galis P, Bartosova L, Farkasova V, Szobi A, Horvath C, Kovacova D, Adameova A, Rajtik T. Intermittent Hypoxic Preconditioning Plays a Cardioprotective Role in Doxorubicin-Induced Cardiomyopathy. Cardiovasc Toxicol 2023:10.1007/s12012-023-09793-7. [PMID: 37119387 DOI: 10.1007/s12012-023-09793-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 04/14/2023] [Indexed: 05/01/2023]
Abstract
Intermittent hypoxic preconditioning (IHP) is a well-established cardioprotective intervention in models of ischemia/reperfusion injury. Nevertheless, the significance of IHP in different cardiac pathologies remains elusive. In order to investigate the role of IHP and its effects on calcium-dependent signalization in HF, we employed a model of cardiomyopathy induced by doxorubicin (Dox), a widely used drug from the class of cardiotoxic antineoplastics, which was i.p. injected to Wistar rats (4 applications of 4 mg/kg/week). IHP-treated group was exposed to IHP for 2 weeks prior to Dox administration. IHP ameliorated Dox-induced reduction in cardiac output. Western blot analysis revealed increased expression of sarco/endoplasmic reticulum Ca2+-ATPase (SERCA2a) while the expression of hypoxia inducible factor (HIF)-1-α, which is a crucial regulator of hypoxia-inducible genes, was not changed. Animals administered with Dox had further decreased expression of TRPV1 and TRPV4 (transient receptor potential, vanilloid subtype) ion channels along with suppressed Ca2+/calmodulin-dependent protein kinase II (CaMKII) activation. In summary, IHP-mediated improvement in cardiac output in the model of Dox-induced cardiomyopathy is likely a result of increased SERCA2a expression which could implicate IHP as a potential protective intervention in Dox cardiomyopathy, however, further analysis of observed effects is still required.
Collapse
Affiliation(s)
- Peter Galis
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University, Odbojárov 10, 832 32, Bratislava, Slovakia
| | - Linda Bartosova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University, Odbojárov 10, 832 32, Bratislava, Slovakia
| | - Veronika Farkasova
- Institute for Heart Research, Slovak Academy of Sciences, Dúbravská Cesta 9, 841 04, Bratislava, Slovakia
| | - Adrian Szobi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University, Odbojárov 10, 832 32, Bratislava, Slovakia
| | - Csaba Horvath
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University, Odbojárov 10, 832 32, Bratislava, Slovakia
| | - Dominika Kovacova
- Faculty of Medicine, Institute of Pathophysiology, Comenius University, Špitálska 24, 813 72, Bratislava, Slovakia
| | - Adriana Adameova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University, Odbojárov 10, 832 32, Bratislava, Slovakia
- Institute for Heart Research, Slovak Academy of Sciences, Dúbravská Cesta 9, 841 04, Bratislava, Slovakia
| | - Tomas Rajtik
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University, Odbojárov 10, 832 32, Bratislava, Slovakia.
- Institute for Heart Research, Slovak Academy of Sciences, Dúbravská Cesta 9, 841 04, Bratislava, Slovakia.
| |
Collapse
|
137
|
Li F, Zhao X, Xie F, Wang Z, Ding H, Wang W, Jiao R, Pan Y, Kong L. Nuciferine blocks MIB2-mediated CARD6 polyubiquitination and degradation in the amelioration of high fructose-induced liver lipid accumulation. Food Funct 2023; 14:4706-4721. [PMID: 37186242 DOI: 10.1039/d2fo03622c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Dietary alkaloid nuciferine isolated from the leaves of Nelumbo nucifera can ameliorate dyslipidemia and liver lipid accumulation, but the underlying mechanism remains unclear. Caspase recruitment domain protein family member 6 (CARD6) is suggested to play an important role in metabolic diseases. This study aimed to investigate the role and the upstream regulator of CARD6 in high fructose-induced liver lipid accumulation and whether and how the anti-lipid accumulation effect of nuciferine was related to CARD6. Herein, we found that high fructose decreased CARD6 expression and increased ASK1 and JNK1/2 phosphorylation in rat livers and hepatocytes, which were attenuated by nuciferine. Furthermore, after the transfection with HA-CARD6, CARD6 siRNA and MIB2 siRNA, the data showed that CARD6 overexpression blocked high fructose-induced upregulation of ASK1 and JNK1/2 phosphorylation as well as lipid accumulation in hepatocytes. CARD6 siRNA reversed the amelioration of nuciferine to high fructose-induced upregulation of ASK1 and JNK1/2 phosphorylation in hepatocyte lipid accumulation. Mechanistically, high fructose upregulated MIB2 expression by interacting with CARD6 and promoting K48-linked CARD6 polyubiquitination and degradation in high fructose-stimulated hepatocytes which were explored by immunoblotting, immunofluorescence, and immunoprecipitation. However, MIB2 siRNA reversed high fructose-induced downregulation of CARD6 and lipid accumulation in hepatocytes. Notably, nuciferine reduced MIB2 expression and thus decreased K48-linked CARD6 polyubiquitination and degradation in the amelioration of high fructose-induced lipid accumulation in hepatocytes. These results suggested that nuciferine exhibited a protective effect against high fructose-induced liver lipid accumulation through blocking MIB2-mediated CARD6 polyubiquitination and degradation.
