1
|
Li ZZ, Wang Z, Chen X, Feng HQ, Yao XY, Song J, Xu B, Jin J, Cao X, Zhuang T. Benzylpiperidine derivatives as new dual μ-opioid and σ 1 receptor ligands with potent antinociceptive effects. Bioorg Chem 2024; 153:107921. [PMID: 39492131 DOI: 10.1016/j.bioorg.2024.107921] [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: 09/03/2024] [Revised: 10/20/2024] [Accepted: 10/24/2024] [Indexed: 11/05/2024]
Abstract
Dual-acting μ-opioid receptor (MOR)/sigma-1 receptor (σ1R) ligands have displayed promise in exerting robust antinociceptive effects while reducing opioid-related side effects. To discover safer and more effective analgesics, we designed, prepared, and evaluated 30 benzylpiperidine derivatives as dual MOR and σ1R ligands. The obtained benzylpiperidine analogs were tested for MOR and σ1R binding affinity in vitro. The best compound 52 showed high affinity for both MOR [Ki (MOR) = 56.4 nM] and σ1R [Ki (σ1R) = 11.0 nM] and produced potent antinociceptive effects in the abdominal contraction test (ED50 = 4.04 mg/kg in mice), carrageenan-induced inflammatory pain model (ED50 = 6.88 mg/kg in mice), formalin test (ED50 = 13.98 mg/kg in rats) and complete Freund's adjuvant (CFA)-induced chronic pain model (ED50 = 7.62 mg/kg in mice). Moreover, 52 had less MOR-related adverse effects than oxycodone, including constipation, acute hyperlocomotion and physical dependence. The above results suggested that 52 may be a promising candidate for the development of safer analgesics.
Collapse
Affiliation(s)
- Zong-Zheng Li
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, School of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Zhen Wang
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, School of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Xiong Chen
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, School of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Hong-Qing Feng
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, School of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Xing-Yu Yao
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, School of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Jie Song
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, School of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Ben Xu
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, School of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Jian Jin
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, School of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China.
| | - Xudong Cao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004, Jiangsu, China.
| | - Tao Zhuang
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, School of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China.
| |
Collapse
|
2
|
Wei H, Vuorenpää A, Laurila J, Domanskyi A, Koivisto A, Pertovaara A. Indirect involvement of α 2-adrenoceptors in the mechanical antihypersensitivity effect induced by the spinally administered imidazoline I 1 receptor ligand LNP599 in a rat model of experimental neuropathy. Brain Res Bull 2024; 217:111089. [PMID: 39341510 DOI: 10.1016/j.brainresbull.2024.111089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 08/28/2024] [Accepted: 09/25/2024] [Indexed: 10/01/2024]
Abstract
Here we assess whether neuropathic pain hypersensitivity is attenuated by spinal administration of the imidazoline I1-receptor agonist LNP599 and whether the attenuation involves co-activation of α2-adrenoceptors. Spared nerve injury (SNI) model of neuropathy was used to induce mechanical hypersensitivity in male and female rats with a chronic catheter for intrathecal drug administrations. Mechanical sensitivity and heat nociception were assessed behaviorally in the injured limb. Additionally, GTPγS radioligand binding assay, β-arrestin recruitment and intracellular cAMP levels were used for receptor profiling in vitro. LNP599 (imidazoline I1 receptor agonist) and clonidine (α2-adrenoceptor agonist) produced equal dose-related mechanical antihypersensitivity effects in both sexes. LNP599 attenuated heat nociception preferentially in males, while clonidine reduced heat nociception equally in males and females. Carbophenyline (another imidazoline I1 receptor agonist) had no significant effect on mechanical hypersensitivity or heat nociception in males or females. Mechanical antihypersensitivity and heat antinociception induced by LNP599 in SNI males was prevented by pretreatments with yohimbine or atipamezole (two α2-adrenoceptor antagonists) but not by efaroxan (a mixed imidazoline I1 receptor/α2-adrenoceptor antagonist). In vitro assays indicated that LNP599 does not activate α2A- or other subtypes of α2-adrenoceptors. However, LNP599 was a weak partial agonist for 5-HT2B receptors and bound to sigma-1 and sigma-2 receptors that all are involved in modulation of spinal nociception. The results indicate that the suppression of neuropathic pain hypersensitivity by LNP599 is not due to action on spinal imidazoline I1 receptors, but rather due to indirect activation of spinal α2-adrenoceptors.
Collapse
Affiliation(s)
- Hong Wei
- Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Anne Vuorenpää
- Pain Research Unit, Orion Pharma, Orion Corporation, Turku, Finland
| | - Jonne Laurila
- Faculty of Medicine, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Andrii Domanskyi
- Pain Research Unit, Orion Pharma, Orion Corporation, Turku, Finland
| | - Ari Koivisto
- Pain Research Unit, Orion Pharma, Orion Corporation, Turku, Finland.
| | - Antti Pertovaara
- Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
3
|
Kamei T, Kudo T, Yamane H, Ishibashi F, Takada Y, Honda S, Maezawa Y, Ikeda K, Oyamada Y. Unique electrophysiological property of a novel Nav1.7, Nav1.8, and Nav1.9 sodium channel blocker, ANP-230. Biochem Biophys Res Commun 2024; 721:150126. [PMID: 38776832 DOI: 10.1016/j.bbrc.2024.150126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 04/28/2024] [Accepted: 05/13/2024] [Indexed: 05/25/2024]
Abstract
Voltage-gated sodium channel subtypes, Nav1.7, Nav1.8, and Nav1.9 are predominantly expressed in peripheral sensory neurons. Recent genetic studies have revealed that they are involved in pathological pain processing and that the blockade of Nav1.7, Nav1.8, or Nav1.9 will become a promising pharmacotherapy especially for neuropathic pain. A growing number of drug discovery programs have targeted either of the subtypes to obtain a selective inhibitor which can provide pain relief without affecting the cardiovascular and central nervous systems, though none of them has been approved yet. Here we describe the in vitro characteristics of ANP-230, a novel sodium channel blocker under clinical development. Surprisingly, ANP-230 was shown to block three pain-related subtypes, human Nav1.7, Nav1.8, and Nav1.9 with similar potency, but had only low inhibitory activity to human cardiac Nav1.5 channel and rat central Nav channels. The voltage clamp experiments using different step pulse protocols revealed that ANP-230 had a "tonic block" mode of action without state- and use-dependency. In addition, ANP-230 caused a depolarizing shift of the activation curve and decelerated gating kinetics in human Nav1.7-stably expressing cells. The depolarizing shift of activation curve was commonly observed in human Nav1.8-stably expressing cells as well as rat dorsal root ganglion neurons. These data suggested a quite unique mechanism of Nav channel inhibition by ANP-230. Finally, ANP-230 reduced excitability of rat dorsal root ganglion neurons in a concentration dependent manner. Collectively, these promising results indicate that ANP-230 could be a potent drug for neuropathic pain.
Collapse
Affiliation(s)
- Tatsuya Kamei
- Pharmacology Research Unit, Research Division, Sumitomo Pharma Co., Ltd., Osaka, 554-0022, Japan; Regenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Kobe, 650-0047, Japan.
| | - Takehiro Kudo
- Pharmacology Research Unit, Research Division, Sumitomo Pharma Co., Ltd., Osaka, 554-0022, Japan
| | - Hana Yamane
- Pharmacology Research Unit, Research Division, Sumitomo Pharma Co., Ltd., Osaka, 554-0022, Japan; Regenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Kobe, 650-0047, Japan
| | - Fumiaki Ishibashi
- Pharmacology Research Unit, Research Division, Sumitomo Pharma Co., Ltd., Osaka, 554-0022, Japan; Platform Technology Research Unit, Research Division, Sumitomo Pharma Co., Ltd., Osaka, 554-0022, Japan
| | - Yoshinori Takada
- Pharmacology Research Unit, Research Division, Sumitomo Pharma Co., Ltd., Osaka, 554-0022, Japan; Global Corporate Strategy, Sumitomo Pharma Co., Ltd., Tokyo, 104-8356, Japan
| | - Shigeyuki Honda
- Pharmacology Research Unit, Research Division, Sumitomo Pharma Co., Ltd., Osaka, 554-0022, Japan; Sumika Chemical Analysis Service, Ltd., Osaka, 554-0022, Japan
| | - Yasuyo Maezawa
- Pharmacology Research Unit, Research Division, Sumitomo Pharma Co., Ltd., Osaka, 554-0022, Japan
| | - Kazuhito Ikeda
- Pharmacology Research Unit, Research Division, Sumitomo Pharma Co., Ltd., Osaka, 554-0022, Japan; Platform Technology Research Unit, Research Division, Sumitomo Pharma Co., Ltd., Osaka, 554-0022, Japan
| | - Yoshihiro Oyamada
- Pharmacology Research Unit, Research Division, Sumitomo Pharma Co., Ltd., Osaka, 554-0022, Japan; AlphaNavi Pharma Inc., Osaka, 564-0053, Japan
| |
Collapse
|
4
|
Ruiz-Cantero MC, Huerta MÁ, Tejada MÁ, Santos-Caballero M, Fernández-Segura E, Cañizares FJ, Entrena JM, Baeyens JM, Cobos EJ. Sigma-1 receptor agonism exacerbates immune-driven nociception: Role of TRPV1 + nociceptors. Biomed Pharmacother 2023; 167:115534. [PMID: 37729726 DOI: 10.1016/j.biopha.2023.115534] [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: 06/21/2023] [Revised: 09/14/2023] [Accepted: 09/17/2023] [Indexed: 09/22/2023] Open
Abstract
The analgesic effects of sigma-1 antagonists are undisputed, but the effects of sigma-1 agonists on pain are not well studied. Here, we used a mouse model to show that the administration of the sigma-1 agonists dextromethorphan (a widely used antitussive drug), PRE-084 (a standard sigma-1 ligand), and pridopidine (a selective drug being investigated in clinical trials for the treatment of neurodegenerative diseases) enhances PGE2-induced mechanical hyperalgesia. Superficial plantar incision induced transient weight-bearing asymmetry at early time points, but the mice appeared to recover at 24 h, despite noticeable edema and infiltration of neutrophils (a well-known cellular source of PGE2) at the injured site. Sigma-1 agonists induced a relapse of weight bearing asymmetry in a manner dependent on the presence of neutrophils. The effects of sigma-1 agonists were all reversed by administration of the sigma-1 antagonist BD-1063 in wild-type mice, and were absent in sigma-1 knockout mice, supporting the selectivity of the effects observed. The proalgesic effects of sigma-1 agonism were also abolished by the TRP antagonist ruthenium red and by in vivo resiniferatoxin ablation of TRPV1 + peripheral sensory neurons. Therefore, sigma-1 agonism exacerbates pain-like responses in mice with a mild inflammatory state through the action of TRPV1 + nociceptors. We also show that sigma-1 receptors are present in most (if not all) mouse and human DRG neurons. If our findings translate to humans, further studies will be needed to investigate potential proalgesic effects induced by sigma-1 agonism in patients treated with sigma-1 agonists.
Collapse
Affiliation(s)
- M Carmen Ruiz-Cantero
- Department of Pharmacology, Faculty of Medicine, University of Granada, 18016 Granada, Spain; Institute of Neuroscience, Biomedical Research Center, University of Granada, Armilla, 18100 Granada, Spain; Biosanitary Research Institute ibs.GRANADA, 18012 Granada, Spain
| | - Miguel Á Huerta
- Department of Pharmacology, Faculty of Medicine, University of Granada, 18016 Granada, Spain; Institute of Neuroscience, Biomedical Research Center, University of Granada, Armilla, 18100 Granada, Spain; Biosanitary Research Institute ibs.GRANADA, 18012 Granada, Spain
| | - Miguel Á Tejada
- Department of Pharmacology, Faculty of Medicine, University of Granada, 18016 Granada, Spain; Institute of Neuroscience, Biomedical Research Center, University of Granada, Armilla, 18100 Granada, Spain; Biosanitary Research Institute ibs.GRANADA, 18012 Granada, Spain
| | - Miriam Santos-Caballero
- Department of Pharmacology, Faculty of Medicine, University of Granada, 18016 Granada, Spain; Institute of Neuroscience, Biomedical Research Center, University of Granada, Armilla, 18100 Granada, Spain; Biosanitary Research Institute ibs.GRANADA, 18012 Granada, Spain
| | - Eduardo Fernández-Segura
- Institute of Neuroscience, Biomedical Research Center, University of Granada, Armilla, 18100 Granada, Spain; Biosanitary Research Institute ibs.GRANADA, 18012 Granada, Spain; Department of Histology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
| | - Francisco J Cañizares
- Institute of Neuroscience, Biomedical Research Center, University of Granada, Armilla, 18100 Granada, Spain; Biosanitary Research Institute ibs.GRANADA, 18012 Granada, Spain; Department of Histology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
| | - José M Entrena
- Department of Pharmacology, Faculty of Medicine, University of Granada, 18016 Granada, Spain; Institute of Neuroscience, Biomedical Research Center, University of Granada, Armilla, 18100 Granada, Spain; Biosanitary Research Institute ibs.GRANADA, 18012 Granada, Spain
| | - José M Baeyens
- Department of Pharmacology, Faculty of Medicine, University of Granada, 18016 Granada, Spain; Institute of Neuroscience, Biomedical Research Center, University of Granada, Armilla, 18100 Granada, Spain; Biosanitary Research Institute ibs.GRANADA, 18012 Granada, Spain
| | - Enrique J Cobos
- Department of Pharmacology, Faculty of Medicine, University of Granada, 18016 Granada, Spain; Institute of Neuroscience, Biomedical Research Center, University of Granada, Armilla, 18100 Granada, Spain; Biosanitary Research Institute ibs.GRANADA, 18012 Granada, Spain; Teófilo Hernando Institute for Drug Discovery, 28029 Madrid, Spain.
