1
|
Reis AS, Paltian JJ, Domingues WB, Novo DLR, Bolea-Fernandez E, Van Acker T, Campos VF, Luchese C, Vanhaecke F, Mesko MF, Wilhelm EA. Platinum Deposition in the Central Nervous System: A Novel Insight into Oxaliplatin-induced Peripheral Neuropathy in Young and Old Mice. Mol Neurobiol 2025; 62:3712-3729. [PMID: 39320565 DOI: 10.1007/s12035-024-04430-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 08/08/2024] [Indexed: 09/26/2024]
Abstract
Numerous factors can contribute to the incidence or exacerbation of peripheral neuropathy induced by oxaliplatin (OXA). Recently, platinum accumulation in the spinal cord of mice after OXA exposure, despite the efficient defenses of the central nervous system, has been demonstrated by our research group, expanding the knowledge about its toxicity. One hypothesis is platinum accumulation in the spinal cord causes oxidative damage to neurons and impairs mitochondrial function. Thus, the main aim of this study was to investigate the relationship between aging and OXA-induced neuropathic pain and its comorbidities, including anxious behavior and cognitive impairment. By using an OXA-induced peripheral neuropathy model, platinum and bioelement concentrations and their influence on oxidative damage, neuroprotection, and neuroplasticity pathways were evaluated in Swiss mice, and our findings showed that treatment with OXA exacerbated pain and anxious behavior, albeit not age-induced cognitive impairment. Platinum deposition in the spinal cord and, for the first time, in the brain of mice exposed to OXA, regardless of age, was identified. We found that alterations in bioelement concentration, oxidative damage, neuroprotection, and neuroplasticity pathways induced by aging contribute to OXA-induced peripheral neuropathy. Our results strive to supply a basis for therapeutic interventions for OXA-induced peripheral neuropathy considering age specificities.
Collapse
Affiliation(s)
- Angélica S Reis
- Programa de Pós-Graduação em Bioquímica e Bioprospecção (PPGBBio), Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas (UFPel), Campus Capão Do Leão, Pelotas, RS, CEP 96010-900, Brazil
| | - Jaini J Paltian
- Programa de Pós-Graduação em Bioquímica e Bioprospecção (PPGBBio), Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas (UFPel), Campus Capão Do Leão, Pelotas, RS, CEP 96010-900, Brazil
| | - William B Domingues
- Programa de Pós-Graduação em Biotecnologia, Laboratório de Genômica Estrutural, Biotecnologia - Universidade Federal de Pelotas, UFPel -, Pelotas, RS, CEP - 96010-900, Brazil
| | - Diogo L R Novo
- Programa de Pós-Graduação em Química, Laboratório de Controle de Contaminantes em Biomateriais, CCQFA - Universidade Federal de Pelotas, UFPel -, Pelotas, RS, CEP - 96010-900, Brazil
| | - Eduardo Bolea-Fernandez
- Department of Chemistry, Atomic & Mass Spectrometry - A&MS Research Unit, Ghent University, Campus Sterre, Krijgslaan 281-S12, 9000, Ghent, Belgium
| | - Thibaut Van Acker
- Department of Chemistry, Atomic & Mass Spectrometry - A&MS Research Unit, Ghent University, Campus Sterre, Krijgslaan 281-S12, 9000, Ghent, Belgium
| | - Vinicius F Campos
- Programa de Pós-Graduação em Biotecnologia, Laboratório de Genômica Estrutural, Biotecnologia - Universidade Federal de Pelotas, UFPel -, Pelotas, RS, CEP - 96010-900, Brazil
| | - Cristiane Luchese
- Programa de Pós-Graduação em Bioquímica e Bioprospecção (PPGBBio), Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas (UFPel), Campus Capão Do Leão, Pelotas, RS, CEP 96010-900, Brazil
| | - Frank Vanhaecke
- Department of Chemistry, Atomic & Mass Spectrometry - A&MS Research Unit, Ghent University, Campus Sterre, Krijgslaan 281-S12, 9000, Ghent, Belgium
| | - Marcia F Mesko
- Programa de Pós-Graduação em Química, Laboratório de Controle de Contaminantes em Biomateriais, CCQFA - Universidade Federal de Pelotas, UFPel -, Pelotas, RS, CEP - 96010-900, Brazil.
| | - Ethel A Wilhelm
- Programa de Pós-Graduação em Bioquímica e Bioprospecção (PPGBBio), Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas (UFPel), Campus Capão Do Leão, Pelotas, RS, CEP 96010-900, Brazil.
| |
Collapse
|
2
|
Takasaki I, Nagashima R, Ueda T, Ashihara Y, Nakamachi T, Okada T, Toyooka N, Miyata A, Kurihara T. Spinal pituitary adenylate cyclase-activating polypeptide and PAC1 receptor signaling system is involved in the oxaliplatin-induced acute cold allodynia in mice. THE JOURNAL OF PAIN 2025; 27:104751. [PMID: 39615811 DOI: 10.1016/j.jpain.2024.104751] [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: 06/28/2024] [Revised: 11/04/2024] [Accepted: 11/25/2024] [Indexed: 02/10/2025]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a type of peripheral neuropathy that develops in patients treated with certain anticancer drugs. Oxaliplatin (OXA) causes CIPN in approximately 80-90 % of patients; thus, it is necessary to elucidate its underlying mechanism and develop effective treatments and prevention methods. The purpose of this study was to determine whether the pituitary adenylate cyclase-activating polypeptide (PACAP)/PAC1 receptor system in the spinal dorsal horn is involved in OXA-induced acute cold allodynia and examine the effect of a PAC1 receptor antagonist. Administration of OXA induced acute cold allodynia in wild-type mice, but not in PACAP-/- mice. In the dorsal root ganglia, OXA upregulated PACAP expression, particularly in small-sized neurons. OXA-induced cold allodynia was ameliorated by intrathecal (i.t.) injection of PACAP6-38 (peptide antagonist for PACAP receptor) and PA-8 (small-molecule antagonist specific for PAC1 receptor). I.t. PACAP, but not vasoactive intestinal polypeptide, resulted in cold allodynia, which was blocked by PA-8. OXA induced the activation of spinal astrocytes in a PAC1 receptor-dependent manner. The results suggest that spinal PACAP/PAC1 receptor systems are involved in OXA-induced acute cold allodynia through astrocyte activation. Furthermore, we demonstrated that the systemic administration of PA-8 resulted in therapeutic and preventative effects on OXA-induced acute cold allodynia. Because PA-8 did not affect the anticancer effects of OXA, we propose PAC1 receptor inhibition as a new strategy for the treatment and prevention of CIPN. PERSPECTIVE: Cold allodynia is a hallmark of OXA-induced peripheral neuropathy. This study demonstrated the involvement of spinal PACAP/PAC1 receptors in OXA-induced acute cold allodynia. We propose PAC1 receptor inhibition as a new strategy for the treatment and prevention of OXA-induced acute cold allodynia.
Collapse
Affiliation(s)
- Ichiro Takasaki
- Laboratory of Pharmacology, Faculty of Engineering, University of Toyama, Toyama 930-8555, Japan; Graduate School of Pharma-Medical Sciences, University of Toyama, Toyama 930-8555, Japan; Graduate School of Innovative Life Science, University of Toyama, Toyama 930-8555, Japan.
| | - Ryota Nagashima
- Laboratory of Pharmacology, Faculty of Engineering, University of Toyama, Toyama 930-8555, Japan
| | - Takahiro Ueda
- Laboratory of Pharmacology, Faculty of Engineering, University of Toyama, Toyama 930-8555, Japan
| | - Yuya Ashihara
- Laboratory of Pharmacology, Faculty of Engineering, University of Toyama, Toyama 930-8555, Japan; Graduate School of Pharma-Medical Sciences, University of Toyama, Toyama 930-8555, Japan
| | - Tomoya Nakamachi
- Laboratory of Regulatory Biology, Faculty of Science, University of Toyama, Toyama 930-8555, Japan
| | - Takuya Okada
- Graduate School of Pharma-Medical Sciences, University of Toyama, Toyama 930-8555, Japan; Graduate School of Innovative Life Science, University of Toyama, Toyama 930-8555, Japan; Laboratory of Bio-functional Molecular Engineering, Faculty of Engineering, University of Toyama, Toyama 930-8555, Japan
| | - Naoki Toyooka
- Graduate School of Pharma-Medical Sciences, University of Toyama, Toyama 930-8555, Japan; Graduate School of Innovative Life Science, University of Toyama, Toyama 930-8555, Japan; Laboratory of Bio-functional Molecular Engineering, Faculty of Engineering, University of Toyama, Toyama 930-8555, Japan
| | - Atsuro Miyata
- Department of Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan
| | - Takashi Kurihara
- Department of Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan
| |
Collapse
|
3
|
Mishra S, Mishra Y, Kumar A. Marine-derived bioactive compounds for neuropathic pain: pharmacology and therapeutic potential. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-024-03667-7. [PMID: 39797987 DOI: 10.1007/s00210-024-03667-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 11/22/2024] [Indexed: 01/13/2025]
Abstract
Neuropathic pain, a challenging condition often associated with diabetes, trauma, or chemotherapy, impairs patients' quality of life. Current treatments often provide inconsistent relief and notable adverse effects, highlighting the urgent need for safer and more effective alternatives. This review investigates marine-derived bioactive compounds as potential novel therapies for neuropathic pain management. Marine organisms, including fungi, algae, cone snails, sponges, soft corals, tunicates, and fish, produce a diverse range of secondary metabolites with significant pharmacological properties. These include peptides (e.g., conopeptides, piscidin 1), non-peptides (e.g., guanidinium toxins, astaxanthin, docosahexaenoic acid, fucoidan, apigenin, fumagillin, aaptamine, flexibilide, excavatolide B, capnellenes, austrasulfones, lemnalol), and crude extracts (e.g., Spirulina platensis, Dunaliella salina, Cliothosa aurivilli). These compounds exhibit diverse mechanisms of action, such as modulating ion channels (e.g., transient receptor potential channels, voltage-gated sodium, calcium, and potassium channels, and G protein-coupled inwardly rectifying potassium channels), interacting with cell-surface receptors (e.g., nicotinic acetylcholine, NMDA, kainate, GABAB, and neurotensin receptors), inhibiting norepinephrine transporters, reducing oxidative stress, and attenuating neuroinflammation. These effects collectively contribute to alleviating nerve degeneration and symptoms of neuropathic pain, including hyperalgesia, allodynia, and associated psychomotor disturbances. Marine-derived bioactive compounds represent promising alternatives to conventional neuropathic pain treatments, to advance their development and assess their integration into neuropathic pain management strategies.