Collapse
Affiliation(s)
- Feng Li
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China.
| | - Xiaojuan Zhao
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China.
| | - Fengyu Xie
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China.
| | - Zixuan Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China.
| | - Hong Ding
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China.
| | - Wanru Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China.
| | - Ruiqing Jiao
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China.
| | - Ying Pan
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China.
| | - Lingdong Kong
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China.
| |
Collapse
|
138
|
Jaque-Fernandez F, Allard B, Monteiro L, Lafoux A, Huchet C, Jaimovich E, Berthier C, Jacquemond V. Probenecid affects muscle Ca2+ homeostasis and contraction independently from pannexin channel block. J Gen Physiol 2023; 155:e202213203. [PMID: 36820799 PMCID: PMC9998970 DOI: 10.1085/jgp.202213203] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 12/21/2022] [Accepted: 02/08/2023] [Indexed: 02/24/2023] Open
Abstract
Tight control of skeletal muscle contractile activation is secured by the excitation-contraction (EC) coupling protein complex, a molecular machinery allowing the plasma membrane voltage to control the activity of the ryanodine receptor Ca2+ release channel in the sarcoplasmic reticulum (SR) membrane. This machinery has been shown to be intimately linked to the plasma membrane protein pannexin-1 (Panx1). We investigated whether the prescription drug probenecid, a widely used Panx1 blocker, affects Ca2+ signaling, EC coupling, and muscle force. The effect of probenecid was tested on membrane current, resting Ca2+, and SR Ca2+ release in isolated mouse muscle fibers, using a combination of whole-cell voltage-clamp and Ca2+ imaging, and on electrically triggered contraction of isolated muscles. Probenecid (1 mM) induces SR Ca2+ leak at rest and reduces peak voltage-activated SR Ca2+ release and contractile force by 40%. Carbenoxolone, another Panx1 blocker, also reduces Ca2+ release, but neither a Panx1 channel inhibitory peptide nor a purinergic antagonist affected Ca2+ release, suggesting that probenecid and carbenoxolone do not act through inhibition of Panx1-mediated ATP release and consequently altered purinergic signaling. Probenecid may act by altering Panx1 interaction with the EC coupling machinery, yet the implication of another molecular target cannot be excluded. Since probenecid has been used both in the clinic and as a masking agent for doping in sports, these results should encourage evaluation of possible effects on muscle function in treated individuals. In addition, they also raise the question of whether probenecid-induced altered Ca2+ homeostasis may be shared by other tissues.
Collapse
Affiliation(s)
- Francisco Jaque-Fernandez
- Université Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5261, INSERM U-1315, Institut NeuroMyoGène—Pathophysiology and Genetics of Neuron and Muscle, Lyon, France
| | - Bruno Allard
- Université Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5261, INSERM U-1315, Institut NeuroMyoGène—Pathophysiology and Genetics of Neuron and Muscle, Lyon, France
| | - Laloé Monteiro
- Université Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5261, INSERM U-1315, Institut NeuroMyoGène—Pathophysiology and Genetics of Neuron and Muscle, Lyon, France
| | - Aude Lafoux
- Therassay Platform, CAPACITES, Université de Nantes, Nantes, France
| | - Corinne Huchet
- Therassay Platform, CAPACITES, Université de Nantes, Nantes, France
- Nantes Gene Therapy Laboratory, Université de Nantes, INSERM UMR 1089, Nantes, France
| | - Enrique Jaimovich
- Centro de Estudios Moleculares de la Célula, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Christine Berthier
- Université Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5261, INSERM U-1315, Institut NeuroMyoGène—Pathophysiology and Genetics of Neuron and Muscle, Lyon, France
| | - Vincent Jacquemond
- Université Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5261, INSERM U-1315, Institut NeuroMyoGène—Pathophysiology and Genetics of Neuron and Muscle, Lyon, France
| |
Collapse
|
139
|
Fialho MFP, Brum ES, Becker G, Brusco I, Oliveira SM. Kinin B2 and B1 Receptors Activation Sensitize the TRPA1 Channel Contributing to Anastrozole-Induced Pain Symptoms. Pharmaceutics 2023; 15:pharmaceutics15041136. [PMID: 37111622 PMCID: PMC10143169 DOI: 10.3390/pharmaceutics15041136] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/25/2023] [Accepted: 03/30/2023] [Indexed: 04/07/2023] Open
Abstract
Aromatase inhibitors (AIs) cause symptoms of musculoskeletal pain, and some mechanisms have been proposed to explain them. However, signaling pathways downstream from kinin B2 (B2R) and B1 (B1R) receptor activation and their possible sensitizing of the Transient Receptor Potential Ankyrin 1 (TRPA1) remain unknown. The interaction between the kinin receptor and the TRPA1 channel in male C57BL/6 mice treated with anastrozole (an AI) was evaluated. PLC/PKC and PKA inhibitors were used to evaluate the signaling pathways downstream from B2R and B1R activation and their effect on TRPA1 sensitization. Anastrozole caused mechanical allodynia and muscle strength loss in mice. B2R (Bradykinin), B1R (DABk), or TRPA1 (AITC) agonists induced overt nociceptive behavior and enhanced and prolonged the painful parameters in anastrozole-treated mice. All painful symptoms were reduced by B2R (Icatibant), B1R (DALBk), or TRPA1 (A967079) antagonists. We observed the interaction between B2R, B1R, and the TRPA1 channel in anastrozole-induced musculoskeletal pain, which was dependent on the activation of the PLC/PKC and PKA signaling pathways. TRPA1 seems to be sensitized by mechanisms dependent on the activation of PLC/PKC, and PKA due to kinin receptors stimulation in anastrozole-treated animals. Thus, regulating this signaling pathway could contribute to alleviating AIs-related pain symptoms, patients’ adherence to therapy, and disease control.