| |
Collapse
|
5
|
Mogilski S, Kubacka M, Świerczek A, Wyska E, Szczepańska K, Sapa J, Kieć-Kononowicz K, Łażewska D. Efficacy of the Multi-Target Compound E153 in Relieving Pain and Pruritus of Different Origins. Pharmaceuticals (Basel) 2023; 16:1481. [PMID: 37895952 PMCID: PMC10609854 DOI: 10.3390/ph16101481] [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: 08/18/2023] [Revised: 10/04/2023] [Accepted: 10/13/2023] [Indexed: 10/29/2023] Open
Abstract
Itch and pain are closely related but distinct sensations that share largely overlapping mediators and receptors. We hypothesized that the novel, multi-target compound E153 has the potential to attenuate pain and pruritus of different origins. After the evaluation of sigma receptor affinity and pharmacokinetic studies, we tested the compound using different procedures and models of pain and pruritus. Additionally, we used pharmacological tools, such as PRE-084, RAMH, JNJ 5207852, and S1RA, to precisely determine the role of histamine H3 and sigma 1 receptors in the analgesic and antipruritic effects of the compound. In vitro studies revealed that the test compound had potent affinity for sigma 1 and sigma 2 receptors, moderate affinity for opioid kappa receptors, and no affinity for delta or μ receptors. Pharmacokinetic studies showed that after intraperitoneal administration, the compound was present at high concentrations in both the peripheral tissues and the central nervous system. The blood-brain barrier-penetrating properties indicate its ability to act centrally at the levels of the brain and spinal cord. Furthermore, the test compound attenuated different types of pain, including acute, inflammatory, and neuropathic. It also showed a broad spectrum of antipruritic activity, attenuating histamine-dependent and histamine-independent itching. Finally, we proved that antagonism of both sigma 1 and histamine H3 receptors is involved in the analgesic activity of the compound, while the antipruritic effect to a greater extent depends on sigma 1 antagonism.
Collapse
Affiliation(s)
- Szczepan Mogilski
- Department of Pharmacodynamics, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland; (M.K.); (J.S.)
| | - Monika Kubacka
- Department of Pharmacodynamics, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland; (M.K.); (J.S.)
| | - Artur Świerczek
- Department of Pharmacokinetics and Physical Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland; (A.Ś.); (E.W.)
| | - Elżbieta Wyska
- Department of Pharmacokinetics and Physical Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland; (A.Ś.); (E.W.)
| | - Katarzyna Szczepańska
- Department of Technology and Biotechnology of Drugs, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland; (K.S.); (K.K.-K.); (D.Ł.)
- Department of Medicinal Chemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Kraków, Poland
| | - Jacek Sapa
- Department of Pharmacodynamics, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland; (M.K.); (J.S.)
| | - Katarzyna Kieć-Kononowicz
- Department of Technology and Biotechnology of Drugs, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland; (K.S.); (K.K.-K.); (D.Ł.)
| | - Dorota Łażewska
- Department of Technology and Biotechnology of Drugs, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland; (K.S.); (K.K.-K.); (D.Ł.)
| |
Collapse
|
6
|
Dickinson JE, Inzunza JAD, Perez-Villa L, Millar TG, Pushparaj AP. Case report: Ibogaine reduced severe neuropathic pain associated with a case of brachial plexus nerve root avulsion. FRONTIERS IN PAIN RESEARCH 2023; 4:1256396. [PMID: 37720911 PMCID: PMC10502345 DOI: 10.3389/fpain.2023.1256396] [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: 07/10/2023] [Accepted: 08/11/2023] [Indexed: 09/19/2023] Open
Abstract
Brachial plexus nerve root avulsion results from complete separation of the nerve root from the spinal cord and is one of the most challenging types of neuropathic pain, coinciding with motor, sensory and autonomic deficits. The severe pain and typical impossibility of root reattachment often leads to requests for amputation. Ibogaine is an indole alkaloid producing psychoactive effects through reported actions upon multiple neurotransmitter systems, including NMDA, κ- and µ-opioid receptors and σ2 receptor sites, along with stimulation of neurotrophic factors GDNF and BDNF. In this case report we describe a 53-year-old male with two decades of severe intractable pain due to brachial plexus nerve root avulsion from vehicular trauma who was successfully treated with both high dose inpatient and low dose outpatient administrations of ibogaine. Though promising for future study, the adverse effects of high dose ibogaine administrations may limit tolerability of this saturation protocol to the most refractory cases.
Collapse
Affiliation(s)
| | | | | | | | - Abhiram P. Pushparaj
- Scientific Advisory, Ambio Life Sciences, Vancouver, BC, Canada
- Consulting Department, +ROI Regulatory Advisory, Toronto, ON, Canada
| |
Collapse
|
7
|
Rossino G, Marra A, Listro R, Peviani M, Poggio E, Curti D, Pellavio G, Laforenza U, Dondio G, Schepmann D, Wünsch B, Bedeschi M, Marino N, Tesei A, Ha HJ, Kim YH, Ann J, Lee J, Linciano P, Di Giacomo M, Rossi D, Collina S. Discovery of RC-752, a Novel Sigma-1 Receptor Antagonist with Antinociceptive Activity: A Promising Tool for Fighting Neuropathic Pain. Pharmaceuticals (Basel) 2023; 16:962. [PMID: 37513874 PMCID: PMC10386076 DOI: 10.3390/ph16070962] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/30/2023] [Accepted: 07/02/2023] [Indexed: 07/30/2023] Open
Abstract
Neuropathic pain (NP) is a chronic condition resulting from damaged pain-signaling pathways. It is a debilitating disorder that affects up to 10% of the world's population. Although opioid analgesics are effective in reducing pain, they present severe risks; so, there is a pressing need for non-opioid pain-relieving drugs. One potential alternative is represented by sigma-1 receptor (S1R) antagonists due to their promising analgesic effects. Here, we report the synthesis and biological evaluation of a series of S1R antagonists based on a 2-aryl-4-aminobutanol scaffold. After assessing affinity toward the S1R and selectivity over the sigma-2 receptor (S2R), we evaluated the agonist/antagonist profile of the compounds by investigating their effects on nerve growth factor-induced neurite outgrowth and aquaporin-mediated water permeability in the presence and absence of oxidative stress. (R/S)-RC-752 emerged as the most interesting compound for S1R affinity (Ki S1R = 6.2 ± 0.9) and functional antagonist activity. Furthermore, it showed no cytotoxic effect in two normal human cell lines or in an in vivo zebrafish model and was stable after incubation in mouse plasma. (R/S)-RC-752 was then evaluated in two animal models of NP: the formalin test and the spinal nerve ligation model. The results clearly demonstrated that compound (R/S)-RC-752 effectively alleviated pain in both animal models, thus providing the proof of concept of its efficacy as an antinociceptive agent.
Collapse
Affiliation(s)
- Giacomo Rossino
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy
| | - Annamaria Marra
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy
| | - Roberta Listro
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy
| | - Marco Peviani
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, 27100 Pavia, Italy
| | - Elena Poggio
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, 27100 Pavia, Italy
| | - Daniela Curti
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, 27100 Pavia, Italy
| | - Giorgia Pellavio
- Human Physiology Unit, Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Umberto Laforenza
- Human Physiology Unit, Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Giulio Dondio
- Aphad SrL, Via della Resistenza, 65, 20090 Buccinasco, Italy
| | - Dirk Schepmann
- Institut für Pharmazeutische und Medizinische Chemie, Westfälische Wilhelms-Universität Münster, Corrensstraße 48, D-48149 Münster, Germany
| | - Bernhard Wünsch
- Institut für Pharmazeutische und Medizinische Chemie, Westfälische Wilhelms-Universität Münster, Corrensstraße 48, D-48149 Münster, Germany
| | - Martina Bedeschi
- BioScience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", 47014 Meldola, Italy
| | - Noemi Marino
- BioScience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", 47014 Meldola, Italy
| | - Anna Tesei
- BioScience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", 47014 Meldola, Italy
| | - Hee-Jin Ha
- Medifron DBT, Seoul 08502, Republic of Korea
| | | | - Jihyae Ann
- Laboratory of Medicinal Chemistry, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
- JMackem Co. Ltd., Seoul 08826, Republic of Korea
| | - Jeewoo Lee
- Laboratory of Medicinal Chemistry, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
- JMackem Co. Ltd., Seoul 08826, Republic of Korea
| | - Pasquale Linciano
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy
| | | | - Daniela Rossi
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy
| | - Simona Collina
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
8
|
Ruiz-Cantero MC, Cortés-Montero E, Jain A, Montilla-García Á, Bravo-Caparrós I, Shim J, Sánchez-Blázquez P, Woolf CJ, Baeyens JM, Cobos EJ. The sigma-1 receptor curtails endogenous opioid analgesia during sensitization of TRPV1 nociceptors. Br J Pharmacol 2023; 180:1148-1167. [PMID: 36478100 DOI: 10.1111/bph.16003] [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: 03/23/2022] [Revised: 08/18/2022] [Accepted: 08/21/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND PURPOSE Peripheral sensitization contributes to pathological pain. While prostaglandin E2 (PGE2) and nerve growth factor (NGF) sensitize peptidergic C-nociceptors (TRPV1+), glial cell line-derived neurotrophic factor (GDNF) sensitizes non-peptidergic C-neurons (IB4+). The sigma-1 receptor (sigma-1R) is a Ca2+ -sensing chaperone known to modulate opoid analgesia. This receptor binds both to TRPV1 and the μ opioid receptor, although the functional repercussions of these physical interactions in peripheral sensitization are unknown. EXPERIMENTAL APPROACH We tested the effects of sigma-1 antagonism on PGE2-, NGF-, and GDNF-induced mechanical and heat hyperalgesia in mice. We used immunohistochemistry to determine the presence of endomorphin-2, an endogenous μ receptor agonist, on dorsal root ganglion (DRG) neurons. Recombinant proteins were used to study the interactions between sigma-1R, μ- receptor, and TRPV1. We used calcium imaging to study the effects of sigma-1 antagonism on PGE2-induced sensitization of TRPV1+ nociceptors. KEY RESULTS Sigma1 antagonists reversed PGE2- and NGF-induced hyperalgesia but not GDNF-induced hyperalgesia. Endomorphin-2 was detected on TRPV1+ but not on IB4+ neurons. Peripheral opioid receptor antagonism by naloxone methiodide or administration of an anti-endomorphin-2 antibody to a sensitized paw reversed the antihyperalgesia induced by sigma-1 antagonists. Sigma-1 antagonism transfers sigma-1R from TRPV1 to μ receptors, suggesting that sigma-1R participate in TRPV1-μ receptor crosstalk. Moreover, sigma-1 antagonism reversed, in a naloxone-sensitive manner, PGE2-induced sensitization of DRG neurons to the calcium flux elicited by capsaicin, the prototypic TRPV1 agonist. CONCLUSION AND IMPLICATIONS Sigma-1 antagonism harnesses endogenous opioids produced by TRPV1+ neurons to reduce hyperalgesia by increasing μ receptor activity.