Collapse
Affiliation(s)
- Swapnil Mishra
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, West Bengal, India
| | - Yogesh Mishra
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Punjab, India
| | - Ashutosh Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Punjab, India.
| |
Collapse
|
4
|
Tae HS, Hung A, Clark RJ, Adams DJ. Molecular determinants of the selectivity and potency of α-conotoxin Vc1.1 for human nicotinic acetylcholine receptors. J Biol Chem 2025; 301:108017. [PMID: 39608712 PMCID: PMC11732461 DOI: 10.1016/j.jbc.2024.108017] [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: 09/03/2024] [Revised: 11/02/2024] [Accepted: 11/17/2024] [Indexed: 11/30/2024] Open
Abstract
The α-conotoxins (α-Ctxs) are short, disulfide-rich peptides derived from the venom of the Conus marine snails, primarily acting as antagonists of nicotinic acetylcholine receptors (nAChRs). Specifically, α-Ctx Vc1.1, a 16-amino acid peptide from Conus victoriae, competitively antagonizes non-muscle nAChRs, inhibits nicotine-induced currents in bovine chromaffin cells, and alleviates neuropathic pain in rat models. Although Vc1.1 selectively inhibits rat α9α10 nAChRs, its potency and selectivity across human nAChR subtypes remain unresolved. In this study, we assessed the activity of Vc1.1 on human (h) nAChRs heterologously expressed in Xenopus laevis oocytes using the two-electrode voltage clamp technique and simulated interactions using computational modeling. Vc1.1 selectively antagonized homomeric α9 and heteromeric α3β2 nAChRs, with half-maximal inhibitory concentrations (IC50) of 160 nM and 232 nM, respectively. At hα9[N179A]α10, Vc1.1 exhibited a 20-fold decrease in potency compared to hα9α10, due to the loss of hydrogen bonding with Vc1.1-D11. Conversely, Vc1.1 was four-fold more potent at hα3β2[E86A] compared to hα3β2, possibly influenced by the proximal residue β2-K104, as suggested by molecular dynamics (MD) simulations. Additionally, Vc1.1's potency doubled at hα9[N213K]α10, whereas it remained unchanged at hα9[N213R]α10 nAChRs. MD simulations indicate that altered interactions between the mutant hα9 N179A, N213K, and N213R side chains and Vc1.1-D5 may partly explain these changes in potency. The inhibitory action of Vc1.1 at α9-containing nAChRs is particularly relevant given their role in neuroinflammation, presenting a potential therapeutic pathway for alleviating neuropathic and inflammatory pain. This study provides valuable insights into the rational design of Vc1.1-derived α-Ctxs with enhanced nAChR subtype selectivity.
Collapse
Affiliation(s)
- Han-Shen Tae
- Molecular Horizons, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, Australia
| | - Andrew Hung
- School of Science, RMIT University, Melbourne, Australia.
| | - Richard J Clark
- School of Biomedical Sciences, The University of Queensland, Brisbane, Australia
| | - David J Adams
- Molecular Horizons, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, Australia.
| |
Collapse
|
5
|
Azam L, Christensen SB, Riaz Z, Kendell A, Cull J, Hone AJ, McIntosh JM. α9-Containing Nicotinic Acetylcholine Receptors Are Required for RgIA-5474 Attenuation of Chemotherapy-Induced Neuropathic Pain. ACS Pharmacol Transl Sci 2024; 7:3935-3944. [PMID: 39698293 PMCID: PMC11651205 DOI: 10.1021/acsptsci.4c00454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 10/22/2024] [Accepted: 11/04/2024] [Indexed: 12/20/2024]
Abstract
Nicotinic acetylcholine receptors containing the α9 subunit have been mechanistically implicated in alleviating chemotherapy-induced neuropathic pain. However, the cell types that underlie these effects are currently unknown. RgIA-5474 is a recently developed, synthetic α-conotoxin analog that is a potent antagonist of human α9α10 nAChRs. We used germline α9 subunit knockout mice, CD3+ T-cell depletion, and conditional knockdown of the α9 subunit in immune cells to examine the role of α9-containing nAChRs that mediate RgIA-5474 alleviation of oxaliplatin-induced neuropathic pain. RgIA-5474 potently and selectively blocked mouse α9α10 nAChRs. A one-time oxaliplatin injection resulted in cold allodynia that was reversed by RgIA-5474 administration in the wild type but not in α9 germline knockout mice. RgIA-5474 also failed to produce analgesia in CD3+ T-cell-depleted male and female animals. Conditional knockdown of the α9 subunit in immune cells of mice by the CreloxP system also eliminated the therapeutic effects of RgIA-5474 in both male and female mice. These results indicate that the α9 nAChR subunit is necessary for the analgesic effects of RgIA-5474 and implicate α9-containing nAChRs in immune cells as a nonopioid target for treating neuropathic pain.
Collapse
Affiliation(s)
- Layla Azam
- School
of Biological Sciences, University of Utah, Salt Lake City, Utah 84112, United States
| | - Sean B. Christensen
- School
of Biological Sciences, University of Utah, Salt Lake City, Utah 84112, United States
| | - Zoha Riaz
- School
of Biological Sciences, University of Utah, Salt Lake City, Utah 84112, United States
| | - Anne Kendell
- School
of Biological Sciences, University of Utah, Salt Lake City, Utah 84112, United States
| | - Jennison Cull
- School
of Biological Sciences, University of Utah, Salt Lake City, Utah 84112, United States
| | - Arik J. Hone
- School
of Biological Sciences, University of Utah, Salt Lake City, Utah 84112, United States
- MIRECC,
George E. Whalen Veterans Affair Medical Center, Salt Lake City, Utah 84148, United States
| | - J. Michael McIntosh
- School
of Biological Sciences, University of Utah, Salt Lake City, Utah 84112, United States
- Psychiatry, University of Utah, Salt Lake City, Utah 84108, United States
- George
E. Whalen Veterans Affair Medical Center, Salt Lake City, Utah 84148, United States
| |
Collapse
|
6
|
Cunha M, Tavares I, Costa-Pereira JT. Centralizing the Knowledge and Interpretation of Pain in Chemotherapy-Induced Peripheral Neuropathy: A Paradigm Shift towards Brain-Centric Approaches. Brain Sci 2024; 14:659. [PMID: 39061400 PMCID: PMC11274822 DOI: 10.3390/brainsci14070659] [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: 05/14/2024] [Revised: 06/17/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024] Open
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a side effect of cancer treatment, often linked with pain complaints. Patients report mechanical and thermal hypersensitivity that may emerge during chemotherapy treatment and may persist after cancer remission. Whereas the latter situation disturbs the quality of life, life itself may be endangered by the appearance of CIPN during cancer treatment. The causes of CIPN have almost entirely been ascribed to the neurotoxicity of chemotherapeutic drugs in the peripheral nervous system. However, the central consequences of peripheral neuropathy are starting to be unraveled, namely in the supraspinal pain modulatory system. Based on our interests and experience in the field, we undertook a review of the brain-centered alterations that may underpin pain in CIPN. The changes in the descending pain modulation in CIPN models along with the functional and connectivity abnormalities in the brain of CIPN patients are analyzed. A translational analysis of preclinical findings about descending pain regulation during CIPN is reviewed considering the main neurochemical systems (serotoninergic and noradrenergic) targeted in CIPN management in patients, namely by antidepressants. In conclusion, this review highlights the importance of studying supraspinal areas involved in descending pain modulation to understand the pathophysiology of CIPN, which will probably allow a more personalized and effective CIPN treatment in the future.
Collapse
Affiliation(s)
- Mário Cunha
- Department of Biomedicine, Unit of Experimental Biology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; (M.C.); (J.T.C.-P.)
| | - Isaura Tavares
- Department of Biomedicine, Unit of Experimental Biology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; (M.C.); (J.T.C.-P.)
- I3S—Institute of Investigation and Innovation in Health, University of Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - José Tiago Costa-Pereira
- Department of Biomedicine, Unit of Experimental Biology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; (M.C.); (J.T.C.-P.)
- I3S—Institute of Investigation and Innovation in Health, University of Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Faculty of Nutrition and Food Sciences, University of Porto, Rua do Campo Alegre 823, 4150-180 Porto, Portugal
| |
Collapse
|
7
|
Andleeb H, Papke RL, Stokes C, Richter K, Herz SM, Chiang K, Kanumuri SRR, Sharma A, Damaj MI, Grau V, Horenstein NA, Thakur GA. Explorations of Agonist Selectivity for the α9* nAChR with Novel Substituted Carbamoyl/Amido/Heteroaryl Dialkylpiperazinium Salts and Their Therapeutic Implications in Pain and Inflammation. J Med Chem 2024; 67:8642-8666. [PMID: 38748608 PMCID: PMC11181317 DOI: 10.1021/acs.jmedchem.3c02429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 04/01/2024] [Accepted: 04/29/2024] [Indexed: 06/14/2024]
Abstract
There is an urgent need for nonopioid treatments for chronic and neuropathic pain to provide effective alternatives amid the escalating opioid crisis. This study introduces novel compounds targeting the α9 nicotinic acetylcholine receptor (nAChR) subunit, which is crucial for pain regulation, inflammation, and inner ear functions. Specifically, it identifies novel substituted carbamoyl/amido/heteroaryl dialkylpiperazinium iodides as potent agonists selective for human α9 and α9α10 over α7 nAChRs, particularly compounds 3f, 3h, and 3j. Compound 3h (GAT2711) demonstrated a 230 nM potency as a full agonist at α9 nAChRs, being 340-fold selective over α7. Compound 3c was 10-fold selective for α9α10 over α9 nAChR. Compounds 2, 3f, and 3h inhibited ATP-induced interleukin-1β release in THP-1 cells. The analgesic activity of 3h was fully retained in α7 knockout mice, suggesting that analgesic effects were potentially mediated through α9* nAChRs. Our findings provide a blueprint for developing α9*-specific therapeutics for pain.