Collapse
Affiliation(s)
- Maria Fernanda Pessano Fialho
- Graduate Program in Biological Sciences, Biochemical Toxicology, Center of Natural and Exact Sciences, Federal University of Santa Maria, Camobi, Santa Maria 97105-900, RS, Brazil
| | - Evelyne Silva Brum
- Graduate Program in Biological Sciences, Biochemical Toxicology, Center of Natural and Exact Sciences, Federal University of Santa Maria, Camobi, Santa Maria 97105-900, RS, Brazil
| | - Gabriela Becker
- Graduate Program in Biological Sciences, Biochemical Toxicology, Center of Natural and Exact Sciences, Federal University of Santa Maria, Camobi, Santa Maria 97105-900, RS, Brazil
| | - Indiara Brusco
- Graduate Program in Biological Sciences, Biochemical Toxicology, Center of Natural and Exact Sciences, Federal University of Santa Maria, Camobi, Santa Maria 97105-900, RS, Brazil
| | - Sara Marchesan Oliveira
- Graduate Program in Biological Sciences, Biochemical Toxicology, Center of Natural and Exact Sciences, Federal University of Santa Maria, Camobi, Santa Maria 97105-900, RS, Brazil
- Department of Biochemical and Molecular Biology, Center of Natural and Exact Sciences, Federal University of Santa Maria, Camobi, Santa Maria 97105-900, RS, Brazil
| |
Collapse
|
140
|
Groom S, Blum NK, Conibear AE, Disney A, Hill R, Husbands SM, Li Y, Toll L, Kliewer A, Schulz S, Henderson G, Kelly E, Bailey CP. A novel G protein-biased agonist at the μ opioid receptor induces substantial receptor desensitisation through G protein-coupled receptor kinase. Br J Pharmacol 2023; 180:943-957. [PMID: 33245558 DOI: 10.1111/bph.15334] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 11/04/2020] [Accepted: 11/16/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND AND PURPOSE G protein-biased μ opioid receptor agonists have the potential to induce less receptor desensitisation and tolerance than balanced opioids. Here, we investigated if the cyclic endomorphin analogue Tyr-c[D-Lys-Phe-Tyr-Gly] (Compound 1) is a G protein-biased μ agonist and characterised its ability to induce rapid receptor desensitisation in mammalian neurones. EXPERIMENTAL APPROACH The signalling and trafficking properties of opioids were characterised using bioluminescence resonance energy transfer assays, enzyme-linked immunosorbent assay and phosphosite-specific immunoblotting in human embryonic kidney 293 cells. Desensitisation of opioid-induced currents were studied in rat locus coeruleus neurones using whole-cell patch-clamp electrophysiology. The mechanism of Compound 1-induced μ receptor desensitisation was probed using kinase inhibitors. KEY RESULTS Compound 1 has similar intrinsic activity for G protein signalling as morphine. As predicted for a G protein-biased μ agonist, Compound 1 induced minimal agonist-induced internalisation and phosphorylation at intracellular μ receptor serine/threonine residues known to be involved in G protein-coupled receptor kinase (GRK)-mediated desensitisation. However, Compound 1 induced robust rapid μ receptor desensitisation in locus coeruleus neurons, to a greater degree than morphine. The extent of Compound 1-induced desensitisation was unaffected by activation or inhibition of protein kinase C (PKC) but was significantly reduced by inhibition of GRK. CONCLUSION AND IMPLICATIONS Compound 1 is a novel G protein-biased μ agonist that induces substantial rapid receptor desensitisation in mammalian neurons. Surprisingly, Compound 1-induced desensitisation was demonstrated to be GRK dependent despite its G protein bias. Our findings refute the assumption that G protein-biased agonists will evade receptor desensitisation and tolerance. LINKED ARTICLES This article is part of a themed issue on Advances in Opioid Pharmacology at the Time of the Opioid Epidemic. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v180.7/issuetoc.
Collapse
Affiliation(s)
- Sam Groom
- Department of Pharmacy and Pharmacology, University of Bath, Bath, UK.,School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Nina K Blum
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | - Alexandra E Conibear
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Alexander Disney
- Department of Pharmacy and Pharmacology, University of Bath, Bath, UK
| | - Rob Hill
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK.,School of Life Sciences, University of Nottingham, Nottingham, UK
| | | | - Yangmei Li
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Columbia, South Carolina, USA
| | - Lawrence Toll
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, Florida, USA
| | - Andrea Kliewer
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | - Stefan Schulz
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | - Graeme Henderson
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Eamonn Kelly
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Chris P Bailey
- Department of Pharmacy and Pharmacology, University of Bath, Bath, UK
| |
Collapse
|
141
|
Liao W, Foo HYC, Tran TNQ, Chai CLL, Wong WSF. Calcaratarin D, a labdane diterpenoid, attenuates mouse asthma via modulating alveolar macrophage function. Br J Pharmacol 2023; 180:1056-1071. [PMID: 36440573 DOI: 10.1111/bph.15993] [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: 06/26/2022] [Revised: 10/04/2022] [Accepted: 11/16/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Alveolar macrophages (AMs) contribute to airway inflammation and remodelling in allergic asthma. Calcaratarin D (CalD), a labdane diterpenoid from rhizomes of the medicinal plant Alpinia calcarata, has recently been shown to possess anti-inflammatory properties. The present study evaluated protective effects of CalD in a house dust mite (HDM)-induced asthma mouse model. EXPERIMENTAL APPROACH The effects of CalD on AMs in contributing to anti-inflammatory effects in asthma were investigated through in vivo, ex vivo, and in vitro experiments. KEY RESULTS CalD reduced total bronchoalveolar lavage fluid and differential cell count, serum IgE levels, mucus hypersecretion, and airway hyperresponsiveness in HDM-challenged mice. Additionally, CalD affected a wide array of pro-inflammatory cytokines and chemokines and oxidative damage markers in isolated lung tissues. CalD suppressed the HDM-induced increase in Arg1 (M2 macrophage marker) in AMs from lung tissue and reduced lung polyamine levels. CalD weakened antigen presentation capability of AMs by reducing CD80 expression, reduced AM-derived CCL17 and CCL22 levels, and lessened Th2 cytokines from CD4+ T-cells from asthma lung digest. CalD blocked the HDM-induced FoxO1/IRF4 pathway and restored impaired the Nrf2/HO-1 antioxidant pathway in lung tissues. CalD inhibited IL-4/IL-13-stimulated JAK1/STAT6 pathway, FoxO1 protein expression, and chemokine production in primary AMs. Structure-activity relationship study revealed the α,β-unsaturated γ-butyrolactone in CalD is capable of forming covalent bonds with cellular protein targets essential for its action. CONCLUSION AND IMPLICATIONS Our results demonstrate for the first time that CalD is a novel anti-inflammatory natural compound for allergic asthma that modulates AM function.