Collapse
Affiliation(s)
- M Carmen Ruiz-Cantero
- Department of Pharmacology, Faculty of Medicine, University of Granada, Granada, Spain.,Institute of Neuroscience, Biomedical Research Center, University of Granada, Granada, Spain.,Biosanitary Research Institute ibs. GRANADA, Granada, Spain
| | - Elsa Cortés-Montero
- Department of Translational Neurosciences, Neuropharmacology, Cajal Institute, CSIC, Madrid, Spain
| | - Aakanksha Jain
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA.,F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
| | - Ángeles Montilla-García
- Department of Pharmacology, Faculty of Medicine, University of Granada, Granada, Spain.,Institute of Neuroscience, Biomedical Research Center, University of Granada, Granada, Spain
| | - Inmaculada Bravo-Caparrós
- Department of Pharmacology, Faculty of Medicine, University of Granada, Granada, Spain.,Institute of Neuroscience, Biomedical Research Center, University of Granada, Granada, Spain
| | - Jaehoon Shim
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA.,F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
| | - Pilar Sánchez-Blázquez
- Department of Translational Neurosciences, Neuropharmacology, Cajal Institute, CSIC, Madrid, Spain
| | - Clifford J Woolf
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA.,F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
| | - José M Baeyens
- Department of Pharmacology, Faculty of Medicine, University of Granada, Granada, Spain.,Institute of Neuroscience, Biomedical Research Center, University of Granada, Granada, Spain.,Biosanitary Research Institute ibs. GRANADA, Granada, Spain
| | - Enrique J Cobos
- Department of Pharmacology, Faculty of Medicine, University of Granada, Granada, Spain.,Institute of Neuroscience, Biomedical Research Center, University of Granada, Granada, Spain.,Biosanitary Research Institute ibs. GRANADA, Granada, Spain.,Teófilo Hernando Institute for Drug Discovery, Madrid, Spain
| |
Collapse
|
9
|
Padín JF, Maroto M, Entrena JM, Egea J, Montell E, Vergés J, López MG, Cobos EJ, García AG. Small Synthetic Hyaluronan Disaccharide BIS014 Mitigates Neuropathic Pain in Mice. THE JOURNAL OF PAIN 2023; 24:68-83. [PMID: 36087908 DOI: 10.1016/j.jpain.2022.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 07/18/2022] [Accepted: 07/31/2022] [Indexed: 02/08/2023]
Abstract
Neuropathic pain (NP) is a challenging condition to treat, as the need for new drugs to treat NP is an unmet goal. We investigated the analgesic potential of a new sulfated disaccharide compound, named BIS014. Oral administration (p.o.) of this compound induced ameliorative effects in formalin-induced nociception and capsaicin-induced secondary mechanical hypersensitivity in mice, but also after partial sciatic nerve transection (spared nerve injury), chemotherapy (paclitaxel)-induced NP, and diabetic neuropathy induced by streptozotocin. Importantly, BIS014, at doses active on neuropathic hypersensitivity (60 mg/kg/p.o.), did not alter exploratory activity or motor coordination (in the rotarod test), unlike a standard dose of gabapentin (40 mg/kg/p.o.) which although inducing antiallodynic effects on the NP models, it also markedly decreased exploration and motor coordination. In docking and molecular dynamic simulation studies, BIS014 interacted with TRPV1, a receptor involved in pain transmission where it behaved as a partial agonist. Additionally, similar to capsaicin, BIS014 increased cytosolic Ca2+ concentration ([Ca2+]c) in neuroblastoma cells expressing TRPV1 receptors; these elevations were blocked by ruthenium red. BIS014 did not block capsaicin-elicited [Ca2+]c transients, but inhibited the increase in the firing rate of action potentials in bradykinin-sensitized dorsal root ganglion neurons stimulated with capsaicin. Perspective: We report that the oral administration of a new sulfated disaccharide compound, named BIS014, decreases neuropathic pain from diverse etiology in mice. Unlike the comparator gabapentin, BIS014 does not induce sedation. Thus, BIS014 has the potential to become a new efficacious non-sedative oral medication for the treatment of neuropathic pain.
Collapse
Affiliation(s)
- Juan-Fernando Padín
- Instituto-Fundación Teófilo Hernando, C/ Faraday 7, Parque Científico del Campus de Cantoblanco, Universidad Autónoma de Madrid, Madrid, Spain; Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, Avda. Arzobispo Morcillo 4, Madrid, Spain; Departamento de Ciencias Médicas (Farmacología), Facultad de Medicina, Universidad de Castilla-La Mancha, Ciudad Real, Spain.
| | - Marcos Maroto
- Instituto-Fundación Teófilo Hernando, C/ Faraday 7, Parque Científico del Campus de Cantoblanco, Universidad Autónoma de Madrid, Madrid, Spain.
| | - José Manuel Entrena
- Unidad de Análisis de Comportamiento Animal, Centro de Instrumentación Científica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Armilla, Granada, Spain.
| | - Javier Egea
- Instituto de Investigación Sanitaria del Hospital Universitario La Princesa (IIS La Princesa), C/Diego de León 62 (1ª planta), Madrid, Spain.
| | - Eulàlia Montell
- Pre-Clinical R&D Department, Bioibérica, S.A., Barcelona, Spain.
| | - Josep Vergés
- Pre-Clinical R&D Department, Bioibérica, S.A., Barcelona, Spain.
| | - Manuela G López
- Instituto-Fundación Teófilo Hernando, C/ Faraday 7, Parque Científico del Campus de Cantoblanco, Universidad Autónoma de Madrid, Madrid, Spain; Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, Avda. Arzobispo Morcillo 4, Madrid, Spain; Instituto de Investigación Sanitaria del Hospital Universitario La Princesa (IIS La Princesa), C/Diego de León 62 (1ª planta), Madrid, Spain.
| | - Enrique J Cobos
- Departamento de Farmacología e Instituto de Neurociencias, Facultad de Medicina, Universidad de Granada e Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain.
| | - Antonio G García
- Instituto-Fundación Teófilo Hernando, C/ Faraday 7, Parque Científico del Campus de Cantoblanco, Universidad Autónoma de Madrid, Madrid, Spain; Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, Avda. Arzobispo Morcillo 4, Madrid, Spain; Instituto de Investigación Sanitaria del Hospital Universitario La Princesa (IIS La Princesa), C/Diego de León 62 (1ª planta), Madrid, Spain.
| |
Collapse
|
10
|
From dopamine 4 to sigma 1: Synthesis, SAR and biological characterization of a piperidine scaffold of σ1 modulators. Eur J Med Chem 2022; 244:114840. [DOI: 10.1016/j.ejmech.2022.114840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 10/07/2022] [Accepted: 10/08/2022] [Indexed: 11/20/2022]
|
11
|
Marcotti A, Fernández-Trillo J, González A, Vizcaíno-Escoto M, Ros-Arlanzón P, Romero L, Vela JM, Gomis A, Viana F, de la Peña E. TRPA1 modulation by Sigma-1 receptor prevents oxaliplatin-induced painful peripheral neuropathy. Brain 2022; 146:475-491. [PMID: 35871491 PMCID: PMC9924907 DOI: 10.1093/brain/awac273] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 05/30/2022] [Accepted: 07/03/2022] [Indexed: 11/13/2022] Open
Abstract
Chemotherapy-induced peripheral neuropathy is a frequent, disabling side effect of anticancer drugs. Oxaliplatin, a platinum compound used in the treatment of advanced colorectal cancer, often leads to a form of chemotherapy-induced peripheral neuropathy characterized by mechanical and cold hypersensitivity. Current therapies for chemotherapy-induced peripheral neuropathy are ineffective, often leading to the cessation of treatment. Transient receptor potential ankyrin 1 (TRPA1) is a polymodal, non-selective cation-permeable channel expressed in nociceptors, activated by physical stimuli and cellular stress products. TRPA1 has been linked to the establishment of chemotherapy-induced peripheral neuropathy and other painful neuropathic conditions. Sigma-1 receptor is an endoplasmic reticulum chaperone known to modulate the function of many ion channels and receptors. Sigma-1 receptor antagonist, a highly selective antagonist of Sigma-1 receptor, has shown effectiveness in a phase II clinical trial for oxaliplatin chemotherapy-induced peripheral neuropathy. However, the mechanisms involved in the beneficial effects of Sigma-1 receptor antagonist are little understood. We combined biochemical and biophysical (i.e. intermolecular Förster resonance energy transfer) techniques to demonstrate the interaction between Sigma-1 receptor and human TRPA1. Pharmacological antagonism of Sigma-1R impaired the formation of this molecular complex and the trafficking of functional TRPA1 to the plasma membrane. Using patch-clamp electrophysiological recordings we found that antagonists of Sigma-1 receptor, including Sigma-1 receptor antagonist, exert a marked inhibition on plasma membrane expression and function of human TRPA1 channels. In TRPA1-expressing mouse sensory neurons, Sigma-1 receptor antagonists reduced inward currents and the firing of actions potentials in response to TRPA1 agonists. Finally, in a mouse experimental model of oxaliplatin neuropathy, systemic treatment with a Sigma-1 receptor antagonists prevented the development of painful symptoms by a mechanism involving TRPA1. In summary, the modulation of TRPA1 channels by Sigma-1 receptor antagonists suggests a new strategy for the prevention and treatment of chemotherapy-induced peripheral neuropathy and could inform the development of novel therapeutics for neuropathic pain.
Collapse
Affiliation(s)
- Aida Marcotti
- Present address: Instituto de Farmacología Experimental de Córdoba (IFEC) – CONICET, Haya de la Torre y Medina Allende, Ciudad Universitaria, Córdoba 5000, Argentina
| | | | - Alejandro González
- Present address: Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Marta Vizcaíno-Escoto
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, 03550 San Juan de Alicante, Spain
| | - Pablo Ros-Arlanzón
- Present address: Institute for Health and Biomedical Research (ISABIAL), 03550 San Juan de Alicante, Spain
| | - Luz Romero
- WeLab Barcelona, Parc Científic de Barcelona, 08028 Barcelona, Spain
| | - José Miguel Vela
- WeLab Barcelona, Parc Científic de Barcelona, 08028 Barcelona, Spain
| | - Ana Gomis
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, 03550 San Juan de Alicante, Spain
| | - Félix Viana
- Correspondence may also be addressed to: Felix Viana E-mail:
| | - Elvira de la Peña
- Correspondence to: Elvira de la Peña Instituto de Neurociencias de Alicante Universidad Miguel Hernández-CSIC 03550 San Juan de Alicante, Spain E-mail:
| |
Collapse
|
12
|
Couly S, Goguadze N, Yasui Y, Kimura Y, Wang SM, Sharikadze N, Wu HE, Su TP. Knocking Out Sigma-1 Receptors Reveals Diverse Health Problems. Cell Mol Neurobiol 2022; 42:597-620. [PMID: 33095392 PMCID: PMC8062587 DOI: 10.1007/s10571-020-00983-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 10/14/2020] [Indexed: 02/07/2023]
Abstract
Sigma-1 receptor (Sig-1R) is a protein present in several organs such as brain, lung, and heart. In a cell, Sig-1R is mainly located across the membranes of the endoplasmic reticulum and more specifically at the mitochondria-associated membranes. Despite numerous studies showing that Sig-1R could be targeted to rescue several cellular mechanisms in different pathological conditions, less is known about its fundamental relevance. In this review, we report results from various studies and focus on the importance of Sig-1R in physiological conditions by comparing Sig-1R KO mice to wild-type mice in order to investigate the fundamental functions of Sig-1R. We note that the Sig-1R deletion induces cognitive, psychiatric, and motor dysfunctions, but also alters metabolism of heart. Finally, taken together, observations from different experiments demonstrate that those dysfunctions are correlated to poor regulation of ER and mitochondria metabolism altered by stress, which could occur with aging.
Collapse
Affiliation(s)
- Simon Couly
- Cellular Pathobiology Section, Integrative Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, DHHS, IRP, NIH, Triad Technology Center 333 Cassell Drive, Baltimore, MD, 21224 NIH, USA.
| | - Nino Goguadze
- Cellular Pathobiology Section, Integrative Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, DHHS, IRP, NIH, Triad Technology Center 333 Cassell Drive, Baltimore, MD, 21224 NIH, USA
| | - Yuko Yasui
- Cellular Pathobiology Section, Integrative Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, DHHS, IRP, NIH, Triad Technology Center 333 Cassell Drive, Baltimore, MD, 21224 NIH, USA
| | - Yuriko Kimura
- Cellular Pathobiology Section, Integrative Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, DHHS, IRP, NIH, Triad Technology Center 333 Cassell Drive, Baltimore, MD, 21224 NIH, USA
| | - Shao-Ming Wang
- Cellular Pathobiology Section, Integrative Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, DHHS, IRP, NIH, Triad Technology Center 333 Cassell Drive, Baltimore, MD, 21224 NIH, USA
| | - Nino Sharikadze
- Cellular Pathobiology Section, Integrative Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, DHHS, IRP, NIH, Triad Technology Center 333 Cassell Drive, Baltimore, MD, 21224 NIH, USA
| | - Hsiang-En Wu
- Cellular Pathobiology Section, Integrative Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, DHHS, IRP, NIH, Triad Technology Center 333 Cassell Drive, Baltimore, MD, 21224 NIH, USA
| | - Tsung-Ping Su
- Cellular Pathobiology Section, Integrative Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, DHHS, IRP, NIH, Triad Technology Center 333 Cassell Drive, Baltimore, MD, 21224 NIH, USA
| |
Collapse
|
13
|
Cottilli P, Gaja-Capdevila N, Navarro X. Effects of Sigma-1 Receptor Ligands on Peripheral Nerve Regeneration. Cells 2022; 11:1083. [PMID: 35406646 PMCID: PMC8998141 DOI: 10.3390/cells11071083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/20/2022] [Accepted: 03/22/2022] [Indexed: 12/01/2022] Open
Abstract
Peripheral nerve injuries lead to the loss of motor, sensory and autonomic functions in the territories supplied by the injured nerve. Currently, nerve injuries are managed by surgical repair procedures, and there are no effective drugs in the clinic for improving the capacity of axonal regeneration. Sigma-1 receptor (Sig-1R) is an endoplasmic reticulum chaperon protein involved in many functions, including neuroprotection and neuroplasticity. A few previous studies using Sig-1R ligands reported results that suggest this receptor as a putative target to enhance regeneration. The aim of this study was to evaluate the possible effects of Sig-1R ligands on axonal regeneration in a sciatic nerve section and repair model in mice. To this end, mice were treated either with the Sig-1R agonist PRE-084 or the antagonist BD1063, and a Sig-1R knock-out (KO) mice group was also studied. The electrophysiological and histological data showed that treatment with Sig-1R ligands, or the lack of this protein, did not markedly modify the process of axonal regeneration and target reinnervation after sciatic nerve injury. Nevertheless, the nociceptive tests provided results indicating a role of Sig-1R in sensory perception after nerve injury, and immunohistochemical labeling indicated a regulatory role in inflammatory cell infiltration in the injured nerve.