Collapse
Affiliation(s)
- Hina Andleeb
- Department
of Chemistry, University of Florida, P.O. Box 117200, Gainesville, Florida 32611-7200, United States
- Department
of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical
Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, Massachusetts 02115, United States
| | - Roger L. Papke
- Department
of Pharmacology and Therapeutics, University
of Florida, P.O. Box 100267, Gainesville, Florida 32610, United States
| | - Clare Stokes
- Department
of Pharmacology and Therapeutics, University
of Florida, P.O. Box 100267, Gainesville, Florida 32610, United States
| | - Katrin Richter
- Department
of General and Thoracic Surgery, Laboratory of Experimental Surgery,
Justus-Liebig-University, German Center for Lung Research [DZL], Cardio-Pulmonary Institute [CPI], Giessen 35385, Germany
| | - Sara M. Herz
- Department
of Pharmacology and Toxicology, Virginia
Commonwealth University, Richmond, Virginia 23298, United States
| | - Ka Chiang
- Department
of Pharmacology and Toxicology, Virginia
Commonwealth University, Richmond, Virginia 23298, United States
| | - Siva R. Raju Kanumuri
- Department
of Pharmaceutics, University of Florida, Gainesville, Florida 32610, United States
| | - Abhisheak Sharma
- Department
of Pharmaceutics, University of Florida, Gainesville, Florida 32610, United States
| | - M. Imad Damaj
- Department
of Pharmacology and Toxicology, Virginia
Commonwealth University, Richmond, Virginia 23298, United States
| | - Veronika Grau
- Department
of General and Thoracic Surgery, Laboratory of Experimental Surgery,
Justus-Liebig-University, German Center for Lung Research [DZL], Cardio-Pulmonary Institute [CPI], Giessen 35385, Germany
| | - Nicole A. Horenstein
- Department
of Chemistry, University of Florida, P.O. Box 117200, Gainesville, Florida 32611-7200, United States
| | - Ganesh A. Thakur
- Department
of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical
Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, Massachusetts 02115, United States
| |
Collapse
|
8
|
Wang H, Li X, Qiao Y, Wang M, Wang W, McIntosh JM, Zhangsun D, Luo S. αO-Conotoxin GeXIVA[1,2] Reduced Neuropathic Pain and Changed Gene Expression in Chronic Oxaliplatin-Induced Neuropathy Mice Model. Mar Drugs 2024; 22:49. [PMID: 38276651 PMCID: PMC10821445 DOI: 10.3390/md22010049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/14/2024] [Accepted: 01/17/2024] [Indexed: 01/27/2024] Open
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a dose-limiting painful neuropathy that occurs commonly during cancer management, which often leads to the discontinuation of medication. Previous studies suggest that the α9α10 nicotinic acetylcholine receptor (nAChR)-specific antagonist αO-conotoxin GeXIVA[1,2] is effective in CIPN models; however, the related mechanisms remain unclear. Here, we analyzed the preventive effect of GeXIVA[1,2] on neuropathic pain in the long-term oxaliplatin injection-induced CIPN model. At the end of treatment, lumbar (L4-L6) spinal cord was extracted, and RNA sequencing and bioinformatic analysis were performed to investigate the potential genes and pathways related to CIPN and GeXIVA[1,2]. GeXIVA[1,2] inhibited the development of mechanical allodynia induced by chronic oxaliplatin treatment. Repeated injections of GeXIVA[1,2] for 3 weeks had no effect on the mice's normal pain threshold or locomotor activity and anxiety-like behavior, as evaluated in the open field test (OFT) and elevated plus maze (EPM). Our RNA sequencing results identified 209 differentially expressed genes (DEGs) in the CIPN model, and simultaneously injecting GeXIVA[1,2] with oxaliplatin altered 53 of the identified DEGs. These reverted genes were significantly enriched in immune-related pathways represented by the cytokine-cytokine receptor interaction pathway. Our findings suggest that GeXIVA[1,2] could be a potential therapeutic compound for chronic oxaliplatin-induced CIPN management.
Collapse
Affiliation(s)
- Huanbai Wang
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou 570228, China; (H.W.); (X.L.); (Y.Q.); (W.W.)
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China;
| | - Xiaodan Li
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou 570228, China; (H.W.); (X.L.); (Y.Q.); (W.W.)
| | - Yamin Qiao
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou 570228, China; (H.W.); (X.L.); (Y.Q.); (W.W.)
| | - Meiting Wang
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China;
| | - Wen Wang
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou 570228, China; (H.W.); (X.L.); (Y.Q.); (W.W.)
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China;
| | - J. Michael McIntosh
- Department of Biology and Department of Psychiatry, University of Utah, Salt Lake City, UT 84112, USA;
- George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, UT 84108, USA
| | - Dongting Zhangsun
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou 570228, China; (H.W.); (X.L.); (Y.Q.); (W.W.)
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China;
| | - Sulan Luo
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou 570228, China; (H.W.); (X.L.); (Y.Q.); (W.W.)
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China;
| |
Collapse
|
9
|
Richter K, Herz SM, Stokes C, Damaj MI, Grau V, Papke RL. Pharmacological profiles and anti-inflammatory activity of pCN-diEPP and mCN-diEPP, new alpha9alpha10 nicotinic receptor ligands. Neuropharmacology 2023; 240:109717. [PMID: 37758018 PMCID: PMC11295495 DOI: 10.1016/j.neuropharm.2023.109717] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/29/2023] [Accepted: 09/13/2023] [Indexed: 10/01/2023]
Abstract
Pain due to inflammation can be reduced by targeting the noncanonical nicotinic receptors (NCNR) in cells of the immune system that regulate the synthesis and release of pro- and anti-inflammatory cytokines. Although NCNR do not generate ion channel currents, the pharmacology of ion-channel forms of the receptors can predict drugs which may be effective regulators of the cholinergic anti-inflammatory system (CAS). Agonists of α7 type receptors have been definitively associated with CAS. Receptors containing α9 and α10 subunits have also been implicated. We have recently characterized two small molecules, pCN-diEPP and mCN-diEPP, as selective α9α10 agonists and antagonists, respectively. We used these drugs, along with nicotine, an α7 agonist and α9α10 antagonist, to probe the mixed populations of receptors that are formed when α7, α9, and α10 are all expressed together in Xenopus oocytes. We also evaluated the effects of the CN-diEPP compounds on regulating the ATP-induced release of interleukin-1β from monocytic THP-1 cells, which express NCNR. The compounds successfully identified separate populations of receptors when all three subunits were co-expressed, including a potential population of homomeric α10 receptors. The α9α10 agonist pCN-diEPP was the more effective regulator of interleukin-1β release in THP-1 cells. pCN-diEPP was also fully effective in a mouse model of inflammatory pain, while mCN-diEPP had only partial effects, requiring a higher dosage. The analgetic effects of pCN-diEPP and mCN-diEPP were retained in α7 knockout mice. Taken together, our results suggest that drugs that selectively activate α9α10 receptors may useful to reduce inflammatory pain through the CAS.
Collapse
Affiliation(s)
- Katrin Richter
- Department of General and Thoracic Surgery, Laboratory of Experimental Surgery, Justus-Liebig-University, German Center for Lung Research [DZL], Cardio-Pulmonary Institute [CPI], Giessen, Germany
| | - Sara M Herz
- Department of Pharmacology and Toxicology, Medical College of Virginia, Virginia Commonwealth University, USA
| | - Clare Stokes
- Department of Pharmacology and Therapeutics, University of Florida, PO Box 100267, Gainesville, FL, 32610, USA
| | - M Imad Damaj
- Department of Pharmacology and Toxicology, Medical College of Virginia, Virginia Commonwealth University, USA
| | - Veronika Grau
- Department of General and Thoracic Surgery, Laboratory of Experimental Surgery, Justus-Liebig-University, German Center for Lung Research [DZL], Cardio-Pulmonary Institute [CPI], Giessen, Germany
| | - Roger L Papke
- Department of Pharmacology and Therapeutics, University of Florida, PO Box 100267, Gainesville, FL, 32610, USA.
| |
Collapse
|
10
|
Montigné E, Balayssac D. Exploring Cholinergic Compounds for Peripheral Neuropathic Pain Management: A Comprehensive Scoping Review of Rodent Model Studies. Pharmaceuticals (Basel) 2023; 16:1363. [PMID: 37895835 PMCID: PMC10609809 DOI: 10.3390/ph16101363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/01/2023] [Accepted: 09/14/2023] [Indexed: 10/29/2023] Open
Abstract
Neuropathic pain affects about 7-8% of the population, and its management still poses challenges with unmet needs. Over the past decades, researchers have explored the cholinergic system (muscarinic and nicotinic acetylcholine receptors: mAChR and nAChR) and compounds targeting these receptors as potential analgesics for neuropathic pain management. This scoping review aims to provide an overview of studies on peripheral neuropathic pain (PNP) in rodent models, exploring compounds targeting cholinergic neurotransmission. The inclusion criteria were original articles on PNP in rodent models that explored the use of compounds directly targeting cholinergic neurotransmission and reported results of nociceptive behavioral assays. The literature search was performed in the PubMed and Web of Science databases (1 January 2000-22 April 2023). The selection process yielded 82 publications, encompassing 62 compounds. The most studied compounds were agonists of α4β2 nAChR and α7 nAChR, and antagonists of α9/α10 nAChR, along with those increasing acetylcholine and targeting mAChRs. Studies mainly reported antinociceptive effects in traumatic PNP models, and to a lesser extent, chemotherapy-induced neuropathy or diabetic models. These preclinical studies underscore the considerable potential of cholinergic compounds in the management of PNP, warranting the initiation of clinical trials.
Collapse
Affiliation(s)
- Edouard Montigné
- INSERM, U1107, NEURO-DOL, Université Clermont Auvergne, F-63000 Clermont-Ferrand, France;
| | - David Balayssac
- INSERM, U1107, NEURO-DOL, Université Clermont Auvergne, Direction de la Recherche Clinique et de l’Innovation, CHU Clermont-Ferrand, F-63000 Clermont-Ferrand, France
| |
Collapse
|
11
|
Xu P, Zhang P, Zhu X, Wu Y, Harvey PJ, Kaas Q, Zhangsun D, Craik DJ, Luo S. Structure-Activity Relationships of Alanine Scan Mutants αO-Conotoxins GeXIVA[1,2] and GeXIVA[1,4]. J Med Chem 2023. [PMID: 37464764 DOI: 10.1021/acs.jmedchem.3c01023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
αO-Conotoxin GeXIVA is a selective α9α10 nicotinic acetylcholine receptor (nAChR) inhibitor displaying two disulfide bonds that can form three isomers. The bead (GeXIVA[1,2]) and ribbon (GeXIVA[1,4]) isomers possess the highest activity on rat and human α9α10 nAChRs. However, the molecular mechanism by which they inhibit the α9α10 nAChR is unknown. Here, an alanine scan of GeXIVA was used to elucidate key interactions between the peptides and the α9α10 nAChR. The majority of GeXIVA[1,2] analogues preserved affinity at α9α10 nAChR, but [R17A]GeXIVA[1,2] enhanced selectivity on the α9α10 nAChR. The I23A replacement of GeXIVA[1,4] increased activity at both rat and human α9α10 nAChRs by 10-fold. Surprisingly, these results do not support the molecular model of an interaction in the orthosteric binding site proposed previously, but rather may involve allosteric coupling with the voltage-sensitive domain of the α9α10 nAChR. These results could help to guide further development of GeXIVA analogues as analgesics.