Collapse
Affiliation(s)
- Wupeng Liao
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore.,Singapore-HUJ Alliance for Research and Enterprise, National University of Singapore, Singapore
| | - Hazel Yu Ci Foo
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore.,Singapore-HUJ Alliance for Research and Enterprise, National University of Singapore, Singapore
| | - Thi Ngoc Quy Tran
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore.,Singapore-HUJ Alliance for Research and Enterprise, National University of Singapore, Singapore.,Drug Discovery and Optimization Platform, Yong Loo Lin School of Medicine, National University Health System, Singapore
| | - Christina Li Lin Chai
- Drug Discovery and Optimization Platform, Yong Loo Lin School of Medicine, National University Health System, Singapore.,Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore
| | - Wai Shiu Fred Wong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore.,Singapore-HUJ Alliance for Research and Enterprise, National University of Singapore, Singapore.,Drug Discovery and Optimization Platform, Yong Loo Lin School of Medicine, National University Health System, Singapore
| |
Collapse
|
142
|
Liu P, Ye L, Ren Y, Zhao G, Zhang Y, Lu S, Li Q, Wu C, Bai L, Zhang Z, Zhao Z, Shi Z, Yin S, Liao M, Lan Z, Feng J, Chen L. Chemotherapy-induced phlebitis via the GBP5/NLRP3 inflammasome axis and the therapeutic effect of aescin. Br J Pharmacol 2023; 180:1132-1147. [PMID: 36479683 DOI: 10.1111/bph.16002] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 10/03/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND PURPOSE Intravenous infusion of chemotherapy drugs can cause severe chemotherapy-induced phlebitis (CIP) in patients. However, the underlying mechanism of CIP development remains unclear. EXPERIMENTAL APPROACH RNA-sequencing analysis was used to identify potential disease targets in CIP. Guanylate binding protein-5 (GBP5) genetic deletion approaches also were used to investigate the role of GBP5 in NLR family pyrin domain containing 3 (NLRP3) inflammasome activation in lipopolysaccharide (LPS) primed murine bone-marrow-derived macrophages (BMDMs) induced by vinorelbine (VIN) in vitro and in mouse models of VIN-induced CIP in vivo. The anti-CIP effect of aescin was evaluated, both in vivo and in vivo. KEY RESULTS Here, we show that the expression of GBP5 was upregulated in human peripheral blood mononuclear cells from CIP patients. Genetic ablation of GBP5 in murine macrophages significantly alleviated VIN-induced CIP in the experimental mouse model. Mechanistically, GBP5 contributed to the inflammatory responses through activating NLRP3 inflammasome and driving the production of the inflammatory cytokine IL-1β. Moreover, aescin, a mixture of triterpene saponins extracted from horse chestnut seed, can alleviate CIP by inhibiting the GBP5/NLRP3 axis. CONCLUSION AND IMPLICATIONS These findings suggest that GBP5 is an important regulator of NLRP3 inflammasome in CIP mouse model. Our work further reveals that aescin may serve as a promising candidate in the clinical treatment of CIP.
Collapse
Affiliation(s)
- Peng Liu
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, China
| | - Lichun Ye
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Yongshen Ren
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, China
| | - Guodun Zhao
- Center for Neurological and Psychiatric Research and Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yun Zhang
- Center for Neurological and Psychiatric Research and Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shaojuan Lu
- School of Medicine, Tongji University, Shanghai, China
| | - Qiang Li
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, China
| | - Chen Wu
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, China
| | - Lijie Bai
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, China
| | - Zhongyun Zhang
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, China
| | - Zhongqiu Zhao
- Center for the Study of Itch, Department of Anesthesiology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA.,Barnes-Jewish Hospital, St. Louis, Missouri, USA
| | - Zhaohua Shi
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Shijin Yin
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, China
| | - Maochuan Liao
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, China
| | - Zhou Lan
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Jing Feng
- Center for Neurological and Psychiatric Research and Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lvyi Chen
- School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, China
| |
Collapse
|
143
|
Lawson R, Čechová P, Zarrouk E, Javellaud J, Bazgier V, Otyepka M, Trouillas P, Picard N, Marquet P, Saint-Marcoux F, El Balkhi S. Metabolic interactions of benzodiazepines with oxycodone ex vivo and toxicity depending on usage patterns in an animal model. Br J Pharmacol 2023; 180:829-842. [PMID: 34855983 DOI: 10.1111/bph.15765] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 09/16/2021] [Accepted: 11/03/2021] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE Opioids and benzodiazepines are frequently combined in medical as well as in non-medical contexts. At high doses, such combinations often result in serious health complications attributed to pharmacodynamics interactions. Here, we investigate the contribution of the metabolic interactions between oxycodone, diazepam and diclazepam (a designer benzodiazepine) in abuse/overdose conditions through ex vivo, in vivo and in silico approaches. EXPERIMENTAL APPROACH A preparation of pooled human liver microsomes was used to study oxycodone metabolism in the presence or absence of diazepam or diclazepam. In mice, diazepam or diclazepam was concomitantly administered with oxycodone to mimic acute intoxication. Diclazepam was introduced on Day 10 in mice continuously infused with oxycodone for 15 days to mimic chronic intoxication. In silico modelling was used to study the molecular interactions of the three drugs with CYP3A4 and 2D6. KEY RESULTS In mice, in acute conditions, both diazepam and diclazepam inhibited the metabolism of oxycodone. In chronic conditions and at pharmacologically equivalent doses, diclazepam drastically enhanced the production of oxymorphone. In silico, the affinity of benzodiazepines was higher than oxycodone for CYP3A4, inhibiting oxycodone metabolism through CYP3A4. Oxycodone metabolism is likely to be diverted towards CYP2D6. CONCLUSION AND IMPLICATIONS Acute doses of diazepam or diclazepam result in the accumulation of oxycodone, whereas chronic administration induces the accumulation of oxymorphone, the toxic metabolite. This suggests that overdoses of opioids in the presence of benzodiazepines are partly due to metabolic interactions, which in turn explain the patterns of toxicity dependent on usage. LINKED ARTICLES This article is part of a themed issue on Advances in Opioid Pharmacology at the Time of the Opioid Epidemic. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v180.7/issuetoc.