Collapse
Affiliation(s)
- Patrick Cottilli
- Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat Autònoma de Barcelona, 01893 Bellaterra, Spain; (P.C.); (N.G.-C.)
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| | - Núria Gaja-Capdevila
- Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat Autònoma de Barcelona, 01893 Bellaterra, Spain; (P.C.); (N.G.-C.)
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| | - Xavier Navarro
- Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat Autònoma de Barcelona, 01893 Bellaterra, Spain; (P.C.); (N.G.-C.)
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| |
Collapse
|
14
|
Dichiara M, Artacho-Cordón A, Turnaturi R, Santos-Caballero M, González-Cano R, Pasquinucci L, Barbaraci C, Rodríguez-Gómez I, Gómez-Guzmán M, Marrazzo A, Cobos EJ, Amata E. Dual Sigma-1 receptor antagonists and hydrogen sulfide-releasing compounds for pain treatment: Design, synthesis, and pharmacological evaluation. Eur J Med Chem 2022; 230:114091. [PMID: 35016113 DOI: 10.1016/j.ejmech.2021.114091] [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: 10/26/2021] [Revised: 12/22/2021] [Accepted: 12/26/2021] [Indexed: 11/04/2022]
Abstract
The development of σ1 receptor antagonists hybridized with a H2S-donor is here reported. We aimed to obtain improved analgesic effects when compared to σ1 receptor antagonists or H2S-donors alone. In an in vivo model of sensory hypersensitivity, thioamide 1a induced analgesia which was synergistically enhanced when associated with the σ1 receptor antagonist BD-1063. The selective σ1 receptor agonist PRE-084 completely reversed this effect. Four thioamide H2S-σ1 receptor hybrids (5a-8a) and their amide derivatives (5b-8b) were synthesized. Compound 7a (AD164) robustly released H2S and showed selectivity for σ1 receptor over σ2 and opioid receptors. This compound induced marked analgesia that was reversed by PRE-084. The amide analogue 7b (AD163) showed only minimal analgesia. Further studies showed that 7a exhibited negligible acute toxicity, together with a favorable pharmacokinetic profile. To the best of our knowledge, compound 7a is the first dual-acting ligand with simultaneous H2S-release and σ1 antagonistic activities.
Collapse
Affiliation(s)
- Maria Dichiara
- Department of Drug and Health Sciences, Medicinal Chemistry Section, Università Degli Studi di Catania, Viale Andrea Doria 6, 95125, Catania, Italy
| | - Antonia Artacho-Cordón
- Department of Pharmacology, Faculty of Medicine and Biomedical Research Center (Neurosciences Institute), University of Granada and Biosanitary Research Institute Ibs.Granada, 18016, Granada, Spain
| | - Rita Turnaturi
- Department of Drug and Health Sciences, Medicinal Chemistry Section, Università Degli Studi di Catania, Viale Andrea Doria 6, 95125, Catania, Italy
| | - Miriam Santos-Caballero
- Department of Pharmacology, Faculty of Medicine and Biomedical Research Center (Neurosciences Institute), University of Granada and Biosanitary Research Institute Ibs.Granada, 18016, Granada, Spain
| | - Rafael González-Cano
- Department of Pharmacology, Faculty of Medicine and Biomedical Research Center (Neurosciences Institute), University of Granada and Biosanitary Research Institute Ibs.Granada, 18016, Granada, Spain
| | - Lorella Pasquinucci
- Department of Drug and Health Sciences, Medicinal Chemistry Section, Università Degli Studi di Catania, Viale Andrea Doria 6, 95125, Catania, Italy
| | - Carla Barbaraci
- Department of Drug and Health Sciences, Medicinal Chemistry Section, Università Degli Studi di Catania, Viale Andrea Doria 6, 95125, Catania, Italy
| | - Isabel Rodríguez-Gómez
- Department of Physiology, Faculty of Medicine, University of Granada, 18016, Granada, Spain
| | - Manuel Gómez-Guzmán
- Department of Pharmacology, Faculty of Pharmacy and Biomedical Research Center, University of Granada and Biosanitary Research Institute Ibs.Granada, 18016, Granada, Spain
| | - Agostino Marrazzo
- Department of Drug and Health Sciences, Medicinal Chemistry Section, Università Degli Studi di Catania, Viale Andrea Doria 6, 95125, Catania, Italy
| | - Enrique J Cobos
- Department of Pharmacology, Faculty of Medicine and Biomedical Research Center (Neurosciences Institute), University of Granada and Biosanitary Research Institute Ibs.Granada, 18016, Granada, Spain.
| | - Emanuele Amata
- Department of Drug and Health Sciences, Medicinal Chemistry Section, Università Degli Studi di Catania, Viale Andrea Doria 6, 95125, Catania, Italy.
| |
Collapse
|
15
|
Rabiner EA, Smith K, Bennett C, Rizzo G, Lewis Y, Mundin G, Dooner H, Oksche A. Pharmacokinetics and brain σ1 receptor occupancy of MR309, a selective σ1 receptor antagonist. Br J Clin Pharmacol 2022; 88:1644-1654. [PMID: 34156715 DOI: 10.1111/bcp.14952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 06/02/2021] [Accepted: 06/09/2021] [Indexed: 11/30/2022] Open
Abstract
AIMS Preclinical studies of MR309, a selective sigma-1 receptor (σ1R) antagonist, support a potential role in treating neuropathic pain. We report 2 studies that provide insight into the pharmacokinetics (PK) and brain σ1R binding of MR309. METHODS Steady-state PK of MR309 (400 mg once daily and 200 mg twice-daily [BID] for 10 days; EudraCT 2015-001818-99 [PK study]) and the relationship between MR309 plasma exposure and brain σ1R occupancy (EudraCT 2017-000670-11 [positron emission tomography study]) were investigated in healthy volunteers. Positron emission tomography using the σ1R ligand [11 C]SA4503 was conducted at baseline, and 2 and 8 hours after a single dose of MR309 (200-800 mg). The relationship between brain σ1R occupancy and MR309 exposure was explored using data-driven model fitting. RESULTS MR309 was well tolerated, brain σ1R occupancy ranged between 30.5 and 74.9% following single-dose MR309 (n = 7). MR309 BID provided a plasma PK profile with less fluctuation than once daily dosing (n = 16). MR309 200 mg BID yielded average steady state plasma concentrations between 2000 and 4000 ng/mL in the PK study, which corresponded to an estimated brain σ1R occupancy of 59-74%. CONCLUSION MR309 200 mg BID dose was below the 75% σ1R occupancy threshold expected to elicit maximal antinociceptive effect as observed in neuropathic pain models. Further investigations of MR309 for neuropathic pain will require higher brain σ1R occupancy, and establish the optimal dose by elucidating the clinical impact of a broad range of brain σ1R occupancy across different neuropathic pain indications.
Collapse
Affiliation(s)
- Eugenii A Rabiner
- Invicro(former Imanova Ltd), A Konica Minolta Company, London, UK
- Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, UK
| | | | | | - Gaia Rizzo
- Invicro(former Imanova Ltd), A Konica Minolta Company, London, UK
- Division of Brain Sciences, Department of Medicine, Imperial College, London, UK
| | - Yvonne Lewis
- Invicro(former Imanova Ltd), A Konica Minolta Company, London, UK
| | | | | | - Alexander Oksche
- Mundipharma Research Limited, Cambridge, UK
- Institut für medizinische und pharmazeutische Prüfungsfragen, Mainz, Germany
- Rudolf-Buchheim Institute of Pharmacology, Giessen, Germany
| |
Collapse
|
16
|
Shin SM, Wang F, Qiu C, Itson-Zoske B, Hogan QH, Yu H. Sigma-1 receptor activity in primary sensory neurons is a critical driver of neuropathic pain. Gene Ther 2022; 29:1-15. [PMID: 32424233 PMCID: PMC7671947 DOI: 10.1038/s41434-020-0157-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 04/21/2020] [Accepted: 05/04/2020] [Indexed: 12/15/2022]
Abstract
The Sigma-1 receptor (σ1R) is highly expressed in the primary sensory neurons (PSNs) that are the critical site of initiation and maintenance of pain following peripheral nerve injury. By immunoblot and immunohistochemistry, we observed increased expression of both σ1R and σ1R-binding immunoglobulin protein (BiP) in the lumbar (L) dorsal root ganglia (DRG) ipsilateral to painful neuropathy induced by spared nerve injury (SNI). To evaluate the therapeutic potential of PSN-targeted σ1R inhibition at a selected segmental level, we designed a recombinant adeno-associated viral (AAV) vector expressing a small hairpin RNA (shRNA) against rat σ1R. Injection of this vector into the L4/L5 DRGs induced downregulation of σ1R in DRG neurons of all size groups, while expression of BiP was not affected. This was accompanied by attenuation of SNI-induced cutaneous mechanical and thermal hypersensitivity. Whole-cell current-clamp recordings of dissociated neurons showed that knockdown of σ1R suppressed neuronal excitability, suggesting that σ1R silencing attenuates pain by reversal of injury-induced neuronal hyperexcitability. These findings support a critical role of σ1R in modulating PSN nociceptive functions, and that the nerve injury-induced elevated σ1R activity in the PSNs can be a significant driver of neuropathic pain. Further understanding the role of PSN-σ1R in pain pathology may open routes to exploit this system for DRG-targeted pain therapy.
Collapse
Affiliation(s)
- Seung Min Shin
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
- Zablocki Veterans Affairs Medical Center, Milwaukee, WI, 53295, USA
| | - Fei Wang
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
- Medical Experiment Center, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi, PR China
| | - Chensheng Qiu
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
- Department of Orthopedic Surgery, Affiliated Hospital of Qingdao University, Qingdao, 266000, PR China
| | - Brandon Itson-Zoske
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Quinn H Hogan
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
- Zablocki Veterans Affairs Medical Center, Milwaukee, WI, 53295, USA
| | - Hongwei Yu
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA.
- Zablocki Veterans Affairs Medical Center, Milwaukee, WI, 53295, USA.
| |
Collapse
|
17
|
Wilson LL, Eans SO, Ramadan-Siraj I, Modica MN, Romeo G, Intagliata S, McLaughlin JP. Examination of the Novel Sigma-1 Receptor Antagonist, SI 1/28, for Antinociceptive and Anti-allodynic Efficacy against Multiple Types of Nociception with Fewer Liabilities of Use. Int J Mol Sci 2022; 23:615. [PMID: 35054797 PMCID: PMC8775934 DOI: 10.3390/ijms23020615] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 12/29/2021] [Indexed: 02/05/2023] Open
Abstract
Neuropathic pain is a significant problem with few effective treatments lacking adverse effects. The sigma-1 receptor (S1R) is a potential therapeutic target for neuropathic pain, as antagonists for this receptor effectively ameliorate pain in both preclinical and clinical studies. The current research examines the antinociceptive and anti-allodynic efficacy of SI 1/28, a recently reported benzylpiperazine derivative and analog of the S1R antagonist SI 1/13, that was 423-fold more selective for S1R over the sigma-2 receptor (S2R). In addition, possible liabilities of respiration, sedation, and drug reinforcement caused by SI 1/28 have been evaluated. Inflammatory and chemical nociception, chronic nerve constriction injury (CCI) induced mechanical allodynia, and adverse effects of sedation in a rotarod assay, conditioned place preference (CPP), and changes in breath rate and locomotor activity were assessed after i.p. administration of SI 1/28. Pretreatment with SI 1/28 produced dose-dependent antinociception in the formalin test, with an ED50 (and 95% C.I.) value of 13.2 (7.42-28.3) mg/kg, i.p. Likewise, SI 1/28 produced dose-dependent antinociception against visceral nociception and anti-allodynia against CCI-induced neuropathic pain. SI 1/28 demonstrated no impairment of locomotor activity, conditioned place preference, or respiratory depression. In summary, SI 1/28 proved efficacious in the treatment of acute inflammatory pain and chronic neuropathy without liabilities at therapeutic doses, supporting the development of S1R antagonists as therapeutics for chronic pain.