Collapse
Affiliation(s)
- Pan Xu
- School of Medicine, Guangxi University, Nanning 530004, China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen 518060, China
| | - Panpan Zhang
- School of Medicine, Guangxi University, Nanning 530004, China
| | - Xiaopeng Zhu
- School of Medicine, Guangxi University, Nanning 530004, China
| | - Yong Wu
- School of Medicine, Guangxi University, Nanning 530004, China
| | - Peta J Harvey
- Institute for Molecular Bioscience, Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Quentin Kaas
- Institute for Molecular Bioscience, Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Dongting Zhangsun
- School of Medicine, Guangxi University, Nanning 530004, China
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou 570228, China
| | - David J Craik
- Institute for Molecular Bioscience, Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Sulan Luo
- School of Medicine, Guangxi University, Nanning 530004, China
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou 570228, China
| |
Collapse
|
12
|
Di Cesare Mannelli L, Pacini A, Micheli L, Femia AP, Maresca M, Zanardelli M, Vannacci A, Gallo E, Bilia AR, Caderni G, Firenzuoli F, Mugelli A, Ghelardini C. Retraction Note: Astragali radix: could it be an adjuvant for oxaliplatin-induced neuropathy? Sci Rep 2023; 13:9979. [PMID: 37339997 DOI: 10.1038/s41598-023-37011-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/22/2023] Open
Affiliation(s)
- Lorenzo Di Cesare Mannelli
- Department of Neuroscience, Psychology, Drug Research and Children's Health - NEUROFARBA - Pharmacology and Toxicology Section, Florence, Italy.
| | - Alessandra Pacini
- Department of Experimental and Clinical Medicine - DECM - Section of Anatomy and Histology, University of Florence, Florence, Italy
| | - Laura Micheli
- Department of Neuroscience, Psychology, Drug Research and Children's Health - NEUROFARBA - Pharmacology and Toxicology Section, Florence, Italy
| | - Angelo Pietro Femia
- Department of Neuroscience, Psychology, Drug Research and Children's Health - NEUROFARBA - Pharmacology and Toxicology Section, Florence, Italy
| | - Mario Maresca
- Department of Neuroscience, Psychology, Drug Research and Children's Health - NEUROFARBA - Pharmacology and Toxicology Section, Florence, Italy
| | - Matteo Zanardelli
- Department of Neuroscience, Psychology, Drug Research and Children's Health - NEUROFARBA - Pharmacology and Toxicology Section, Florence, Italy
| | - Alfredo Vannacci
- Department of Neuroscience, Psychology, Drug Research and Children's Health - NEUROFARBA - Pharmacology and Toxicology Section, Florence, Italy
| | - Eugenia Gallo
- Department of Neuroscience, Psychology, Drug Research and Children's Health - NEUROFARBA - Pharmacology and Toxicology Section, Florence, Italy
| | - Anna Rita Bilia
- Department of Chemistry, University of Florence, Florence, Italy
| | - Giovanna Caderni
- Department of Neuroscience, Psychology, Drug Research and Children's Health - NEUROFARBA - Pharmacology and Toxicology Section, Florence, Italy
| | - Fabio Firenzuoli
- Department of Neuroscience, Psychology, Drug Research and Children's Health - NEUROFARBA - Pharmacology and Toxicology Section, Florence, Italy
| | - Alessandro Mugelli
- Department of Neuroscience, Psychology, Drug Research and Children's Health - NEUROFARBA - Pharmacology and Toxicology Section, Florence, Italy
| | - Carla Ghelardini
- Department of Neuroscience, Psychology, Drug Research and Children's Health - NEUROFARBA - Pharmacology and Toxicology Section, Florence, Italy
| |
Collapse
|
13
|
Tan M, Feng Z, Chen H, Min L, Wen H, Liu H, Hou J. Transcranial direct current stimulation regulates phenotypic transformation of microglia to relieve neuropathic pain induced by spinal cord injury. Front Behav Neurosci 2023; 17:1147693. [PMID: 37081929 PMCID: PMC10110883 DOI: 10.3389/fnbeh.2023.1147693] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 03/21/2023] [Indexed: 04/07/2023] Open
Abstract
ObjectiveNeuropathic pain is a common complication after spinal cord injury (SCI). Transcranial direct current stimulation (tDCS) has been confirmed to be effective in relieving neuropathic pain in patients with SCI. The aim of this study is to investigate the effect of tDCS on neuropathic pain induced by SCI and its underlying mechanism.Materials and methodsThe SCI model was induced by a clip-compression injury and tDCS stimulation was performed for two courses (5 days/each). The motor function was evaluated by Basso-Beattie-Bresnahan (BBB) score, and the thermal withdrawal threshold was evaluated by the thermal radiation method. The effects of tDCS on the cerebral cortex, thalamus, midbrain, and medulla were detected by the enzyme-linked immunosorbent assay (ELISA) and immunofluorescence.ResultsThe results showed that SCI reduced the thermal withdrawal threshold and increased the concentration of inflammatory cytokines in the cortex, thalamus, midbrain, and medulla, including the tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6). In addition, the activation of microglia and the proportion of M1 phenotypic polarization increased significantly in the ventral posterolateral (VPL), ventral tegmental (VTA), and periaqueductal gray (PAG) regions after SCI. After tDCS treatment, the thermal withdrawal threshold and motor function of SCI rats were significantly improved compared to the vehicle group. Meanwhile, tDCS effectively reduced the concentration of pro-inflammatory cytokines in the cortex, thalamus, midbrain, and medulla and increased the concentration of anti-inflammatory cytokines interleukin-10 (IL-10) in the thalamus. In addition, tDCS reduced the proportion of the M1 phenotype of microglia in VPL, VTA, and PAG regions and increase the proportion of the M2 phenotype.ConclusionThe results suggest that tDCS can effectively relieve SCI-induced neuropathic pain. Its mechanism may be related to regulating the inflammatory and anti-inflammatory cytokines in corresponding brain regions via promoting the phenotypic transformation of microglia.
Collapse
Affiliation(s)
- Mingliang Tan
- Department of Rehabilitation, Southwest Hospital, Army Medical University, Chongqing, China
| | - Zhou Feng
- Department of Rehabilitation, Southwest Hospital, Army Medical University, Chongqing, China
| | - Hui Chen
- Department of Rehabilitation, Southwest Hospital, Army Medical University, Chongqing, China
| | - Lingxia Min
- Department of Rehabilitation, Southwest Hospital, Army Medical University, Chongqing, China
| | - Huizhong Wen
- Department of Neurobiology, College of Basic Medical Science, Army Medical University, Chongqing, China
| | - Hongliang Liu
- Department of Rehabilitation, Southwest Hospital, Army Medical University, Chongqing, China
- *Correspondence: Hongliang Liu,
| | - Jingming Hou
- Department of Rehabilitation, Southwest Hospital, Army Medical University, Chongqing, China
- Jingming Hou,
| |
Collapse
|
14
|
Hone AJ, McIntosh JM. Nicotinic acetylcholine receptors: Therapeutic targets for novel ligands to treat pain and inflammation. Pharmacol Res 2023; 190:106715. [PMID: 36868367 PMCID: PMC10691827 DOI: 10.1016/j.phrs.2023.106715] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/22/2023] [Accepted: 02/28/2023] [Indexed: 03/05/2023]
Abstract
Nicotinic acetylcholine receptors (nAChRs) have been historically defined as ligand-gated ion channels and function as such in the central and peripheral nervous systems. Recently, however, non-ionic signaling mechanisms via nAChRs have been demonstrated in immune cells. Furthermore, the signaling pathways where nAChRs are expressed can be activated by endogenous ligands other than the canonical agonists acetylcholine and choline. In this review, we discuss the involvement of a subset of nAChRs containing α7, α9, and/or α10 subunits in the modulation of pain and inflammation via the cholinergic anti-inflammatory pathway. Additionally, we review the most recent advances in the development of novel ligands and their potential as therapeutics.
Collapse
Affiliation(s)
- Arik J Hone
- School of Biological Sciences University of Utah, Salt Lake City, UT, USA; MIRECC, George E. Whalen Veterans Affairs Medical Center, Salt Lake City, UT, USA.
| | - J Michael McIntosh
- School of Biological Sciences University of Utah, Salt Lake City, UT, USA; Department of Psychiatry, University of Utah, Salt Lake City, UT, USA; George E. Whalen Veterans Affairs Medical Center, Salt Lake City, UT, USA.
| |
Collapse
|
15
|
Shelukhina I, Siniavin A, Kasheverov I, Ojomoko L, Tsetlin V, Utkin Y. α7- and α9-Containing Nicotinic Acetylcholine Receptors in the Functioning of Immune System and in Pain. Int J Mol Sci 2023; 24:ijms24076524. [PMID: 37047495 PMCID: PMC10095066 DOI: 10.3390/ijms24076524] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/26/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023] Open
Abstract
Nicotinic acetylcholine receptors (nAChRs) present as many different subtypes in the nervous and immune systems, muscles and on the cells of other organs. In the immune system, inflammation is regulated via the vagus nerve through the activation of the non-neuronal α7 nAChR subtype, affecting the production of cytokines. The analgesic properties of α7 nAChR-selective compounds are mostly based on the activation of the cholinergic anti-inflammatory pathway. The molecular mechanism of neuropathic pain relief mediated by the inhibition of α9-containing nAChRs is not fully understood yet, but the role of immune factors in this process is becoming evident. To obtain appropriate drugs, a search of selective agonists, antagonists and modulators of α7- and α9-containing nAChRs is underway. The naturally occurring three-finger snake α-neurotoxins and mammalian Ly6/uPAR proteins, as well as neurotoxic peptides α-conotoxins, are not only sophisticated tools in research on nAChRs but are also considered as potential medicines. In particular, the inhibition of the α9-containing nAChRs by α-conotoxins may be a pathway to alleviate neuropathic pain. nAChRs are involved in the inflammation processes during AIDS and other viral infections; thus they can also be means used in drug design. In this review, we discuss the role of α7- and α9-containing nAChRs in the immune processes and in pain.