Collapse
Affiliation(s)
- Roland Lawson
- University of Limoges, IPPRITT, Limoges, France.,INSERM, IPPRITT, U1248, Limoges, France
| | - Petra Čechová
- Regional Center of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute (CATRIN), Palacký University Olomouc, Olomouc, Czechia
| | - Eliès Zarrouk
- University of Limoges, IPPRITT, Limoges, France.,INSERM, IPPRITT, U1248, Limoges, France
| | - James Javellaud
- University of Limoges, IPPRITT, Limoges, France.,INSERM, IPPRITT, U1248, Limoges, France
| | - Václav Bazgier
- Department of Physical Chemistry, Faculty of Science, Palacký University Olomouc, Olomouc, Czechia
| | - Michal Otyepka
- Regional Center of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute (CATRIN), Palacký University Olomouc, Olomouc, Czechia
| | - Patrick Trouillas
- University of Limoges, IPPRITT, Limoges, France.,INSERM, IPPRITT, U1248, Limoges, France.,Regional Center of Advanced Technologies and Materials, Czech Advanced Technology and Research Institute (CATRIN), Palacký University Olomouc, Olomouc, Czechia
| | - Nicolas Picard
- University of Limoges, IPPRITT, Limoges, France.,INSERM, IPPRITT, U1248, Limoges, France.,Department of Pharmacology, Toxicology and Pharmacovigilance, CHU Limoges, Limoges, France
| | - Pierre Marquet
- University of Limoges, IPPRITT, Limoges, France.,INSERM, IPPRITT, U1248, Limoges, France.,Department of Pharmacology, Toxicology and Pharmacovigilance, CHU Limoges, Limoges, France
| | - Franck Saint-Marcoux
- University of Limoges, IPPRITT, Limoges, France.,INSERM, IPPRITT, U1248, Limoges, France.,Department of Pharmacology, Toxicology and Pharmacovigilance, CHU Limoges, Limoges, France
| | - Souleiman El Balkhi
- INSERM, IPPRITT, U1248, Limoges, France.,Department of Pharmacology, Toxicology and Pharmacovigilance, CHU Limoges, Limoges, France
| |
Collapse
|
144
|
Gabel F, Hovhannisyan V, Andry V, Goumon Y. Central metabolism as a potential origin of sex differences in morphine antinociception but not induction of antinociceptive tolerance in mice. Br J Pharmacol 2023; 180:843-861. [PMID: 34986502 DOI: 10.1111/bph.15792] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 12/07/2021] [Accepted: 12/28/2021] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND AND PURPOSE In rodents, morphine antinociception is influenced by sex. However, conflicting results have been reported regarding the interaction between sex and morphine antinociceptive tolerance. Morphine is metabolised in the liver and brain into morphine-3-glucuronide (M3G). Sex differences in morphine metabolism and differential metabolic adaptations during tolerance development might contribute to behavioural discrepancies. This article investigates the differences in peripheral and central morphine metabolism after acute and chronic morphine treatment in male and female mice. EXPERIMENTAL APPROACH Sex differences in morphine antinociception and tolerance were assessed using the tail-immersion test. After acute and chronic morphine treatment, morphine and M3G metabolic kinetics in the blood were evaluated using LC-MS/MS. They were also quantified in several CNS regions. Finally, the blood-brain barrier (BBB) permeability of M3G was assessed in male and female mice. KEY RESULTS This study demonstrated that female mice showed weaker morphine antinociception and faster induction of tolerance than males. Additionally, female mice showed higher levels of M3G in the blood and in several pain-related CNS regions than male mice, whereas lower levels of morphine were observed in these regions. M3G brain/blood ratios after injection of M3G indicated no sex differences in M3G BBB permeability, and these ratios were lower than those obtained after injection of morphine. CONCLUSION These differences are attributable mainly to morphine central metabolism, which differed between males and females in pain-related CNS regions, consistent with weaker morphine antinociceptive effects in females. However, the role of morphine metabolism in antinociceptive tolerance seemed limited. LINKED ARTICLES This article is part of a themed issue on Advances in Opioid Pharmacology at the Time of the Opioid Epidemic. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v180.7/issuetoc.