Collapse
Affiliation(s)
- Lisa L. Wilson
- Department of Pharmacodynamics, The University of Florida, Gainesville, FL 32610, USA; (L.L.W.); (S.O.E.); (I.R.-S.)
| | - Shainnel O. Eans
- Department of Pharmacodynamics, The University of Florida, Gainesville, FL 32610, USA; (L.L.W.); (S.O.E.); (I.R.-S.)
| | - Insitar Ramadan-Siraj
- Department of Pharmacodynamics, The University of Florida, Gainesville, FL 32610, USA; (L.L.W.); (S.O.E.); (I.R.-S.)
| | - Maria N. Modica
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy; (M.N.M.); (G.R.)
| | - Giuseppe Romeo
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy; (M.N.M.); (G.R.)
| | - Sebastiano Intagliata
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy; (M.N.M.); (G.R.)
| | - Jay P. McLaughlin
- Department of Pharmacodynamics, The University of Florida, Gainesville, FL 32610, USA; (L.L.W.); (S.O.E.); (I.R.-S.)
| |
Collapse
|
18
|
Sałaciak K, Pytka K. Revisiting the sigma-1 receptor as a biological target to treat affective and cognitive disorders. Neurosci Biobehav Rev 2022; 132:1114-1136. [PMID: 34736882 PMCID: PMC8559442 DOI: 10.1016/j.neubiorev.2021.10.037] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/25/2021] [Accepted: 10/28/2021] [Indexed: 12/21/2022]
Abstract
Depression and cognitive disorders are diseases with complex and not-fully understood etiology. Unfortunately, the COVID-19 pandemic dramatically increased the prevalence of both conditions. Since the current treatments are inadequate in many patients, there is a constant need for discovering new compounds, which will be more effective in ameliorating depressive symptoms and treating cognitive decline. Proteins attracting much attention as potential targets for drugs treating these conditions are sigma-1 receptors. Sigma-1 receptors are multi-functional proteins localized in endoplasmic reticulum membranes, which play a crucial role in cellular signal transduction by interacting with receptors, ion channels, lipids, and kinases. Changes in their functions and expression may lead to various diseases, including depression or memory impairments. Thus, sigma-1 receptor modulation might be useful in treating these central nervous system diseases. Importantly, two sigma-1 receptor ligands entered clinical trials, showing that this compound group possesses therapeutic potential. Therefore, based on preclinical studies, this review discusses whether the sigma-1 receptor could be a promising target for drugs treating affective and cognitive disorders.
Collapse
Affiliation(s)
- Kinga Sałaciak
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland
| | - Karolina Pytka
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland.
| |
Collapse
|
19
|
Choi SR, Han HJ, Beitz AJ, Lee JH. Intrathecal interleukin-1β decreases sigma-1 receptor expression in spinal astrocytes in a murine model of neuropathic pain. Biomed Pharmacother 2021; 144:112272. [PMID: 34607109 DOI: 10.1016/j.biopha.2021.112272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/18/2021] [Accepted: 09/27/2021] [Indexed: 10/20/2022] Open
Abstract
The sigma-1 receptor (Sig-1R) plays an important role in spinal pain transmission by increasing phosphorylation of the N-methyl-D-aspartate (NMDA) receptor GluN1 subunit (pGluN1). As a result Sig-1R has been suggested as a novel therapeutic target for prevention of chronic pain. Here we investigated whether interleukin-1β (IL-1β) modulates the expression of the Sig-1R in spinal astrocytes during the early phase of nerve injury, and whether this modulation affects spinal pGluN1 expression and the development of neuropathic pain following chronic constriction injury (CCI) of the sciatic nerve. Repeated intrathecal (i.t.) administration of IL-1β from days 0-3 post-surgery significantly reduced the increased pGluN1 expression at the Ser896 and Ser897 sites in the ipsilateral spinal cord, as well as, the development of mechanical allodynia and thermal hyperalgesia in the ipsilateral hind paw of CCI mice, which were restored by co-administration of IL-1 receptor antagonist with IL-1β. Sciatic nerve injury increased the expression of Sig-1R in astrocytes of the ipsilateral spinal cord, and this increase was suppressed by i.t. administration of IL-1β. Agonistic stimulation of the Sig-1R with PRE084 restored pGluN1 expression and the development of mechanical allodynia that were originally suppressed by IL-1β in CCI mice. Collectively these results demonstrate that IL-1β administration during the induction phase of neuropathic pain produces an analgesic effect on neuropathic pain development by controlling the expression of Sig-1R in spinal astrocytes.
Collapse
Affiliation(s)
- Sheu-Ran Choi
- Department of Pharmacology, Catholic Kwandong University College of Medicine, Gangneung 25601, Republic of Korea; Department of Veterinary Physiology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea.
| | - Ho Jae Han
- Department of Veterinary Physiology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea.
| | - Alvin J Beitz
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, MN 55108, USA.
| | - Jang-Hern Lee
- Department of Veterinary Physiology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
20
|
Rodríguez-Muñoz M, Cortés-Montero E, Onetti Y, Sánchez-Blázquez P, Garzón-Niño J. The σ1 Receptor and the HINT1 Protein Control α2δ1 Binding to Glutamate NMDA Receptors: Implications in Neuropathic Pain. Biomolecules 2021; 11:1681. [PMID: 34827679 PMCID: PMC8615847 DOI: 10.3390/biom11111681] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/29/2021] [Accepted: 11/08/2021] [Indexed: 02/01/2023] Open
Abstract
Nerve injury produces neuropathic pain through the binding of α2δ1 proteins to glutamate N-methyl-D-aspartate receptors (NMDARs). Notably, mice with a targeted deletion of the sigma 1 receptor (σ1R) gene do not develop neuropathy, whereas mice lacking the histidine triad nucleotide-binding protein 1 (Hint1) gene exhibit exacerbated allodynia. σ1R antagonists more effectively diminish neuropathic pain of spinal origin when administered by intracerebroventricular injection than systemically. Thus, in mice subjected to unilateral sciatic nerve chronic constriction injury (CCI), we studied the participation of σ1Rs and HINT1 proteins in the formation of α2δ1-NMDAR complexes within the supraspinal periaqueductal gray (PAG). We found that δ1 peptides required σ1Rs in order to interact with the NMDAR NR1 variant that contains the cytosolic C1 segment. σ1R antagonists or low calcium levels provoke the dissociation of σ1R-NR1 C1 dimers, while they barely affect the integrity of δ1-σ1R-NR1 C1 trimers. However, HINT1 does remove δ1 peptides from the trimer, thereby facilitating the subsequent dissociation of σ1Rs from NMDARs. In σ1R-/- mice, CCI does not promote the formation of NMDAR-α2δ1 complexes and allodynia does not develop. The levels of α2δ1-σ1R-NMDAR complexes increase in HINT1-/- mice and after inducing CCI, degradation of α2δ1 proteins is observed. Notably, σ1R antagonists but not gabapentinoids alleviate neuropathic pain in these mice. During severe neuropathy, the metabolism of α2δ1 proteins may account for the failure of many patients to respond to gabapentinoids. Therefore, σ1Rs promote and HINT1 proteins hinder the formation α2δ1-NMDAR complexes in the PAG, and hence, the appearance of mechanical allodynia depends on the interplay between these proteins.
Collapse
Affiliation(s)
- María Rodríguez-Muñoz
- Neuropharmacology, Department of Translational Neuroscience, Cajal Institute, CSIC, 28002 Madrid, Spain; (M.R.-M.); (E.C.-M.); (Y.O.); (P.S.-B.)
| | - Elsa Cortés-Montero
- Neuropharmacology, Department of Translational Neuroscience, Cajal Institute, CSIC, 28002 Madrid, Spain; (M.R.-M.); (E.C.-M.); (Y.O.); (P.S.-B.)
| | - Yara Onetti
- Neuropharmacology, Department of Translational Neuroscience, Cajal Institute, CSIC, 28002 Madrid, Spain; (M.R.-M.); (E.C.-M.); (Y.O.); (P.S.-B.)
| | - Pilar Sánchez-Blázquez
- Neuropharmacology, Department of Translational Neuroscience, Cajal Institute, CSIC, 28002 Madrid, Spain; (M.R.-M.); (E.C.-M.); (Y.O.); (P.S.-B.)
| | - Javier Garzón-Niño
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Doctor Arce 37, 28002 Madrid, Spain
| |
Collapse
|
21
|
Zhuang T, Xiong J, Hao S, Du W, Liu Z, Liu B, Zhang G, Chen Y. Bifunctional μ opioid and σ 1 receptor ligands as novel analgesics with reduced side effects. Eur J Med Chem 2021; 223:113658. [PMID: 34175542 DOI: 10.1016/j.ejmech.2021.113658] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/13/2021] [Accepted: 06/15/2021] [Indexed: 12/13/2022]
Abstract
Opioid analgesics are highly effective painkillers for the treatment of moderate or severe pain, but they are associated with a number of undesirable adverse effects, including the development of tolerance, addiction, constipation and life-threatening respiratory depression. The development of new and safer analgesics with innovative mechanisms of action, which can enhance the efficacy in comparison to available treatments and reduce their side effects, is urgently needed. The sigma-1 receptor (σ1R), a unique Ca2+-sensing chaperone protein, is expressed throughout pain-modulating tissues and affects neurotransmission by interacting with different protein partners, including molecular targets that participate in nociceptive signalling, such as the μ-opioid receptor (MOR), N-methyl-d-aspartate receptor (NMDAR) and cannabinoid 1 receptor (CB1R). Overwhelming pharmacological and genetic evidence indicates that σ1R antagonists induce anti-hypersensitive effects in sensitising pain conditions (e.g. chemically induced, inflammatory and neuropathic pain) and enhance opioid analgesia but not opioid-mediated detrimental effects. It has been suggested that balanced modulation of MORs and σ1Rs may improve both the therapeutic efficacy and safety of opioids. This review summarises the functional profiles of ligands with mixed MOR agonist and σ1R antagonist activities and highlights their therapeutic potentials for pain management. Dual MOR agonism/σ1R antagonism represents a promising avenue for the development of potent and safer analgesics.
Collapse
MESH Headings
- Analgesics, Opioid/adverse effects
- Analgesics, Opioid/chemistry
- Analgesics, Opioid/metabolism
- Analgesics, Opioid/therapeutic use
- Benzopyrans/chemistry
- Benzopyrans/metabolism
- Humans
- Ligands
- Pain/drug therapy
- Piperazines/chemistry
- Piperazines/metabolism
- Receptors, Opioid, delta/antagonists & inhibitors
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/metabolism
- Receptors, sigma/antagonists & inhibitors
- Receptors, sigma/metabolism
- Sigma-1 Receptor
Collapse
Affiliation(s)
- Tao Zhuang
- Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Jiaying Xiong
- Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Shuaishuai Hao
- Jiangsu Key Laboratory of Marine Biological Resources and Environment, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, School of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Wei Du
- Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Zhenming Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
| | - Bifeng Liu
- Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Guisen Zhang
- Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China; Jiangsu Key Laboratory of Marine Biological Resources and Environment, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, School of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China.
| | - Yin Chen
- Jiangsu Key Laboratory of Marine Biological Resources and Environment, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, School of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China.
| |
Collapse
|
22
|
Sensory Neuron TLR4 mediates the development of nerve-injury induced mechanical hypersensitivity in female mice. Brain Behav Immun 2021; 97:42-60. [PMID: 34174335 PMCID: PMC8453057 DOI: 10.1016/j.bbi.2021.06.011] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 06/08/2021] [Accepted: 06/18/2021] [Indexed: 02/07/2023] Open
Abstract
Recent studies have brought to light the necessity to discern sex-specific differences in various pain states and different cell-types that mediate these differences. These studies have uncovered the role of neuroimmune interactions to mediate pain states in a sex-specific fashion. While investigating immune function in pain development, we discovered that females utilize immune components of sensory neurons to mediate neuropathic pain development. We utilized two novel transgenic mouse models that eitherrestore expression of toll-like receptor (TLR) 4 inNav1.8 nociceptors on a TLR4-null background (TLR4LoxTB) or remove TLR4 specifically from Nav1.8 nociceptors (TLR4fl/fl). After spared nerve injury (SNI), a model of neuropathic injury, we observed a robust female-specific onset of mechanical hypersensitivity in our transgenic animals. Female Nav1.8-TLR4fl/fl knockout animals were less mechanically sensitive than cre-negative TLR4fl/fl littermates. Conversely, female Nav1.8-TLR4LoxTB reactivated animals were as mechanically sensitive as their wild-type counterparts. These sex and cell-specific effects were not recapitulated in male animals of either strain. Additionally, we find the danger associated molecular pattern, high mobility group box-1 (HGMB1), a potent TLR4 agonist, localization and ATF3 expression in females is dependent on TLR4 expression in dorsal root ganglia (DRG) populations following SNI. These experiments provide novel evidence toward sensory neuron specific modulation of pain in a sex-dependent manner.