Collapse
Affiliation(s)
| | | | | | | | | | - Yuri Utkin
- Correspondence: or ; Tel.: +7-495-3366522
| |
Collapse
|
16
|
Groome JR. Historical Perspective of the Characterization of Conotoxins Targeting Voltage-Gated Sodium Channels. Mar Drugs 2023; 21:md21040209. [PMID: 37103349 PMCID: PMC10142487 DOI: 10.3390/md21040209] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/21/2023] [Accepted: 03/24/2023] [Indexed: 03/30/2023] Open
Abstract
Marine toxins have potent actions on diverse sodium ion channels regulated by transmembrane voltage (voltage-gated ion channels) or by neurotransmitters (nicotinic acetylcholine receptor channels). Studies of these toxins have focused on varied aspects of venom peptides ranging from evolutionary relationships of predator and prey, biological actions on excitable tissues, potential application as pharmacological intervention in disease therapy, and as part of multiple experimental approaches towards an understanding of the atomistic characterization of ion channel structure. This review examines the historical perspective of the study of conotoxin peptides active on sodium channels gated by transmembrane voltage, which has led to recent advances in ion channel research made possible with the exploitation of the diversity of these marine toxins.
Collapse
Affiliation(s)
- James R Groome
- Department of Biological Sciences, Idaho State University, Pocatello, ID 83209, USA
| |
Collapse
|
17
|
Tae HS, Adams DJ. Nicotinic acetylcholine receptor subtype expression, function, and pharmacology: Therapeutic potential of α-conotoxins. Pharmacol Res 2023; 191:106747. [PMID: 37001708 DOI: 10.1016/j.phrs.2023.106747] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/22/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023]
Abstract
The pentameric nicotinic acetylcholine receptors (nAChRs) are typically classed as muscle- or neuronal-type, however, the latter has also been reported in non-neuronal cells. Given their broad distribution, nAChRs mediate numerous physiological and pathological processes including synaptic transmission, presynaptic modulation of transmitter release, neuropathic pain, inflammation, and cancer. There are 17 different nAChR subunits and combinations of these subunits produce subtypes with diverse pharmacological properties. The expression and role of some nAChR subtypes have been extensively deciphered with the aid of knock-out models. Many nAChR subtypes expressed in heterologous systems are selectively targeted by the disulfide-rich α-conotoxins. α-Conotoxins are small peptides isolated from the venom of cone snails, and a number of them have potential pharmaceutical value.
Collapse
|
18
|
Fiorotti HB, Figueiredo SG, Campos FV, Pimenta DC. Cone snail species off the Brazilian coast and their venoms: a review and update. J Venom Anim Toxins Incl Trop Dis 2023; 29:e20220052. [PMID: 36756364 PMCID: PMC9897318 DOI: 10.1590/1678-9199-jvatitd-2022-0052] [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: 08/15/2022] [Accepted: 12/14/2022] [Indexed: 01/31/2023] Open
Abstract
The genus Conus includes over 900 species of marine invertebrates known as cone snails, whose venoms are among the most powerful described so far. This potency is mainly due to the concerted action of hundreds of small bioactive peptides named conopeptides, which target different ion channels and membrane receptors and thus interfere with crucial physiological processes. By swiftly harpooning and injecting their prey and predators with such deadly cocktails, the slow-moving cone snails guarantee their survival in the harsh, competitive marine environment. Each cone snail species produces a unique venom, as the mature sequences of conopeptides from the venoms of different species share very little identity. This biochemical diversity, added to the numerous species and conopeptides contained in their venoms, results in an immense biotechnological and therapeutic potential, still largely unexplored. That is especially true regarding the bioprospection of the venoms of cone snail species found off the Brazilian coast - a region widely known for its biodiversity. Of the 31 species described in this region so far, only four - Conus cancellatus, Conus regius, Conus villepinii, and Conus ermineus - have had their venoms partially characterized, and, although many bioactive molecules have been identified, only a few have been actually isolated and studied. In addition to providing an overview on all the cone snail species found off the Brazilian coast to date, this review compiles the information on the structural and pharmacological features of conopeptides and other molecules identified in the venoms of the four aforementioned species, paving the way for future studies.
Collapse
Affiliation(s)
- Helena B. Fiorotti
- Laboratory of Biochemistry and Biophysics, Butantan Institute, São
Paulo, SP, Brazil.,Graduate Program in Biochemistry, Laboratory of Protein Chemistry
(LQP), Federal University of Espírito Santo, Vitória, ES, Brazil
| | - Suely G. Figueiredo
- Graduate Program in Biochemistry, Laboratory of Protein Chemistry
(LQP), Federal University of Espírito Santo, Vitória, ES, Brazil
| | - Fabiana V. Campos
- Laboratory of Biochemistry and Biophysics, Butantan Institute, São
Paulo, SP, Brazil.,Graduate Program in Biochemistry, Laboratory of Protein Chemistry
(LQP), Federal University of Espírito Santo, Vitória, ES, Brazil
| | - Daniel C. Pimenta
- Laboratory of Biochemistry and Biophysics, Butantan Institute, São
Paulo, SP, Brazil.,Correspondence:
| |
Collapse
|
19
|
Liang J, Tae HS, Zhao Z, Li X, Zhang J, Chen S, Jiang T, Adams DJ, Yu R. Mechanism of Action and Structure-Activity Relationship of α-Conotoxin Mr1.1 at the Human α9α10 Nicotinic Acetylcholine Receptor. J Med Chem 2022; 65:16204-16217. [PMID: 36137181 DOI: 10.1021/acs.jmedchem.2c00494] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
α-Conotoxins (α-CTxs) can selectively target nicotinic acetylcholine receptors (nAChRs) and are important drug leads for the treatment of cancer, chronic pain, and neuralgia. Here, we chemically synthesized a formerly defined rat α7 nAChR targeting α-CTx Mr1.1 and evaluated its activity at human nAChRs. Mr1.1 was most potent at the human (h) α9α10 nAChR with a half-maximal inhibitory concentration (IC50) of 92.0 nM. Molecular dynamic simulations suggested that Mr1.1 favorably binds at the α10(+)α9(-) and α9(+)α9(-) sites via hydrogen bonds and salt bridges, stabilizing the channel in a closed conformation. Although Mr1.1 and another antagonist, α-CTx Vc1.1 share high sequence similarity and disulfide-bond framework, Mr1.1 has distinct orientations at hα9α10. Based on the Mr1.1-hα9α10 model, analogues were generated, and the more potent Mr1.1[S4Dap], antagonized hα9α10 with an IC50 of 4.0 nM. Furthermore, Mr1.1[S4Dap] displayed analgesic activity in the rat chronic constriction injury (CCI) pain model and therefore presents a promising drug candidate.
Collapse
Affiliation(s)
- Jiazhen Liang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao266003, China.,Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao266003, China
| | - Han-Shen Tae
- Illawarra Health and Medical Research Institute (IHMRI), University of Wollongong, Wollongong, New South Wales2522, Australia
| | - Zitong Zhao
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao266003, China.,Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao266003, China
| | - Xiao Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao266003, China.,Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao266003, China
| | - Jinghui Zhang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao266003, China.,Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao266003, China
| | - Shen Chen
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao266003, China.,Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao266003, China
| | - Tao Jiang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao266003, China.,Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao266003, China.,Innovation Center for Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao266003, China
| | - David J Adams
- Illawarra Health and Medical Research Institute (IHMRI), University of Wollongong, Wollongong, New South Wales2522, Australia
| | - Rilei Yu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao266003, China.,Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao266003, China.,Innovation Center for Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao266003, China
| |
Collapse
|
20
|
Margiotta F, Micheli L, Ciampi C, Ghelardini C, McIntosh JM, Di Cesare Mannelli L. Conus regius-Derived Conotoxins: Novel Therapeutic Opportunities from a Marine Organism. Mar Drugs 2022; 20:773. [PMID: 36547920 PMCID: PMC9783627 DOI: 10.3390/md20120773] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/03/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
Abstract
Conus regius is a marine venomous mollusk of the Conus genus that captures its prey by injecting a rich cocktail of bioactive disulfide bond rich peptides called conotoxins. These peptides selectively target a broad range of ion channels, membrane receptors, transporters, and enzymes, making them valuable pharmacological tools and potential drug leads. C. regius-derived conotoxins are particularly attractive due to their marked potency and selectivity against specific nicotinic acetylcholine receptor subtypes, whose signalling is involved in pain, cognitive disorders, drug addiction, and cancer. However, the species-specific differences in sensitivity and the low stability and bioavailability of these conotoxins limit their clinical development as novel therapeutic agents for these disorders. Here, we give an overview of the main pharmacological features of the C. regius-derived conotoxins described so far, focusing on the molecular mechanisms underlying their potential therapeutic effects. Additionally, we describe adoptable chemical engineering solutions to improve their pharmacological properties for future potential clinical translation.
Collapse
Affiliation(s)
- Francesco Margiotta
- Department of Neuroscience, Psychology, Drug Research and Child Health—NEUROFARBA, Pharmacology and Toxicology Section, University of Florence, 50139 Florence, Italy
| | - Laura Micheli
- Department of Neuroscience, Psychology, Drug Research and Child Health—NEUROFARBA, Pharmacology and Toxicology Section, University of Florence, 50139 Florence, Italy
| | - Clara Ciampi
- Department of Neuroscience, Psychology, Drug Research and Child Health—NEUROFARBA, Pharmacology and Toxicology Section, University of Florence, 50139 Florence, Italy
| | - Carla Ghelardini
- Department of Neuroscience, Psychology, Drug Research and Child Health—NEUROFARBA, Pharmacology and Toxicology Section, University of Florence, 50139 Florence, Italy
| | - J. Michael McIntosh
- George E. Wohlen Veterans Affairs Medical Center, Salt Lake City, UT 84148, USA
- Department of Psychiatry, University of Utah, Salt Lake City, UT 84108, USA
- School of Biological Sciences University of Utah, Salt Lake City, UT 84112, USA
| | - Lorenzo Di Cesare Mannelli
- Department of Neuroscience, Psychology, Drug Research and Child Health—NEUROFARBA, Pharmacology and Toxicology Section, University of Florence, 50139 Florence, Italy
| |
Collapse
|
21
|
A Single Amino Acid Replacement Boosts the Analgesic Activity of α-Conotoxin AuIB through the Inhibition of the GABA BR-Coupled N-Type Calcium Channel. Mar Drugs 2022; 20:md20120750. [PMID: 36547897 PMCID: PMC9781320 DOI: 10.3390/md20120750] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/17/2022] [Accepted: 11/22/2022] [Indexed: 11/30/2022] Open
Abstract
α-conotoxin AuIB is the only one of the 4/6 type α-conotoxins (α-CTxs) that inhibits the γ-aminobutyric acid receptor B (GABABR)-coupled N-type calcium channel (CaV2.2). To improve its inhibitory activity, a series of variants were synthesized and evaluated according to the structure-activity relationships of 4/7 type α-CTxs targeting GABABR-coupled CaV2.2. Surprisingly, only the substitution of Pro7 with Arg results in a 2-3-fold increase in the inhibition of GABABR-coupled CaV2.2 (IC50 is 0.74 nM); substitutions of position 9-12 with basic or hydrophobic amino acid and the addition of hydrophobic amino acid Leu or Ile at the second loop to mimic 4/7 type α-CTxs all failed to improve the inhibitory activity of AuIB against GABABR-coupled CaV2.2. Interestingly, the most potent form of AuIB[P7R] has disulfide bridges of "1-4, 2-3" (ribbon), which differs from the "1-3, 2-4" (globular) in the isoforms of wildtype AuIB. In addition, AuIB[P7R](globular) displays potent analgesic activity in the acetic acid writhing model and the partial sciatic nerve injury (PNL) model. Our study demonstrated that 4/6 type α-CTxs, with the disulfide bridge connectivity "1-4, 2-3," are also potent inhibitors for GABABR-coupled CaV2.2, exhibiting potent analgesic activity.