Collapse
Affiliation(s)
- Florian Gabel
- CNRS UPR3212, Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique and University of Strasbourg, Strasbourg, France
| | - Volodya Hovhannisyan
- CNRS UPR3212, Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique and University of Strasbourg, Strasbourg, France
| | - Virginie Andry
- CNRS UPR3212, Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique and University of Strasbourg, Strasbourg, France.,SMPMS-INCI, Mass Spectrometry Facilities of the CNRS UPR3212, CNRS UPR3212, Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique and University of Strasbourg, Strasbourg, France
| | - Yannick Goumon
- CNRS UPR3212, Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique and University of Strasbourg, Strasbourg, France.,SMPMS-INCI, Mass Spectrometry Facilities of the CNRS UPR3212, CNRS UPR3212, Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique and University of Strasbourg, Strasbourg, France
| |
Collapse
|
145
|
Zhu W, Wang M, Jin L, Yang B, Bai B, Mutsinze RN, Zuo W, Chattipakorn N, Huh JY, Liang G, Wang Y. Licochalcone A protects against LPS-induced inflammation and acute lung injury by directly binding with myeloid differentiation factor 2 (MD2). Br J Pharmacol 2023; 180:1114-1131. [PMID: 36480410 DOI: 10.1111/bph.15999] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 09/07/2022] [Accepted: 09/12/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND PURPOSE Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is a challenging clinical syndrome that leads to various respiratory sequelae and even high mortality in patients with severe disease. The novel pharmacological strategies and therapeutic drugs are urgently needed. Natural products have played a fundamental role and provided an abundant pool in drug discovery. EXPERIMENTAL APPROACH A compound library containing 160 natural products was used to screen potential anti-inflammatory compounds. Mice with LPS-induced ALI was then used to verify the preventive and therapeutic effects of the selected compounds. KEY RESULTS Licochalcone A was discovered from the anti-inflammatory screening of natural products in macrophages. A qPCR array validated the inflammation-regulatory effects of licochalcone A and indicated that the potential targets of licochalcone A may be the upstream proteins in LPS pro-inflammatory signalling. Further studies showed that licochalcone A directly binds to myeloid differentiation factor 2 (MD2), an assistant protein of toll-like receptor 4 (TLR4), to block both LPS-induced TRIF- and MYD88-dependent pathways. LEU61 and PHE151 in MD2 protein are the two key residues that contribute to the binding of MD2 to licochalcone A. In vivo, licochalcone A treatment alleviated ALI in LPS-challenged mice through significantly reducing immunocyte infiltration, suppressing activation of TLR4 pathway and inflammatory cytokine induction. CONCLUSION AND IMPLICATIONS In summary, our study identified MD2 as a direct target of licochalcone A for its anti-inflammatory activity and suggested that licochalcone A might serve as a novel MD2 inhibitor and a potential drug for developing ALI/ARDS therapy.
Collapse
Affiliation(s)
- Weiwei Zhu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China.,College of Pharmacy, Chonnam National University, Gwangju, Korea
| | - Minxiu Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Leiming Jin
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Bin Yang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Bin Bai
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Rumbidzai Natasha Mutsinze
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Wei Zuo
- Affiliated Xiangshan Hospital of Wenzhou Medical University (Xiangshan First People's Hospital Medical and Health Group), Xiangshan, China
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Joo Young Huh
- College of Pharmacy, Chonnam National University, Gwangju, Korea
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China.,School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, China
| | - Yi Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China.,College of Pharmacy, Chonnam National University, Gwangju, Korea.,Affiliated Xiangshan Hospital of Wenzhou Medical University (Xiangshan First People's Hospital Medical and Health Group), Xiangshan, China
| |
Collapse
|
146
|
Ding H, Jiang M, Lau CW, Luo J, Chan AM, Wang L, Huang Y. Curaxin CBL0137 inhibits endothelial inflammation and atherogenesis via suppression of the Src-YAP signalling axis. Br J Pharmacol 2023; 180:1168-1185. [PMID: 36495259 DOI: 10.1111/bph.16007] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 10/10/2022] [Accepted: 12/01/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND AND PURPOSE Atherosclerotic vascular disease is the leading cause of mortality and morbidity worldwide. Our previous study uncovered that endothelium-specific knockdown of YAP suppresses atherogenesis, suggesting that YAP is a promising therapeutic target against atherosclerotic vascular disease. We established a drug screening platform, which aimed to identify new YAP inhibitors for anti-atherosclerotic treatment. EXPERIMENTAL APPROACH Drug screening was performed by a luciferase reporter gene assay. RNA sequencing was performed to acquire the transcriptomic profile of CBL0137-treated endothelial cells. We assessed and validated the inhibitory effect of CBL0137 on YAP activity and inflammatory response in HUVECs and HAECs. We evaluated the vasoprotective effect of CBL0137 in vivo against plaque formation in ApoE-/- mice, using both disturbed flow-induced and chronic western diet-induced atherosclerotic models. KEY RESULTS We identified CBL0137 as a novel YAP inhibitor from an FDA drug library. CBL0137 inhibited YAP activity by restraining its phosphorylation at Y357. CBL0137 inhibited YAP activity to repress endothelial inflammation. Mechanistically, CBL0137 suppressed YAP phosphorylation at Y357 via the tyrosine-protein kinase Src. Furthermore, administration of CBL0137 ameliorated endothelial inflammation and the atherogenesis induced by disturbed flow and consumption of an atherogenic diet in ApoE-/- mice. CONCLUSION AND IMPLICATIONS To our knowledge, this is the first study to identify CBL0137 as a novel YAP inhibitor. We have demonstrated that pharmacologically targeting YAP by CBL0137 inhibits atherogenesis. The present results suggest that CBL0137 holds promise as a new drug for the treatment of atherosclerotic vascular disease.