Collapse
|
23
|
Kopp N, Civenni G, Marson D, Laurini E, Pricl S, Catapano CV, Humpf HU, Almansa C, Nieto FR, Schepmann D, Wünsch B. Chemoenzymatic synthesis of 2,6-disubstituted tetrahydropyrans with high σ 1 receptor affinity, antitumor and analgesic activity. Eur J Med Chem 2021; 219:113443. [PMID: 33901806 DOI: 10.1016/j.ejmech.2021.113443] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/22/2021] [Accepted: 03/29/2021] [Indexed: 02/06/2023]
Abstract
1,3-Dioxanes 1 and cyclohexanes 2 bearing a phenyl ring and an aminoethyl moiety in 1,3-relationship to each other represent highly potent σ1 receptor antagonists. In order to increase the chemical stability of the acetalic 1,3-dioxanes 1 and the polarity of the cyclohexanes 2, tetrahydropyran derivatives 3 equipped with the same substituents were designed, synthesized and pharmacologically evaluated. The key step of the synthesis was a lipase-catalyzed enantioselective acetylation of the alcohol (R)-5 leading finally to enantiomerically pure test compounds 3a-g. With respect to σ1 receptor affinity and selectivity over a broad range of related (σ2, PCP binding site) and further targets, the enantiomeric benzylamines 3a and cyclohexylmethylamines 3b represent the most promising drug candidates of this series. However, the eudismic ratio for σ1 binding is only in the range of 2.5-3.3. Classical molecular dynamics (MD) simulations confirmed the same binding pose for both the tetrahydropyran 3 and cyclohexane derivatives 2 at the σ1 receptor, according to which: i) the protonated amino moiety of (2S,6R)-3a engages the same key polar interactions with Glu172 (ionic) and Phe107 (π-cation), ii) the lipophilic parts of (2S,6R)-3a are hosted in three hydrophobic regions of the σ1 receptor, and iii) the O-atom of the tetrahydropyran derivatives 3 does not show a relevant interaction with the σ1 receptor. Further in silico evidences obtained by the application of free energy perturbation and steered MD techniques fully supported the experimentally observed difference in receptor/ligand affinities. Tetrahydropyrans 3 require a lower dissociative force peak than cyclohexane analogs 2. Enantiomeric benzylamines 3a and cyclohexylmethylamines 3b were able to inhibit the growth of the androgen negative human prostate cancer cell line DU145. The cyclohexylmethylamine (2S,6R)-3b showed the highest σ1 affinity (Ki(σ1) = 0.95 nM) and the highest analgesic activity in vivo (67%).
Collapse
Affiliation(s)
- Nicole Kopp
- Institut für Pharmazeutische und Medizinische Chemie, Westfälische Wilhelms-Universität Münster, Corrensstraße 48, D-48149, Münster, Germany
| | - Gianluca Civenni
- Institute of Oncology Research, Università della Svizzera Italiana (USI), Via Vincenzo Vela 6, CH-6500, Bellinzona, Switzerland
| | - Domenico Marson
- Molecular Biology and Nanotechnology Laboratory (MolBNL@UniTS), DEA, University of Trieste, 34127, Trieste, Italy
| | - Erik Laurini
- Molecular Biology and Nanotechnology Laboratory (MolBNL@UniTS), DEA, University of Trieste, 34127, Trieste, Italy
| | - Sabrina Pricl
- Molecular Biology and Nanotechnology Laboratory (MolBNL@UniTS), DEA, University of Trieste, 34127, Trieste, Italy; Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Carlo V Catapano
- Institute of Oncology Research, Università della Svizzera Italiana (USI), Via Vincenzo Vela 6, CH-6500, Bellinzona, Switzerland
| | - Hans-Ulrich Humpf
- Institut für Lebensmittelchemie, Westfälische Wilhelms-Universität Münster, Corrensstraße 45, D-48149, Münster, Germany
| | - Carmen Almansa
- Esteve Pharmaceuticals S.A., Baldiri Reixach 4-8, 08028, Barcelona, Spain
| | - Francisco Rafael Nieto
- Department of Pharmacology and Neurosciences Institute (Biomedical Research Center), University of Granada and Biosanitary Research Institute, 18010, Granada, Spain
| | - Dirk Schepmann
- Institut für Pharmazeutische und Medizinische Chemie, Westfälische Wilhelms-Universität Münster, Corrensstraße 48, D-48149, Münster, Germany
| | - Bernhard Wünsch
- Institut für Pharmazeutische und Medizinische Chemie, Westfälische Wilhelms-Universität Münster, Corrensstraße 48, D-48149, Münster, Germany; GRK 2515, Chemical Biology of Ion Channels (Chembion), Westfälische Wilhelms-Universität Münster, Germany.
| |
Collapse
|
24
|
Abstract
This paper is the forty-second consecutive installment of the annual anthological review of research concerning the endogenous opioid system, summarizing articles published during 2019 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides and receptors as well as effects of opioid/opiate agonists and antagonists. The review is subdivided into the following specific topics: molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors (1), the roles of these opioid peptides and receptors in pain and analgesia in animals (2) and humans (3), opioid-sensitive and opioid-insensitive effects of nonopioid analgesics (4), opioid peptide and receptor involvement in tolerance and dependence (5), stress and social status (6), learning and memory (7), eating and drinking (8), drug abuse and alcohol (9), sexual activity and hormones, pregnancy, development and endocrinology (10), mental illness and mood (11), seizures and neurologic disorders (12), electrical-related activity and neurophysiology (13), general activity and locomotion (14), gastrointestinal, renal and hepatic functions (15), cardiovascular responses (16), respiration and thermoregulation (17), and immunological responses (18).
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, 65-30 Kissena Blvd., Flushing, NY, 11367, United States.
| |
Collapse
|
25
|
Romeo G, Bonanno F, Wilson LL, Arena E, Modica MN, Pittalà V, Salerno L, Prezzavento O, McLaughlin JP, Intagliata S. Development of New Benzylpiperazine Derivatives as σ 1 Receptor Ligands with in Vivo Antinociceptive and Anti-Allodynic Effects. ACS Chem Neurosci 2021; 12:2003-2012. [PMID: 34019387 PMCID: PMC8291485 DOI: 10.1021/acschemneuro.1c00106] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 05/10/2021] [Indexed: 12/22/2022] Open
Abstract
σ-1 receptors (σ1R) modulate nociceptive signaling, driving the search for selective antagonists to take advantage of this promising target to treat pain. In this study, a new series of benzylpiperazinyl derivatives has been designed, synthesized, and characterized for their affinities toward σ1R and selectivity over the σ-2 receptor (σ2R). Notably, 3-cyclohexyl-1-{4-[(4-methoxyphenyl)methyl]piperazin-1-yl}propan-1-one (15) showed the highest σ1R receptor affinity (Ki σ1 = 1.6 nM) among the series with a significant improvement of the σ1R selectivity (Ki σ2/Ki σ1= 886) compared to the lead compound 8 (Ki σ2/Ki σ1= 432). Compound 15 was further tested in a mouse formalin assay of inflammatory pain and chronic nerve constriction injury (CCI) of neuropathic pain, where it produced dose-dependent (3-60 mg/kg, i.p.) antinociception and anti-allodynic effects. Moreover, compound 15 demonstrated no significant effects in a rotarod assay, suggesting that this σ1R antagonist did not produce sedation or impair locomotor responses. Overall, these results encourage the further development of our benzylpiperazine-based σ1R antagonists as potential therapeutics for chronic pain.
Collapse
Affiliation(s)
- Giuseppe Romeo
- Department
of Drug and Health Sciences, University
of Catania, viale A. Doria 6, 95125 Catania, Italy
| | - Federica Bonanno
- Department
of Drug and Health Sciences, University
of Catania, viale A. Doria 6, 95125 Catania, Italy
| | - Lisa L. Wilson
- Department
of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Emanuela Arena
- Department
of Drug and Health Sciences, University
of Catania, viale A. Doria 6, 95125 Catania, Italy
| | - Maria N. Modica
- Department
of Drug and Health Sciences, University
of Catania, viale A. Doria 6, 95125 Catania, Italy
| | - Valeria Pittalà
- Department
of Drug and Health Sciences, University
of Catania, viale A. Doria 6, 95125 Catania, Italy
| | - Loredana Salerno
- Department
of Drug and Health Sciences, University
of Catania, viale A. Doria 6, 95125 Catania, Italy
| | - Orazio Prezzavento
- Department
of Drug and Health Sciences, University
of Catania, viale A. Doria 6, 95125 Catania, Italy
| | - Jay P. McLaughlin
- Department
of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Sebastiano Intagliata
- Department
of Drug and Health Sciences, University
of Catania, viale A. Doria 6, 95125 Catania, Italy
| |
Collapse
|
26
|
Ruiz-Cantero MC, González-Cano R, Tejada MÁ, Santos-Caballero M, Perazzoli G, Nieto FR, Cobos EJ. Sigma-1 receptor: A drug target for the modulation of neuroimmune and neuroglial interactions during chronic pain. Pharmacol Res 2021; 163:105339. [PMID: 33276102 DOI: 10.1016/j.phrs.2020.105339] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/22/2020] [Accepted: 11/23/2020] [Indexed: 12/12/2022]
Abstract
Immune and glial cells play a pivotal role in chronic pain. Therefore, it is possible that the pharmacological modulation of neurotransmission from an exclusively neuronal perspective may not be enough for adequate pain management, and the modulation of complex interactions between neurons and other cell types might be needed for successful pain relief. In this article, we review the current scientific evidence for the modulatory effects of sigma-1 receptors on communication between the immune and nervous systems during inflammation, as well as the influence of this receptor on peripheral and central neuroinflammation. Several experimental models of pathological pain are considered, including peripheral and central neuropathic pain, osteoarthritic, and cancer pain. Sigma-1 receptor inhibition prevents peripheral (macrophage infiltration into the dorsal root ganglion) and central (activation of microglia and astrocytes) neuroinflammation in several pain models, and enhances immune-driven peripheral opioid analgesia during painful inflammation, maximizing the analgesic potential of peripheral immune cells. Therefore, sigma-1 antagonists may constitute a new class of analgesics with an unprecedented mechanism of action and potential utility in several painful disorders.
Collapse
Affiliation(s)
- M Carmen Ruiz-Cantero
- Department of Pharmacology, and Neurosciences Institute (Biomedical Research Center), University of Granada, Granada, Spain; Biosanitary Research Institute ibs.GRANADA, Granada, Spain
| | - Rafael González-Cano
- Department of Pharmacology, and Neurosciences Institute (Biomedical Research Center), University of Granada, Granada, Spain; Biosanitary Research Institute ibs.GRANADA, Granada, Spain
| | - Miguel Á Tejada
- Department of Pharmacology, and Neurosciences Institute (Biomedical Research Center), University of Granada, Granada, Spain; INCLIVA Health Research Institute, Valencia, Spain
| | - Miriam Santos-Caballero
- Department of Pharmacology, and Neurosciences Institute (Biomedical Research Center), University of Granada, Granada, Spain; Biosanitary Research Institute ibs.GRANADA, Granada, Spain
| | - Gloria Perazzoli
- Biosanitary Research Institute ibs.GRANADA, Granada, Spain; Department of Nursing, Physiotherapy and Medicine, University of Almería, Almería, Spain
| | - Francisco R Nieto
- Department of Pharmacology, and Neurosciences Institute (Biomedical Research Center), University of Granada, Granada, Spain; Biosanitary Research Institute ibs.GRANADA, Granada, Spain.
| | - Enrique J Cobos
- Department of Pharmacology, and Neurosciences Institute (Biomedical Research Center), University of Granada, Granada, Spain; Biosanitary Research Institute ibs.GRANADA, Granada, Spain; Teófilo Hernando Institute for Drug Discovery, Madrid, Spain.
| |
Collapse
|
27
|
Jiménez-López J, Bravo-Caparrós I, Cabeza L, Nieto FR, Ortiz R, Perazzoli G, Fernández-Segura E, Cañizares FJ, Baeyens JM, Melguizo C, Prados J. Paclitaxel antitumor effect improvement in lung cancer and prevention of the painful neuropathy using large pegylated cationic liposomes. Biomed Pharmacother 2021; 133:111059. [PMID: 33378963 DOI: 10.1016/j.biopha.2020.111059] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 11/18/2020] [Accepted: 11/20/2020] [Indexed: 12/24/2022] Open
Abstract
Paclitaxel (PTX), a drug widely used in lung cancer, has serious limitations including the development of peripheral neurotoxicity, which may lead to treatment discontinuation and therapy failure. The transport of PTX in large cationic liposomes could avoid this undesirable effect, improving the patient's prognosis. PTX was encapsulated in cationic liposomes with two different sizes, MLV (180-200 nm) and SUV (80-100 nm). In both cases, excellent biocompatibility and improved internalization and antitumor effect of PTX were observed in human and mice lung cancer cells in culture, multicellular spheroids and cancer stem cells (CSCs). In addition, both MLV and SUV with a polyethylene glycol (PEG) shell, induced a greater tumor volume reduction than PTX (56.4 % and 57.1 % vs. 36.7 %, respectively) in mice. Interestingly, MLV-PEG-PTX did not induce either mechanical or heat hypersensitivity whereas SUV-PEG-PTX produced a similar response to free PTX. Analysis of PTX distribution showed a very low concentration of the drug in the dorsal root ganglia (DRG) with MLV-PEG-PTX, but not with SUV-PEG-PTX or free PTX. These results support the hypothesis that PTX induces peripheral neuropathy by penetrating the endothelial fenestrations of the DRG (80-100 nm, measured in mice). In conclusion, our larger liposomes (MLV-PEG-PTX) not only showed biocompatibility, antitumor activity against CSCs, and in vitro and in vivo antitumor effect that improved PTX free activity, but also protected from PTX-induced painful peripheral neuropathy. These advantages could be used as a new strategy of lung cancer chemotherapy to increase the PTX activity and reduce its side effects.