Collapse
|
22
|
Huynh PN, Christensen SB, McIntosh JM. RgIA4 Prevention of Acute Oxaliplatin-Induced Cold Allodynia Requires α9-Containing Nicotinic Acetylcholine Receptors and CD3 + T-Cells. Cells 2022; 11:cells11223561. [PMID: 36428990 PMCID: PMC9688540 DOI: 10.3390/cells11223561] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 11/10/2022] [Accepted: 11/10/2022] [Indexed: 11/13/2022] Open
Abstract
Chemotherapy-induced neuropathic pain is a debilitating and dose-limiting side effect. Oxaliplatin is a third-generation platinum and antineoplastic compound that is commonly used to treat colorectal cancer and commonly yields neuropathic side effects. Available drugs such as duloxetine provide only modest benefits against oxaliplatin-induced neuropathy. A particularly disruptive symptom of oxaliplatin is painful cold sensitivity, known as cold allodynia. Previous studies of the Conus regius peptide, RgIA, and its analogs have demonstrated relief from oxaliplatin-induced cold allodynia, yielding improvement that persists even after treatment cessation. Moreover, underlying inflammatory and neuronal protection were shown at the cellular level in chronic constriction nerve injury models, consistent with disease-modifying effects. Despite these promising preclinical outcomes, the underlying molecular mechanism of action of RgIA4 remains an area of active investigation. This study aimed to determine the necessity of the α9 nAChR subunit and potential T-cell mechanisms in RgIA4 efficacy against acute oxaliplatin-induced cold allodynia. A single dose of oxaliplatin (10 mg/kg) was utilized followed by four daily doses of RgIA4. Subcutaneous administration of RgIA4 (40 µg/kg) prevented cold allodynia in wildtype mice but not in mice lacking the α9 nAChR-encoding gene, chrna9. RgIA4 also failed to reverse allodynia in mice depleted of CD3+ T-cells. In wildtype mice treated with oxaliplatin, quantitated circulating T-cells remained unaffected by RgIA4. Together, these results show that RgIA4 requires both chrna9 and CD3+ T-cells to exert its protective effects against acute cold-allodynia produced by oxaliplatin.
Collapse
Affiliation(s)
- Peter N. Huynh
- School of Biological Sciences, University of Utah, Salt Lake City, UT 84112, USA
- Correspondence:
| | - Sean B. Christensen
- School of Biological Sciences, University of Utah, Salt Lake City, UT 84112, USA
| | - J. Michael McIntosh
- School of Biological Sciences, University of Utah, Salt Lake City, UT 84112, USA
- George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, UT 84112, USA
- Department of Psychiatry, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
23
|
Bedeir MM, Ninoyu Y, Nakamura T, Tsujikawa T, Hirano S. Multiplex immunohistochemistry reveals cochlear macrophage heterogeneity and local auditory nerve inflammation in cisplatin-induced hearing loss. Front Neurol 2022; 13:1015014. [PMID: 36341090 PMCID: PMC9633043 DOI: 10.3389/fneur.2022.1015014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 09/26/2022] [Indexed: 08/11/2023] Open
Abstract
Inner ear macrophages play a vital role in cochlear homeostasis. Recent studies have demonstrated the existence of macrophages at different sites of the cochlea, with increased cochlear infiltration as an inflammatory response mechanism to injury. However, current methods, such as conventional immunohistochemistry and flow cytometry, provide limited information about the diversity of cochlear macrophages. Recently, multiplex immunohistochemistry (mIHC) successfully identified the heterogeneity of immune cells in cancer tissue and thereby improved our understanding of the disease prognosis. In this study, we modified the mIHC technique for cochlear tissue and utilized it to investigate cochlear macrophage behavior and heterogeneity before and after exposure to ototoxic drugs such as cisplatin. Four-week-old C57BL/6N female mice were intraperitoneally injected with cisplatin at 5 mg/kg/day consecutively for 6 days. Their hearing levels were assessed before and after the injection. Their cochleae were harvested before (day 0) and on days 8 and 15 after the cisplatin injection. Paraffin-embedded sections were sequentially immunostained using macrophage surface markers to identify the different categories of macrophages. Each immunostaining cycle included incubation with primary antibody, incubation with secondary antibody, chromogenic staining, and image scanning. Thereafter, all antibodies were stripped out, and antigen retrieval was performed to prepare the tissue for the next cycle. The results revealed that activated cochlear macrophages were not entirely differentiated into M1 or M2 categories but into multi-marker M1/M2 mixed macrophages. Furthermore, the ratio of these mixed (M1/M2) macrophages to Iba1+ macrophages increased in the auditory nerve after cisplatin exposure, suggesting local auditory nerve inflammation. The increase in the population of activated macrophages in the auditory nerve region was concomitant with the temporary shift of hearing threshold on day 8 post-cisplatin injection. The findings of this study indicate the effectiveness of mIHC in identifying cochlear macrophage heterogeneity both in the resting state and after cisplatin exposure. Therefore, mIHC could be a powerful tool in cochlear immunology research. Our findings may provide new insights into the co-relation between the cochlear macrophage and cisplatin exposure.
Collapse
|
24
|
Hone AJ, McIntosh JM. Alkaloid ligands enable function of homomeric human α10 nicotinic acetylcholine receptors. Front Pharmacol 2022; 13:981760. [PMID: 36188578 PMCID: PMC9523446 DOI: 10.3389/fphar.2022.981760] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 08/23/2022] [Indexed: 01/29/2023] Open
Abstract
In the nervous system, nicotinic acetylcholine receptors (nAChRs) rapidly transduce a chemical signal into one that is electrical via ligand-gated ion flux through the central channel of the receptor. However, some nAChR subunits are expressed by non-excitable cells where signal transduction apparently occurs through non-ionic mechanisms. One such nAChR subunit, α10, is present in a discreet subset of immune cells and has been implicated in pathologies including cancer, neuropathic pain, and chronic inflammation. Longstanding convention holds that human α10 subunits require co-assembly with α9 subunits for function. Here we assessed whether cholinergic ligands can enable or uncover ionic functions from homomeric α10 nAChRs. Xenopus laevis oocytes expressing human α10 subunits were exposed to a panel of ligands and examined for receptor activation using voltage-clamp electrophysiology. Functional expression of human α10 nAChRs was achieved by exposing the oocytes to the alkaloids strychnine, brucine, or methyllycaconitine. Furthermore, acute exposure to the alkaloid ligands significantly enhanced ionic responses. Acetylcholine-gated currents mediated by α10 nAChRs were potently inhibited by the snake toxins α-bungarotoxin and α-cobratoxin but not by α-conotoxins that target α9 and α9α10 nAChRs. Our findings indicate that human α10 homomers are expressed in oocytes and exposure to certain ligands can enable ionic functions. To our knowledge, this is the first demonstration that human α10 subunits can assemble as functional homomeric nAChRs. These findings have potential implications for receptor regulatory-mechanisms and will enable structural, functional, and further pharmacological characterization of human α10 nAChRs.
Collapse
Affiliation(s)
- Arik J. Hone
- School of Biological Sciences, University of Utah, Salt Lake City, UT, United States
- MIRECC, George E. Whalen Veterans Affairs Medical Center, Salt Lake City, UT, United States
| | - J. Michael McIntosh
- School of Biological Sciences, University of Utah, Salt Lake City, UT, United States
- Department of Psychiatry, University of Utah, Salt Lake City, UT, United States
- George E. Whalen Veterans Affairs Medical Center, Salt Lake City, UT, United States
| |
Collapse
|
25
|
Zhu X, Yuan M, Wang H, Zhangsun D, Yu G, Che J, Luo S. Novel αO-conotoxin GeXIVA[1,2] Nonaddictive Analgesic with Pharmacokinetic Modelling-Based Mechanistic Assessment. Pharmaceutics 2022; 14:pharmaceutics14091789. [PMID: 36145535 PMCID: PMC9505004 DOI: 10.3390/pharmaceutics14091789] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/19/2022] [Accepted: 08/24/2022] [Indexed: 11/17/2022] Open
Abstract
αO-conotoxin GeXIVA[1,2] was isolated in our laboratory from Conus generalis, a snail native to the South China Sea, and is a novel, nonaddictive, intramuscularly administered analgesic targeting the α9α10 nicotinic acetylcholine receptor (nAChR) with an IC50 of 4.61 nM. However, its pharmacokinetics and related mechanisms underlying the analgesic effect remain unknown. Herein, pharmacokinetics and multiscale pharmacokinetic modelling in animals were subjected systematically to mechanistic assessment for αO-conotoxin GeXIVA[1,2]. The intramuscular bioavailability in rats and dogs was 11.47% and 13.37%, respectively. The plasma exposure of GeXIVA[1,2] increased proportionally with the experimental dose. The plasma protein binding of GeXIVA[1,2] differed between the tested animal species. The one-compartment model with the first-order absorption population pharmacokinetics model predicted doses for humans with bodyweight as the covariant. The pharmacokinetics-pharmacodynamics relationships were characterized using an inhibitory loss indirect response model with an effect compartment. Model simulations have provided potential mechanistic insights into the analgesic effects of GeXIVA[1,2] by inhibiting certain endogenous substances, which may be a key biomarker. This report is the first concerning the pharmacokinetics of GeXIVA[1,2] and its potential analgesic mechanisms based on a top-down modelling approach.