Collapse
Affiliation(s)
- Huanyu Ding
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Minchun Jiang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Chi Wai Lau
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Jianfang Luo
- Department of Cardiology, Guangdong Provincial People's Hospital, Guangdong Cardiovascular Institute, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Andrew M Chan
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Li Wang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Yu Huang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| |
Collapse
|
147
|
Imig JD, Khan MAH, Stavniichuk A, Jankiewicz WK, Goorani S, Yeboah MM, El-Meanawy A. Salt-sensitive hypertension after reversal of unilateral ureteral obstruction. Biochem Pharmacol 2023; 210:115438. [PMID: 36716827 PMCID: PMC10107073 DOI: 10.1016/j.bcp.2023.115438] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 01/23/2023] [Accepted: 01/24/2023] [Indexed: 01/28/2023]
Abstract
The incidence of ureter obstruction is increasing and patients recovering from this kidney injury often progress to chronic kidney injury. There is evidence that a long-term consequence of recovery from ureter obstruction is an increased risk for salt-sensitive hypertension. A reversal unilateral ureteral obstruction (RUUO) model was used to study long-term kidney injury and salt-sensitive hypertension. In this model, we removed the ureteral obstruction at day 10 in mice. Mice were divided into four groups: (1) normal salt diet, (2) high salt diet, (3) RUUO normal salt diet, and (4) RUUO high salt diet. At day 10, the mice were fed a normal or high salt diet for 4 weeks. Blood pressure was measured, and urine and kidney tissue collected. There was a progressive increase in blood pressure in the RUUO high salt diet group. RUUO high salt group had decreased sodium excretion and glomerular injury. Renal epithelial cell injury was evident in RUUO normal and high salt mice as assessed by neutrophil gelatinase-associated lipocalin (NGAL). Kidney inflammation in the RUUO high salt group involved an increase in F4/80 positive macrophages; however, CD3+ positive T cells were not changed. Importantly, RUUO normal and high salt mice had decreased vascular density. RUUO was also associated with renal fibrosis that was further elevated in RUUO mice fed a high salt diet. Overall, these findings demonstrate long-term renal tubular injury, inflammation, decreased vascular density, and renal fibrosis following reversal of unilateral ureter obstruction that could contribute to impaired sodium excretion and salt-sensitive hypertension.
Collapse
Affiliation(s)
- John D Imig
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Drug Discovery Center, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA.
| | - Md Abdul Hye Khan
- Drug Discovery Center, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Anna Stavniichuk
- Drug Discovery Center, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA; Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Wojciech K Jankiewicz
- Drug Discovery Center, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Samaneh Goorani
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Drug Discovery Center, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Michael M Yeboah
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Ashraf El-Meanawy
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
148
|
Pisanò CA, Mercatelli D, Mazzocchi M, Brugnoli A, Morella I, Fasano S, Zaveri NT, Brambilla R, O'Keeffe GW, Neubig RR, Morari M. Regulator of G-Protein Signalling 4 (RGS4) negatively modulates nociceptin/orphanin FQ opioid receptor signalling: Implication for l-Dopa-induced dyskinesia. Br J Pharmacol 2023; 180:927-942. [PMID: 34767639 DOI: 10.1111/bph.15730] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 10/11/2021] [Accepted: 10/18/2021] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Regulator of G-protein signalling 4 (RGS4) is a signal transduction protein that accelerates intrinsic GTPase activity of Gαi/o and Gαq subunits, suppressing GPCR signalling. Here, we investigate whether RGS4 modulates nociceptin/orphanin FQ (N/OFQ) opioid (NOP) receptor signalling and if this modulation has relevance for l-Dopa-induced dyskinesia. EXPERIMENTAL APPROACH HEK293T cells transfected with NOP, NOP/RGS4 or NOP/RGS19 were challenged with N/OFQ and the small-molecule NOP agonist AT-403, using D1-stimulated cAMP levels as a readout. Primary rat striatal neurons and adult mouse striatal slices were challenged with either N/OFQ or AT-403 in the presence of the experimental RGS4 chemical probe, CCG-203920, and D1-stimulated cAMP or phosphorylated extracellular signal regulated kinase 1/2 (pERK) responses were monitored. In vivo, CCG-203920 was co-administered with AT-403 and l-Dopa to 6-hydroxydopamine hemilesioned rats, and dyskinetic movements, striatal biochemical correlates of dyskinesia (pERK and pGluR1 levels) and striatal RGS4 levels were measured. KEY RESULTS RGS4 expression reduced NOFQ and AT-403 potency and efficacy in HEK293T cells. CCG-203920 increased N/OFQ potency in primary rat striatal neurons and potentiated AT-403 response in mouse striatal slices. CCG-203920 enhanced AT-403-mediated inhibition of dyskinesia and its biochemical correlates, without compromising its motor-improving effects. Unilateral dopamine depletion caused bilateral reduction of RGS4 levels, which was reversed by l-Dopa. l-Dopa acutely up-regulated RGS4 in the lesioned striatum. CONCLUSIONS AND IMPLICATIONS RGS4 physiologically inhibits NOP receptor signalling. CCG-203920 enhanced NOP responses and improved the antidyskinetic potential of NOP receptor agonists, mitigating the effects of striatal RGS4 up-regulation occurring during dyskinesia expression. LINKED ARTICLES This article is part of a themed issue on Advances in Opioid Pharmacology at the Time of the Opioid Epidemic. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v180.7/issuetoc.