Collapse
Affiliation(s)
- Julia Jiménez-López
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100, Granada, Spain; Instituto Biosanitario de Granada (ibs. GRANADA), 18014, Granada, Spain; Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18012, Granada, Spain
| | - Inmaculada Bravo-Caparrós
- Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18012, Granada, Spain
| | - Laura Cabeza
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100, Granada, Spain; Instituto Biosanitario de Granada (ibs. GRANADA), 18014, Granada, Spain; Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18012, Granada, Spain
| | - Francisco R Nieto
- Department of Pharmacology, Institute of Neuroscience, Biomedical Research Center (CIBM), University of Granada, 18100, Granada, Spain
| | - Raúl Ortiz
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100, Granada, Spain; Instituto Biosanitario de Granada (ibs. GRANADA), 18014, Granada, Spain; Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18012, Granada, Spain
| | - Gloria Perazzoli
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100, Granada, Spain; Instituto Biosanitario de Granada (ibs. GRANADA), 18014, Granada, Spain
| | - Eduardo Fernández-Segura
- Department of Histology, Institute of Neuroscience, Biomedical Research Center (CIBM), University of Granada, 18100, Granada, Spain
| | - Francisco J Cañizares
- Department of Histology, Institute of Neuroscience, Biomedical Research Center (CIBM), University of Granada, 18100, Granada, Spain
| | - José M Baeyens
- Department of Pharmacology, Institute of Neuroscience, Biomedical Research Center (CIBM), University of Granada, 18100, Granada, Spain
| | - Consolación Melguizo
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100, Granada, Spain; Instituto Biosanitario de Granada (ibs. GRANADA), 18014, Granada, Spain; Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18012, Granada, Spain.
| | - José Prados
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100, Granada, Spain; Instituto Biosanitario de Granada (ibs. GRANADA), 18014, Granada, Spain; Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18012, Granada, Spain
| |
Collapse
|
28
|
Ye N, Qin W, Tian S, Xu Q, Wold EA, Zhou J, Zhen XC. Small Molecules Selectively Targeting Sigma-1 Receptor for the Treatment of Neurological Diseases. J Med Chem 2020; 63:15187-15217. [PMID: 33111525 DOI: 10.1021/acs.jmedchem.0c01192] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The sigma-1 (σ1) receptor, an enigmatic protein originally classified as an opioid receptor subtype, is now understood to possess unique structural and functional features of its own and play critical roles to widely impact signaling transduction by interacting with receptors, ion channels, lipids, and kinases. The σ1 receptor is implicated in modulating learning, memory, emotion, sensory systems, neuronal development, and cognition and accordingly is now an actively pursued drug target for various neurological and neuropsychiatric disorders. Evaluation of the five selective σ1 receptor drug candidates (pridopidine, ANAVEX2-73, SA4503, S1RA, and T-817MA) that have entered clinical trials has shown that reaching clinical approval remains an evasive and important goal. This review provides up-to-date information on the selective targeting of σ1 receptors, including their history, function, reported crystal structures, and roles in neurological diseases, as well as a useful collation of new chemical entities as σ1 selective orthosteric ligands or allosteric modulators.
Collapse
Affiliation(s)
- Na Ye
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Wangzhi Qin
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Sheng Tian
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Qingfeng Xu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Eric A Wold
- Chemical Biology Program, Department of Pharmacology and Toxicology, and Center for Addiction Research, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Jia Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, and Center for Addiction Research, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Xue-Chu Zhen
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| |
Collapse
|
29
|
Sánchez-Blázquez P, Cortés-Montero E, Rodríguez-Muñoz M, Merlos M, Garzón-Niño J. The Sigma 2 receptor promotes and the Sigma 1 receptor inhibits mu-opioid receptor-mediated antinociception. Mol Brain 2020; 13:150. [PMID: 33176836 PMCID: PMC7659117 DOI: 10.1186/s13041-020-00676-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 09/22/2020] [Indexed: 01/10/2023] Open
Abstract
The Sigma-1 receptor (σ1R) has emerged as an interesting pharmacological target because it inhibits analgesia mediated by mu-opioid receptors (MOR), and also facilitates the development of neuropathic pain. Based on these findings, the recent cloning of the Sigma-2 receptor (σ2R) led us to investigate its potential role as a regulator of opioid analgesia and of pain hypersensitivity in σ2R knockout mice. In contrast to σ1R deficient mice, σ2R knockout mice developed mechanical allodynia following establishment of chronic constriction injury-induced neuropathic pain, which was alleviated by the σ1R antagonist S1RA. The analgesic effects of morphine, [D-Ala, N-MePhe, Gly-ol]-encephalin (DAMGO) and β-endorphin increased in σ1R-/- mice and diminished in σ2R-/- mice. The analgesic effect of morphine was increased in σ2R-/- mice by treatment with S1RA. However, σ2R-/- mice and wild-type mice exhibited comparable antinociceptive responses to the delta receptor agonist [D-Pen2,5]-encephalin (DPDPE), the cannabinoid type 1 receptor agonist WIN55,212-2 and the α2-adrenergic receptor agonist clonidine. Therefore, while σR1 inhibits and σ2R facilitates MOR-mediated analgesia these receptors exchange their roles when regulating neuropathic pain perception. Our study may help identify new pharmacological targets for diminishing pain perception and improving opioid detoxification therapies.
Collapse
MESH Headings
- Analgesics/pharmacology
- Animals
- Constriction, Pathologic
- Hyperalgesia/metabolism
- Hyperalgesia/pathology
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Morphine/pharmacology
- Nociception/drug effects
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Opioid, mu/antagonists & inhibitors
- Receptors, Opioid, mu/metabolism
- Receptors, sigma/metabolism
- Sigma-1 Receptor
Collapse
Affiliation(s)
- Pilar Sánchez-Blázquez
- Neuropharmacology, Cajal Institute, Consejo Superior de Investigaciones Científicas (CSIC), Doctor Arce 37, 28002, Madrid, Spain.
| | - Elsa Cortés-Montero
- Neuropharmacology, Cajal Institute, Consejo Superior de Investigaciones Científicas (CSIC), Doctor Arce 37, 28002, Madrid, Spain
| | - María Rodríguez-Muñoz
- Neuropharmacology, Cajal Institute, Consejo Superior de Investigaciones Científicas (CSIC), Doctor Arce 37, 28002, Madrid, Spain
| | - Manuel Merlos
- Drug Discovery & Preclinical Development, Esteve, Barcelona, Spain
| | - Javier Garzón-Niño
- Neuropharmacology, Cajal Institute, Consejo Superior de Investigaciones Científicas (CSIC), Doctor Arce 37, 28002, Madrid, Spain
| |
Collapse
|
30
|
González-Cano R, Artacho-Cordón A, Romero L, Tejada MA, Nieto FR, Merlos M, Cañizares FJ, Cendán CM, Fernández-Segura E, Baeyens JM. Urinary bladder sigma-1 receptors: A new target for cystitis treatment. Pharmacol Res 2020; 155:104724. [PMID: 32105755 DOI: 10.1016/j.phrs.2020.104724] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 02/12/2020] [Accepted: 02/23/2020] [Indexed: 02/06/2023]
Abstract
No adequate treatment is available for painful urinary bladder disorders such as interstitial cystitis/bladder pain syndrome, and the identification of new urological therapeutic targets is an unmet need. The sigma-1 receptor (σ1-R) modulates somatic pain, but its role in painful urological disorders is unexplored. The urothelium expresses many receptors typical of primary sensory neurons (e.g. TRPV1, TRPA1 and P2X3) and high levels of σ1-R have been found in these neurons; we therefore hypothesized that σ1-R may also be expressed in the urothelium and may have functional relevance in this tissue. With western blotting and immunohistochemical methods, we detected σ1-R in the urinary bladder in wild-type (WT) but not in σ1-R-knockout (σ1-KO) mice. Interestingly, σ1-R was located in the bladder urothelium not only in mouse, but also in human bladder sections. The severity of histopathological (edema, hemorrhage and urothelial desquamation) and biochemical alterations (enhanced myeloperoxidase activity and phosphorylation of extracellular regulated kinases 1/2 [pERK1/2]) that characterize cyclophosphamide-induced cystitis was lower in σ1-KO than in WT mice. Moreover, cyclophosphamide-induced pain behaviors and referred mechanical hyperalgesia were dose-dependently reduced by σ1-R antagonists (BD-1063, NE-100 and S1RA) in WT but not in σ1-KO mice. In contrast, the analgesic effect of morphine was greater in σ1-KO than in WT mice. Together these findings suggest that σ1-R plays a functional role in the mechanisms underlying cyclophosphamide-induced cystitis, and modulates morphine analgesia against urological pain. Therefore, σ1-R may represent a new drug target for urinary bladder disorders.
Collapse
Affiliation(s)
- Rafael González-Cano
- Department of Pharmacology, Faculty of Medicine, University of Granada, Granada, 18016, Spain; Anesthesia Department and Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA; Institute of Neuroscience, Biomedical Research Center, University of Granada, Armilla (Granada), 18100, Spain; Instituto de Investigación Biosanitaria, Ibs Granada, Spain
| | - Antonia Artacho-Cordón
- Department of Pharmacology, Faculty of Medicine, University of Granada, Granada, 18016, Spain; Institute of Neuroscience, Biomedical Research Center, University of Granada, Armilla (Granada), 18100, Spain
| | - Lucía Romero
- Department of Pharmacology, Faculty of Medicine, University of Granada, Granada, 18016, Spain; Institute of Neuroscience, Biomedical Research Center, University of Granada, Armilla (Granada), 18100, Spain
| | - Miguel A Tejada
- Department of Pharmacology, Faculty of Medicine, University of Granada, Granada, 18016, Spain; Institute of Neuroscience, Biomedical Research Center, University of Granada, Armilla (Granada), 18100, Spain
| | - Francisco R Nieto
- Department of Pharmacology, Faculty of Medicine, University of Granada, Granada, 18016, Spain; Institute of Neuroscience, Biomedical Research Center, University of Granada, Armilla (Granada), 18100, Spain; Instituto de Investigación Biosanitaria, Ibs Granada, Spain
| | - Manuel Merlos
- Drug Discovery and Preclinical Development, Esteve Pharmaceuticals SA, Barcelona, 08028, Spain
| | - Francisco J Cañizares
- Institute of Neuroscience, Biomedical Research Center, University of Granada, Armilla (Granada), 18100, Spain; Instituto de Investigación Biosanitaria, Ibs Granada, Spain; Department of Histology, Faculty of Medicine, University of Granada, Granada, 18016, Spain
| | - Cruz M Cendán
- Department of Pharmacology, Faculty of Medicine, University of Granada, Granada, 18016, Spain; Institute of Neuroscience, Biomedical Research Center, University of Granada, Armilla (Granada), 18100, Spain; Instituto de Investigación Biosanitaria, Ibs Granada, Spain
| | - Eduardo Fernández-Segura
- Institute of Neuroscience, Biomedical Research Center, University of Granada, Armilla (Granada), 18100, Spain; Instituto de Investigación Biosanitaria, Ibs Granada, Spain; Department of Histology, Faculty of Medicine, University of Granada, Granada, 18016, Spain
| | - José M Baeyens
- Department of Pharmacology, Faculty of Medicine, University of Granada, Granada, 18016, Spain; Institute of Neuroscience, Biomedical Research Center, University of Granada, Armilla (Granada), 18100, Spain; Instituto de Investigación Biosanitaria, Ibs Granada, Spain.
| |
Collapse
|
31
|
Linciano P, Rossino G, Listro R, Rossi D, Collina S. Sigma-1 receptor antagonists: promising players in fighting neuropathic pain. Pharm Pat Anal 2020; 9:77-85. [PMID: 32539668 DOI: 10.4155/ppa-2020-0007] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Sigma-1 receptors (S1Rs) are strongly correlated to neuropathic pain (NP), since their inactivation may decrease allodynia or dysesthesia, promoting analgesic effects. In the recent patent landscape, S1R antagonists endowed with nanomolar S1Rs affinity emerged as potent antinociceptive agents. So far, three patented compounds have been proposed for counteracting NP. Particularly PV-752 and AV1066, disclosed by the University of Pavia (Italy) and Anavex, respectively, showed good analgesic activity in preclinical studies. Moreover, E-52862 developed by Esteve (Spain) has been proved to be effective, both in preclinical and Phase II clinical trials, against several symptoms of NP. These patents ascertain S1R antagonists as potential drugs, alone or in combination with other analgesic drugs, for managing NP in humans.