Collapse
Affiliation(s)
- Xiaoyu Zhu
- Key Laboratory of Tropical Biological Resources, Ministry of Education, Hainan University, Haikou 570228, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Mei Yuan
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Huanbai Wang
- Key Laboratory of Tropical Biological Resources, Ministry of Education, Hainan University, Haikou 570228, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Dongting Zhangsun
- Key Laboratory of Tropical Biological Resources, Ministry of Education, Hainan University, Haikou 570228, China
| | - Gang Yu
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
- Correspondence: (G.Y.); (J.C.); (S.L.)
| | - Jinjing Che
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
- Correspondence: (G.Y.); (J.C.); (S.L.)
| | - Sulan Luo
- Key Laboratory of Tropical Biological Resources, Ministry of Education, Hainan University, Haikou 570228, China
- Medical School, Guangxi University, Nanning 530004, China
- Correspondence: (G.Y.); (J.C.); (S.L.)
| |
Collapse
|
26
|
Wang S, Bartels P, Zhao C, Yousuf A, Liu Z, Yu S, Bony AR, Ma X, Dai Q, Sun T, Liu N, Yang M, Yu R, Du W, Adams DJ, Dai Q. A 4/8 Subtype α-Conotoxin Vt1.27 Inhibits N-Type Calcium Channels With Potent Anti-Allodynic Effect. Front Pharmacol 2022; 13:881732. [PMID: 35754473 PMCID: PMC9230573 DOI: 10.3389/fphar.2022.881732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 04/08/2022] [Indexed: 11/22/2022] Open
Abstract
A novel 4/8 subtype α-conotoxin, Vt1.27 (NCCMFHTCPIDYSRFNC-NH2), was identified from Conus vitulinus in the South China Sea by RACE methods. The peptide was synthesized and structurally characterized. Similar to other α-conotoxins that target neuronal nicotinic acetylcholine receptor (nAChR) subtypes, Vt1.27 inhibited the rat α3β2 nAChR subtype (IC50 = 1160 nM) and was inactive at voltage-gated sodium and potassium channels in rat sensory neurons. However, Vt1.27 inhibited high voltage-activated N-type (CaV2.2) calcium channels expressed in HEK293T cells with an IC50 of 398 nM. An alanine scan of the peptide showed that residues Phe5, Pro9, Ile10, and Ser13 contribute significantly to the inhibitory activity of Vt1.27. The molecular dockings indicate that Vt1.27 inhibits the transmembrane region of CaV2.2, which is different from that of ω-conotoxins. Furthermore, Vt1.27 exhibited potent anti-allodynic effect in rat partial sciatic nerve injury (PNL) and chronic constriction injury (CCI) pain models at 10 nmol/kg level with the intramuscular injection. The pain threshold elevation of Vt1.27 groups was higher than that of α-conotoxin Vc1.1 in CCI rat models. These findings expand our knowledge of targets of α-conotoxins and potentially provide a potent, anti-allodynic peptide for the treatment of neuropathic pain.
Collapse
Affiliation(s)
- Shuo Wang
- Beijing Institute of Biotechnology, Beijing, China
- Department of Pharmacy, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Peter Bartels
- Illawarra Health and Medical Research Institute (IHMRI), University of Wollongong, Wollongong, NSW, Australia
| | - Cong Zhao
- Department of Chemistry, Renmin University of China, Beijing, China
| | - Arsalan Yousuf
- Illawarra Health and Medical Research Institute (IHMRI), University of Wollongong, Wollongong, NSW, Australia
| | - Zhuguo Liu
- Beijing Institute of Biotechnology, Beijing, China
| | - Shuo Yu
- Beijing Institute of Biotechnology, Beijing, China
| | - Anuja R. Bony
- Illawarra Health and Medical Research Institute (IHMRI), University of Wollongong, Wollongong, NSW, Australia
| | - Xiaoli Ma
- Beijing Institute of Biotechnology, Beijing, China
| | - Qin Dai
- Beijing Institute of Biotechnology, Beijing, China
| | - Ting Sun
- Beijing Institute of Biotechnology, Beijing, China
| | - Na Liu
- Beijing Institute of Biotechnology, Beijing, China
| | - Mengke Yang
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Rilei Yu
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Weihong Du
- Department of Chemistry, Renmin University of China, Beijing, China
- *Correspondence: Qiuyun Dai, ; David J. Adams, ; Weihong Du,
| | - David J. Adams
- Illawarra Health and Medical Research Institute (IHMRI), University of Wollongong, Wollongong, NSW, Australia
- *Correspondence: Qiuyun Dai, ; David J. Adams, ; Weihong Du,
| | - Qiuyun Dai
- Beijing Institute of Biotechnology, Beijing, China
- *Correspondence: Qiuyun Dai, ; David J. Adams, ; Weihong Du,
| |
Collapse
|
27
|
Papke RL, Andleeb H, Stokes C, Quadri M, Horenstein NA. Selective Agonists and Antagonists of α9 Versus α7 Nicotinic Acetylcholine Receptors. ACS Chem Neurosci 2022; 13:624-637. [PMID: 35167270 PMCID: PMC9547379 DOI: 10.1021/acschemneuro.1c00747] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Nicotinic acetylcholine receptors containing α9 subunits are essential for the auditory function and have been implicated, along with α7-containing nicotinic receptors, as potential targets for the treatment of inflammatory and neuropathic pain. The study of α9-containing receptors has been hampered by the lack of selective agonists. The only α9-selective antagonists previously identified are peptide conotoxins. Curiously, the activity of α7 and α9 receptors as modulators of inflammatory pain appears to not rely strictly on ion channel activation, which led to the identification of α7 "silent agonists" and phosphocholine as an "unconventional agonist" for α9 containing receptors. The parallel testing of the α7 silent agonist p-CF3-diEPP and phosphocholine led to the discovery that p-CF3-diEPP was an α9 agonist. In this report, we compared the activity of α7 and α9 with a family of structurally related compounds, most of which were previously shown to be α7 partial or silent agonists. We identify several potent α9-selective agonists as well as numerous potent and selective α9 antagonists and describe the structural basis for these activities. Several of these compounds have previously been shown to be effective in animal models of inflammatory pain, an activity that was assumed to be due to α7 silent agonism but may, in fact, be due to α9 activity. The α9-selective conotoxin antagonists have also been shown to reduce pain in similar models. Our identification of these new α9 agonists and antagonists may prove to be invaluable for defining an optimal approach for treating pain, allowing for reduced use of opioid drugs.
Collapse
Affiliation(s)
- Roger L. Papke
- Department of Pharmacology and Therapeutics, University of Florida, PO Box 100267 Gainesville, FL 32610 USA (CS, RLP),To whom correspondence should be addressed: Roger L. Papke, Ph.D., , Department of Pharmacology and Therapeutics, University of Florida, PO Box 100267, Gainesville FL, 32610-0267
| | - Hina Andleeb
- Department of Chemistry, University of Florida, PO Box 117200, Gainesville, FL 32611-7200 USA (HA, MQ, NAH)
| | - Clare Stokes
- Department of Pharmacology and Therapeutics, University of Florida, PO Box 100267 Gainesville, FL 32610 USA (CS, RLP)
| | - Marta Quadri
- Department of Chemistry, University of Florida, PO Box 117200, Gainesville, FL 32611-7200 USA (HA, MQ, NAH)
| | - Nicole A. Horenstein
- Department of Chemistry, University of Florida, PO Box 117200, Gainesville, FL 32611-7200 USA (HA, MQ, NAH)
| |
Collapse
|
28
|
Peng S, Ying AF, Chan NJH, Sundar R, Soon YY, Bandla A. Prevention of Oxaliplatin-Induced Peripheral Neuropathy: A Systematic Review and Meta-Analysis. Front Oncol 2022; 12:731223. [PMID: 35186722 PMCID: PMC8853097 DOI: 10.3389/fonc.2022.731223] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 01/10/2022] [Indexed: 01/06/2023] Open
Abstract
Background Oxaliplatin-induced peripheral neuropathy (OIPN) has significant clinical impact on the quality of life for cancer patients and is a dose limiting toxicity. Trials studying preventive measures have been inconclusive. A systematic review and meta-analysis were conducted to evaluate the existing pharmacological and non-pharmacological interventions to prevent chronic OIPN. Methods Literature databases PubMed-MEDLINE, Embase and Scopus, were searched from 1 Jan 2005 to 08 Aug 2020 and major conferences’ abstracts were reviewed for randomized controlled trials that examined the efficacy of any preventive measure for OIPN. The primary outcome measure was the incidence of chronic OIPN with a preventive intervention as compared to placebo or no intervention. The pooled risk ratio and its 95% confidence interval were calculated using a random effects model. A network meta-analysis was conducted to derive indirect evidence of any preventive effect of an intervention against placebo when original trials compared one intervention against another. Results Forty-four trials were analyzed describing 29 chemoprotective interventions, including combinations, and 1 non-pharmacological intervention. Ratings were assessed via a combination of outcomes with quality assessment using the Grading of Recommendations, Assessment, Development and Evaluations (GRADE) framework. Of the 30 interventions examined, there were six interventions supporting potential efficacy, 11 interventions with insufficient evidence and 13 interventions not recommended. Conclusion Currently, there is insufficient certainty to support any intervention as effective in preventing OIPN. Of note is that most of these studies have focused on pharmacological interventions; non-pharmacological interventions are underexplored. Further research on ways to limit OIPN is needed. Systematic Review Registration https://www.crd.york.ac.uk/prospero/display_record.php?RecordID=225095, Prospero Registration Number: CRD42021225095.
Collapse
Affiliation(s)
- Siyu Peng
- Department of Medicine, National University Health System, Singapore, Singapore
| | - Ariel Fangting Ying
- Health Services and System Research, Duke-National University of Singapore (NUS) Medical School, Singapore, Singapore
| | | | - Raghav Sundar
- Department of Haematology-Oncology, National University Cancer Institute Singapore, National University Health System, Singapore, Singapore.,The N.1 Institute for Health, National University of Singapore, Singapore, Singapore.,Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yu Yang Soon
- Department of Radiation Oncology, National University Cancer Institute Singapore, National University Health System, Singapore, Singapore
| | - Aishwarya Bandla
- Department of Haematology-Oncology, National University Cancer Institute Singapore, National University Health System, Singapore, Singapore.,The N.1 Institute for Health, National University of Singapore, Singapore, Singapore
| |
Collapse
|
29
|
Akgül Ö, Lucarini E, Mannelli LDC, Ghelardini C, D'Ambrosio K, Buonanno M, Monti SM, De Simone G, Angeli A, Supuran CT, Carta F. Sultam based Carbonic Anhydrase VII inhibitors for the management of neuropathic pain. Eur J Med Chem 2022; 227:113956. [PMID: 34731762 DOI: 10.1016/j.ejmech.2021.113956] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 10/24/2021] [Accepted: 10/26/2021] [Indexed: 12/24/2022]
Abstract
We report a series of compounds 1-17 derived from the antiepileptic drug Sulthiame (SLT) from which both the benzenesulfonamide and the sultam moiety were retained. All compounds were tested in vitro for their inhibition activity against the human (h) Carbonic Anhydrase (CA; EC 4.2.1.1) I, II, VII, IX and XII isoforms. Among the series, derivatives 1 and 11 showed great enhancement of both inhibition potency and selectivity towards the hCA VII isoform, when compared to the reference SLT drug. The binding mode of 11 within the hCA VII active site was deciphered by means of X-ray crystallography and revealed the sultam moiety being exposed to the rim of the active site. In vivo experiments on a model of neuropathic pain induced by oxaliplatin clearly showed 11 being an effective pain relieving agent and therefore worth of further exploitation towards the validation of the hCA VII as new target for the management of neuropathies.