Collapse
Affiliation(s)
- Clarissa A Pisanò
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Daniela Mercatelli
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Martina Mazzocchi
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Alberto Brugnoli
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Ilaria Morella
- Neuroscience and Mental Health Research Institute, Division of Neuroscience, School of Biosciences, Cardiff University, Cardiff, UK
| | - Stefania Fasano
- Neuroscience and Mental Health Research Institute, Division of Neuroscience, School of Biosciences, Cardiff University, Cardiff, UK
| | - Nurulain T Zaveri
- Astraea Therapeutics, Medicinal Chemistry Division, Mountain View, California, USA
| | - Riccardo Brambilla
- Neuroscience and Mental Health Research Institute, Division of Neuroscience, School of Biosciences, Cardiff University, Cardiff, UK
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| | - Gerard W O'Keeffe
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Richard R Neubig
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, USA
| | - Michele Morari
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| |
Collapse
|
149
|
Zhao J, Bai D, Qi L, Cao W, Du J, Gu C, Zhou C, Gao Y, Zhang L, Zhao Y, Lu N. The flavonoid GL-V9 alleviates liver fibrosis by triggering senescence by regulating the transcription factor GATA4 in activated hepatic stellate cells. Br J Pharmacol 2023; 180:1072-1089. [PMID: 36455594 DOI: 10.1111/bph.15997] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 11/14/2022] [Accepted: 11/23/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND AND PURPOSE Liver fibrosis is a critical risk factor for the progression from chronic liver injury to hepatocellular carcinoma. Clinically, there is a lack of therapeutic drugs for liver fibrosis. Previous studies have confirmed that GL-V9, a newly synthesized flavonoid derivative, exhibits anti-inflammatory activity, but whether it has anti-hepatic fibrosis actions remains unclear. This study aimed to investigate the anti-fibrotic activities and potential mechanisms of GL-V9. EXPERIMENTAL APPROACH Bile duct ligation (BDL) and carbon tetrachloride (CCl4 ) challenges were used to assess the anti-fibrotic effects of GL-V9 in vivo. Mouse primary hepatic stellate cells (pHSCs) and the human HSC line LX2 also served as a liver fibrosis model in vitro. Cellular functions and molecular mechanism were analysed using senescence-associated beta-galactosidase staining, real-time PCR, western blotting, immunofluorescence, and co-immunoprecipitation. KEY RESULTS GL-V9 attenuated hepatic histopathological injury and collagen accumulation, as well as decreasing the expression of fibrotic genes in vivo. GL-V9 promoted senescence and inhibited the expression of fibrogenic genes in HSCs in vitro. Mechanistic studies revealed that GL-V9 induced senescence by upregulating GATA4 expression in HSCs. Further studies confirmed that GL-V9 stabilized GATA4 by promoting autophagic degradation of P62. CONCLUSION AND IMPLICATIONS GL-V9 exerted potent anti-fibrotic effects both in vivo and in vitro by stabilizing GATA4, thereby promoting the senescence of HSCs, and by avoiding its activation and ultimately inhibiting liver fibrosis. This action indicated that the flavonoid GL-V9 is a potential therapeutic candidate for the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Jiawei Zhao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, China
| | - Dongsheng Bai
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, China
| | - Lei Qi
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, China
| | - Wangjia Cao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, China
| | - Jiaying Du
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, China
| | - Chunyang Gu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, China
| | - Chen Zhou
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, China
| | - Yuan Gao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, China
| | - Lulu Zhang
- Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Yue Zhao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, China
| | - Na Lu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, China
| |
Collapse
|
150
|
López-Cano M, Font J, Aso E, Sahlholm K, Cabré G, Giraldo J, De Koninck Y, Hernando J, Llebaria A, Fernández-Dueñas V, Ciruela F. Remote local photoactivation of morphine produces analgesia without opioid-related adverse effects. Br J Pharmacol 2023; 180:958-974. [PMID: 34363210 DOI: 10.1111/bph.15645] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 07/19/2021] [Accepted: 07/23/2021] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Opioid-based drugs are the gold standard medicines for pain relief. However, tolerance and several side effects (i.e. constipation and dependence) may occur upon chronic opioid administration. Photopharmacology is a promising approach to improve the benefit/risk profiles of these drugs. Thus, opioids can be locally activated with high spatiotemporal resolution, potentially minimizing systemic-mediated adverse effects. Here, we aimed at developing a morphine photo-derivative (photocaged morphine), which can be activated upon light irradiation both in vitro and in vivo. EXPERIMENTAL APPROACH Light-dependent activity of pc-morphine was assessed in cell-based assays (intracellular calcium accumulation and electrophysiology) and in mice (formalin animal model of pain). In addition, tolerance, constipation and dependence were investigated in vivo using experimental paradigms. KEY RESULTS In mice, pc-morphine was able to elicit antinociceptive effects, both using external light-irradiation (hind paw) and spinal cord implanted fibre-optics. In addition, remote morphine photoactivation was devoid of common systemic opioid-related undesired effects, namely, constipation, tolerance to the analgesic effects, rewarding effects and naloxone-induced withdrawal. CONCLUSION AND IMPLICATIONS Light-dependent opioid-based drugs may allow effective analgesia without the occurrence of tolerance or the associated and severe opioid-related undesired effects. LINKED ARTICLES This article is part of a themed issue on Advances in Opioid Pharmacology at the Time of the Opioid Epidemic. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v180.7/issuetoc.
Collapse
Affiliation(s)
- Marc López-Cano
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain.,Neuropharmacology & Pain Group, Neuroscience Program, Bellvitge Institute for Biomedical Research, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Joan Font
- MCS, Laboratory of Medicinal Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona, Spain.,Institut de Génomique Fonctionnelle (IGF), University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Ester Aso
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain.,Neuropharmacology & Pain Group, Neuroscience Program, Bellvitge Institute for Biomedical Research, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Kristoffer Sahlholm
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain.,Neuropharmacology & Pain Group, Neuroscience Program, Bellvitge Institute for Biomedical Research, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain.,Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.,Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | - Gisela Cabré
- Departament de Química, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Jesús Giraldo
- Laboratory of Molecular Neuropharmacology and Bioinformatics, Unitat de Bioestadística and Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Spain.,Unitat de Neurociència Traslacional, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí (I3PT), Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain.,Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM, Barcelona, Spain
| | - Yves De Koninck
- Institut Universitaire en Santé Mentale de Québec, Québec, Quebec, Canada.,Department of Psychiatry and Neuroscience, Université Laval, Québec, Quebec, Canada
| | - Jordi Hernando
- Departament de Química, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Amadeu Llebaria
- MCS, Laboratory of Medicinal Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona, Spain
| | - Víctor Fernández-Dueñas
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain.,Neuropharmacology & Pain Group, Neuroscience Program, Bellvitge Institute for Biomedical Research, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Francisco Ciruela
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain.,Neuropharmacology & Pain Group, Neuroscience Program, Bellvitge Institute for Biomedical Research, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| |
Collapse
|