Collapse
Affiliation(s)
- Pasquale Linciano
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100, Pavia, Italy
| | - Giacomo Rossino
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100, Pavia, Italy
| | - Roberta Listro
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100, Pavia, Italy
| | - Daniela Rossi
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100, Pavia, Italy
| | - Simona Collina
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100, Pavia, Italy
| |
Collapse
|
32
|
Turnaturi R, Pasquinucci L, Chiechio S, Grasso M, Marrazzo A, Amata E, Dichiara M, Prezzavento O, Parenti C. Exploiting the Power of Stereochemistry in Drug Action: 3-[(2 S,6 S,11 S)-8-Hydroxy-6,11-dimethyl-1,4,5,6-tetrahydro-2,6-methano-3-benzazocin-3(2 H)-yl]- N-phenylpropanamide as Potent Sigma-1 Receptor Antagonist. ACS Chem Neurosci 2020; 11:999-1005. [PMID: 32186844 DOI: 10.1021/acschemneuro.9b00688] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
(+)-(2S,6S,11S)- and (-)-(2R,6R,11R)-Benzomorphan derivatives have a different binding affinity for sigma-1 (σ1R) and opioid receptors, respectively. In this study, we describe the synthesis of the (+)-enantiomer [(+)-LP1] of the benzomorphan MOR agonist/DOR antagonist LP1 [(-)-LP1]. The binding affinity of both (+)-LP1 and (-)-LP1 for σ1R and sigma-2 receptor (σ2R) was tested. Moreover, (+)-LP1 opioid receptor binding affinity was also investigated. Finally, (+)-LP1 was tested in a mouse model of inflammatory pain. Our results showed a nanomolar σ1R and binding affinity for (+)-LP1. Both (+)-LP1 and (-)-LP1 elicited a significant analgesic effect in a formalin test. Differently from (-)-LP1, the analgesic effect of (+)-LP1 was not reversed by naloxone, suggesting a σ1R antagonist profile. Furthermore, σ1R agonist PRE-084 was able to unmask the σ1R antagonistic component of the benzomorphan compound. (+)-LP1 could constitute an useful lead compound to develop new analgesics based on mechanisms of action alternative to opioid receptor activation.
Collapse
MESH Headings
- Analgesics/chemistry
- Analgesics/pharmacology
- Analgesics, Opioid/pharmacology
- Animals
- Benzomorphans/chemical synthesis
- Benzomorphans/pharmacology
- Disease Models, Animal
- Mice
- Pain/drug therapy
- Receptors, Opioid/drug effects
- Receptors, Opioid/metabolism
- Receptors, Opioid, delta/drug effects
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, mu/drug effects
- Receptors, Opioid, mu/metabolism
- Receptors, sigma/antagonists & inhibitors
- Structure-Activity Relationship
- Sigma-1 Receptor
Collapse
Affiliation(s)
- Rita Turnaturi
- Department of Drug Sciences, Medicinal Chemistry Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Lorella Pasquinucci
- Department of Drug Sciences, Medicinal Chemistry Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Santina Chiechio
- Department of Drug Sciences, Pharmacology and Toxicology Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
- Oasi Research Institute-IRCCS, Troina 94018, Italy
| | - Margherita Grasso
- Department of Drug Sciences, Pharmacology and Toxicology Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
- Oasi Research Institute-IRCCS, Troina 94018, Italy
| | - Agostino Marrazzo
- Department of Drug Sciences, Medicinal Chemistry Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Emanuele Amata
- Department of Drug Sciences, Medicinal Chemistry Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Maria Dichiara
- Department of Drug Sciences, Medicinal Chemistry Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Orazio Prezzavento
- Department of Drug Sciences, Medicinal Chemistry Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Carmela Parenti
- Department of Drug Sciences, Pharmacology and Toxicology Section, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| |
Collapse
|
33
|
Bravo-Caparrós I, Ruiz-Cantero MC, Perazzoli G, Cronin SJF, Vela JM, Hamed MF, Penninger JM, Baeyens JM, Cobos EJ, Nieto FR. Sigma-1 receptors control neuropathic pain and macrophage infiltration into the dorsal root ganglion after peripheral nerve injury. FASEB J 2020; 34:5951-5966. [PMID: 32157739 DOI: 10.1096/fj.201901921r] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 02/17/2020] [Accepted: 02/27/2020] [Indexed: 12/14/2022]
Abstract
Neuron-immune interaction in the dorsal root ganglia (DRG) plays a pivotal role in the neuropathic pain development after nerve injury. Sigma-1 receptor (Sig-1R) is expressed by DRG neurons but its role in neuropathic pain is not fully understood. We investigated the effect of peripheral Sig-1R on neuroinflammation in the DRG after spared (sciatic) nerve injury (SNI) in mice. Nerve injury induced a decrease in NeuN staining along with the nuclear eccentricity and ATF3 expression in the injured DRG. Sig-1R was present in all DRG neurons examined, and after SNI this receptor translocated to the periphery of the soma and the vicinity of the nucleus, especially in injured ATF3 + neurons. In WT mice, injured DRG produced the chemokine CCL2, and this was followed by massive infiltration of macrophages/monocytes, which clustered mainly around sensory neurons with translocated Sig-1R, accompanied by robust IL-6 increase and mechanical allodynia. In contrast, Sig-1R knockout (Sig-1R-KO) mice showed reduced levels of CCL2, decreased macrophage/monocyte infiltration into DRG, and less IL-6 and neuropathic mechanical allodynia after SNI. Our findings point to an important role of peripheral Sig-1R in sensory neuron-macrophage/monocyte communication in the DRG after peripheral nerve injury; thus, these receptors may contribute to the neuropathic pain phenotype.
Collapse
Affiliation(s)
- Inmaculada Bravo-Caparrós
- Department of Pharmacology, School of Medicine, University of Granada, Granada, Spain
- Institute of Neuroscience, Biomedical Research Center, University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria IBS. GRANADA, Granada, Spain
| | - M Carmen Ruiz-Cantero
- Department of Pharmacology, School of Medicine, University of Granada, Granada, Spain
- Institute of Neuroscience, Biomedical Research Center, University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria IBS. GRANADA, Granada, Spain
| | - Gloria Perazzoli
- Instituto de Investigación Biosanitaria IBS. GRANADA, Granada, Spain
- Department of Human Anatomy and Embryology, School of Medicine, University of Granada, Granada, Spain
| | | | - José M Vela
- Drug Discovery and Preclinical Development, Esteve, Barcelona, Spain
| | - Mohamed F Hamed
- Department of Pathology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Josef M Penninger
- Institute of Molecular Biotechnology, Vienna, Austria
- Department of Medical Genetics, Life Science Institute, University of British Columbia, Vancouver, Canada
| | - José M Baeyens
- Department of Pharmacology, School of Medicine, University of Granada, Granada, Spain
- Institute of Neuroscience, Biomedical Research Center, University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria IBS. GRANADA, Granada, Spain
| | - Enrique J Cobos
- Department of Pharmacology, School of Medicine, University of Granada, Granada, Spain
- Institute of Neuroscience, Biomedical Research Center, University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria IBS. GRANADA, Granada, Spain
- Teófilo Hernando Institute for Drug Discovery, Madrid, Spain
| | - Francisco R Nieto
- Department of Pharmacology, School of Medicine, University of Granada, Granada, Spain
- Institute of Neuroscience, Biomedical Research Center, University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria IBS. GRANADA, Granada, Spain
| |
Collapse
|
34
|
Pigatto GR, Quinteiro MH, Nunes‐de‐Souza RL, Coimbra NC, Parizotto NA. Low‐Intensity Photobiomodulation Decreases Neuropathic Pain in Paw Ischemia‐Reperfusion and Spared Nervus Ischiadicus Injury Experimental Models. Pain Pract 2020; 20:371-386. [DOI: 10.1111/papr.12862] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 11/14/2019] [Accepted: 11/25/2019] [Indexed: 12/27/2022]
Affiliation(s)
- Glauce R. Pigatto
- Laboratory of Regenerative Medicine University of Araraquara (UNIARA) Araraquara Brazil
| | - Maiara H.S. Quinteiro
- Laboratory of Regenerative Medicine University of Araraquara (UNIARA) Araraquara Brazil
| | - Ricardo L. Nunes‐de‐Souza
- Laboratory of Neuropsychopharmacology School of Pharmaceutical Sciences São Paulo State University (UNESP) Araraquara Brazil
| | - Norberto C. Coimbra
- Laboratory of Neuroanatomy & Neuropsychobiology Department of Pharmacology Ribeirão Preto Medical School of the University of São Paulo (FMRP‐USP) Ribeirão Preto Brazil
| | - Nivaldo A. Parizotto
- Laboratory of Regenerative Medicine University of Araraquara (UNIARA) Araraquara Brazil
- Department of Physical Therapy Federal University of São Carlos (UFSCar) São Carlos Brazil
- Biomedical Engineering Program University of Brasil (UNIBRASIL) São Paulo Brazil
| |
Collapse
|
35
|
Zhai M, Liu C, Li Y, Zhang P, Yu Z, Zhu H, Zhang L, Zhang Q, Wang J, Wang J. Dexmedetomidine inhibits neuronal apoptosis by inducing Sigma-1 receptor signaling in cerebral ischemia-reperfusion injury. Aging (Albany NY) 2019; 11:9556-9568. [PMID: 31682592 PMCID: PMC6874446 DOI: 10.18632/aging.102404] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 10/26/2019] [Indexed: 12/17/2022]
Abstract
Dexmedetomidine is known to alleviate cerebral ischemia-reperfusion injury (CIRI). We established a rat model of CIRI, which exhibited higher neurological deficit scores and a greater number of apoptotic cells in the cerebral ischemic penumbra than controls. Dexmedetomidine reversed the neuronal apoptosis and improved neurological function in this model. We then examined Sigma-1 receptor (Sig-1R) expression on the endoplasmic reticulum (ER) in brain tissues at different reperfusion time points. Sig-1R expression increased with CIRI and decreased with increasing reperfusion times. After 24 hours of reperfusion, dexmedetomidine upregulated Sig-1R expression, and ER stress proteins (GRP78, CHOP, JNK and Caspase-3) were detected in brain tissues with Western blotting. Moreover, GRP78 expression followed a pattern similar to Sig-1R. Dexmedetomidine induced GRP78 expression but inhibited CHOP, Caspase-3 and phosphorylated-JNK expression in brain tissues. A Sig-1R-specific inhibitor reduced GRP78 expression and partially inhibited the upregulation of GRP78 by dexmedetomidine. The inhibitor also increased CHOP and Caspase-3 expression and partially reversed the inhibitory effects of dexmedetomidine on these pro-apoptotic ER stress proteins. These results suggest that dexmedetomidine at least partially inhibits ER stress-induced apoptosis by activating Sig-1R, thereby attenuating brain damage after 24 hours of ischemia-reperfusion.
Collapse
Affiliation(s)
- Meili Zhai
- Department of Anesthesiology, Tianjin Central Hospital of Gynecology Obstetrics, Gynecology Obstetrics Hospital of Nankai University, Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin 300052, China
| | - Chong Liu
- Department of Anesthesiology, Central Laboratory, Tianjin 4th Centre Hospital, The Fourth Central Hospital Affiliated to Nankai University, Tianjin 300140, China
| | - Yuexiang Li
- Department of Anesthesiology, Tianjin Xiqing Hospital, Tianjin 300380, China
| | - Peijun Zhang
- Department of Anesthesiology, Tianjin Central Hospital of Gynecology Obstetrics, Gynecology Obstetrics Hospital of Nankai University, Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin 300052, China
| | - Zhiqiang Yu
- Department of Anesthesiology, Tianjin Central Hospital of Gynecology Obstetrics, Gynecology Obstetrics Hospital of Nankai University, Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin 300052, China
| | - He Zhu
- Department of Anesthesiology, Tianjin Central Hospital of Gynecology Obstetrics, Gynecology Obstetrics Hospital of Nankai University, Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin 300052, China
| | - Li Zhang
- Department of Anesthesiology, Tianjin Central Hospital of Gynecology Obstetrics, Gynecology Obstetrics Hospital of Nankai University, Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin 300052, China
| | - Qian Zhang
- Department of Anesthesiology, Tianjin Central Hospital of Gynecology Obstetrics, Gynecology Obstetrics Hospital of Nankai University, Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin 300052, China
| | - Jianbo Wang
- Department of Anesthesiology, Tianjin Central Hospital of Gynecology Obstetrics, Gynecology Obstetrics Hospital of Nankai University, Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin 300052, China
| | - Jinhua Wang
- Department of Neurology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang Province 318000, China
| |
Collapse
|