Collapse
Affiliation(s)
- Özlem Akgül
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ege University, 35100, Bornova, İzmir, Turkey
| | - Elena Lucarini
- NEUROFARBA Department, Section of Pharmacology and Toxicology, Università degli Studi di Firenze, Viale Pieraccini 6, 50139, Florence, Italy
| | - Lorenzo Di Cesare Mannelli
- NEUROFARBA Department, Section of Pharmacology and Toxicology, Università degli Studi di Firenze, Viale Pieraccini 6, 50139, Florence, Italy
| | - Carla Ghelardini
- NEUROFARBA Department, Section of Pharmacology and Toxicology, Università degli Studi di Firenze, Viale Pieraccini 6, 50139, Florence, Italy
| | - Katia D'Ambrosio
- Institute of Biostructures and Bioimaging, CNR, via Mezzocannone 16, 80134, Naples, Italy
| | - Martina Buonanno
- Institute of Biostructures and Bioimaging, CNR, via Mezzocannone 16, 80134, Naples, Italy
| | - Simona Maria Monti
- Institute of Biostructures and Bioimaging, CNR, via Mezzocannone 16, 80134, Naples, Italy
| | - Giuseppina De Simone
- Institute of Biostructures and Bioimaging, CNR, via Mezzocannone 16, 80134, Naples, Italy
| | - Andrea Angeli
- Università degli Studi di Firenze, NEUROFARBA Dept., Sezione di Scienze Farmaceutiche e Nutraceutiche, Via Ugo Schiff 6, 50019, Sesto Fiorentino (Florence), Italy
| | - Claudiu T Supuran
- Università degli Studi di Firenze, NEUROFARBA Dept., Sezione di Scienze Farmaceutiche e Nutraceutiche, Via Ugo Schiff 6, 50019, Sesto Fiorentino (Florence), Italy
| | - Fabrizio Carta
- Università degli Studi di Firenze, NEUROFARBA Dept., Sezione di Scienze Farmaceutiche e Nutraceutiche, Via Ugo Schiff 6, 50019, Sesto Fiorentino (Florence), Italy.
| |
Collapse
|
30
|
Morgese MG, Bove M, Di Cesare Mannelli L, Schiavone S, Colia AL, Dimonte S, Mhillaj E, Sikora V, Tucci P, Ghelardini C, Trabace L. Precision Medicine in Alzheimer's Disease: Investigating Comorbid Common Biological Substrates in the Rat Model of Amyloid Beta-Induced Toxicity. Front Pharmacol 2022; 12:799561. [PMID: 35046821 PMCID: PMC8763383 DOI: 10.3389/fphar.2021.799561] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 12/01/2021] [Indexed: 12/30/2022] Open
Abstract
Alzheimer's disease (AD), one of the most widespread neurodegenerative disorder, is a fatal global burden for the elder population. Although many efforts have been made, the search of a curative therapy is still ongoing. Individuating phenotypic traits that might help in investigating treatment response is of growing interest in AD research. AD is a complex pathology characterized by many comorbidities, such as depression and increased susceptibility to pain perception, leading to postulate that these conditions may rely on common biological substrates yet to be determined. In order to investigate those biological determinants to be associable with phenotypic traits, we used the rat model of amyloid beta-induced toxicity. This established model of early phase of AD is obtained by the intracerebroventricular injection of soluble amyloid beta1-42 (Aβ) peptide 7 days before performing experiments. In this model, we have previously reported increased immobility in the forced swimming test, reduced cortical serotonin levels and subtle alterations in the cognitive domain a depressive-like phenotype associated with subtle alteration in memory processes. In light of evaluating pain perception in this animal model, we performed two different behavioral tests commonly used, such as the paw pressure test and the cold plate test, to analyze mechanical hyperalgesia and thermal allodynia, respectively. Behavioural outcomes confirmed the memory impairment in the social recognition test and, compared to sham, Aβ-injected rats showed an increased selective susceptibility to mechanical but not to thermal stimulus. Behavioural data were then corroborated by neurochemical and biochemical biomarker analyses either at central or peripheral level. Data showed that the peptide injection evoked a significant increase in hypothalamic glutamate, kynurenine and dopamine content, while serotonin levels were reduced. Plasma Cystatin-C, a cysteine protease, was increased while serotonin and melatonin levels were decreased in Aβ-injected rats. Urinary levels paralleled plasma quantifications, indicating that Aβ-induced deficits in pain perception, mood and cognitive domain may also depend on these biomarkers. In conclusion, in the present study, we demonstrated that this animal model can mimic several comorbid conditions typical of the early phase of AD. Therefore, in the perspective of generating novel therapeutic strategies relevant to precision medicine in AD, this animal model and the biomarkers evaluated herein may represent an advantageous approach.
Collapse
Affiliation(s)
- Maria Grazia Morgese
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Maria Bove
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Lorenzo Di Cesare Mannelli
- Pharmacology and Toxicology Section, Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Firenze, Firenze, Italy
| | - Stefania Schiavone
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Anna Laura Colia
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Stefania Dimonte
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Emanuela Mhillaj
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Vladyslav Sikora
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
- Department of Pathology, Sumy State University, Sumy, Ukraine
| | - Paolo Tucci
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Carla Ghelardini
- Pharmacology and Toxicology Section, Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Firenze, Firenze, Italy
| | - Luigia Trabace
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| |
Collapse
|
31
|
Dyachenko IA, Palikova YA, Palikov VA, Korolkova YV, Kazakov VA, Egorova NS, Garifulina AI, Utkin YN, Tsetlin VI, Kryukova EV. α-Conotoxin RgIA and oligoarginine R8 in the mice model alleviate long-term oxaliplatin induced neuropathy. Biochimie 2021; 194:127-136. [PMID: 34979156 DOI: 10.1016/j.biochi.2021.12.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/27/2021] [Accepted: 12/28/2021] [Indexed: 12/28/2022]
Abstract
Оligoarginines were recently discovered (Lebedev et al., 2019 Nov) [1] as a novel class of nicotinic acetylcholine receptors (nAChRs) inhibitors, octaoligoarginine R8 showing a relatively high affinity (40 nM) for the α9/α10 nAChR. Since the inhibition of α9/α10 nAChR by α-conotoxin RgIA and its analogs is a possible way to drugs against neuropathic pain, here in a mice model we compared R8 with α-conotoxin RgIA in the effects on the chemotherapy-induced peripheral neuropathy (CIPN), namely on the long-term oxaliplatin induced neuropathy. Tests of cold allodynia, hot plate, Von Frey and grip strength analysis revealed for R8 and α-conotoxin RgIA similar positive effects, expressed most prominently after two weeks of administration. Histological analysis of the dorsal root ganglia sections showed for R8 and RgIA a similar partial correction of changes in the nuclear morphology of neurons. Since α9/α10 nAChR might be not the only drug target for R8, we analyzed the R8 action on rat TRPV1 and TRPA1, well-known nociceptive receptors. Against rTRPV1 at 25 μM there was no inhibition, while for rTRPA1 IC50 was about 20 μM. Thus, involvement of rTRPA1 cannot be excluded, but in view of the R8 much higher affinity for α9/α10 nAChR the latter seems to be the main target and the easily synthesized R8 can be considered as a potential candidate for a drug design.
Collapse
Affiliation(s)
- I A Dyachenko
- Branch of the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 6 Nauki Avenue, 142290, Pushchino, Moscow, Russia.
| | - Yu A Palikova
- Branch of the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 6 Nauki Avenue, 142290, Pushchino, Moscow, Russia.
| | - V A Palikov
- Branch of the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 6 Nauki Avenue, 142290, Pushchino, Moscow, Russia.
| | - Y V Korolkova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997, Moscow, Russia.
| | - V A Kazakov
- Branch of the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 6 Nauki Avenue, 142290, Pushchino, Moscow, Russia.
| | - N S Egorova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997, Moscow, Russia.
| | - A I Garifulina
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, A-1090, Vienna, Austria.
| | - Y N Utkin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997, Moscow, Russia.
| | - V I Tsetlin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997, Moscow, Russia.
| | - E V Kryukova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997, Moscow, Russia.
| |
Collapse
|
32
|
Tanini D, Carradori S, Capperucci A, Lupori L, Zara S, Ferraroni M, Ghelardini C, Mannelli L, Micheli L, Lucarini E, Carta F, Angeli A, Supuran CT. Chalcogenides-incorporating carbonic anhydrase inhibitors concomitantly reverted oxaliplatin-induced neuropathy and enhanced antiproliferative action. Eur J Med Chem 2021; 225:113793. [PMID: 34507012 DOI: 10.1016/j.ejmech.2021.113793] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 08/17/2021] [Accepted: 08/18/2021] [Indexed: 12/12/2022]
Abstract
Platinum-based chemotherapy is widely used for the treatment of different tumors but is associated with serious side effects, among which neuropathic pain. Carbonic anhydrase (CA, EC 4.2.1.1) inhibitors have recently been validated as therapeutic agents in neuropathic pain and as antitumor agents. We report the synthesis of new organochalcogenides bearing the benzensulfonamide moiety acting as potent inhibitors of several human CA isoforms and, in particular, against hCA II and VII endowed with potent neuropathic pain attenuating effects. Moreover, in combination with cisplatin or doxorubicin, some of the new CA inhibitors enhanced the effects of the anticancer drugs capability in counteracting breast cancer MCF7 cell viability. The concomitant anti-neuropathic pain and antiproliferative effects of the new chalcogenide-based CA inhibitors represent an innovative approach for the counteraction and management of side effects associated with clinically platinum drugs as antitumor agents.
Collapse
Affiliation(s)
- Damiano Tanini
- University of Florence, Department of Chemistry "Ugo Schiff", Via Della Lastruccia 3-13, I-50019, Sesto Fiorentino, Italy
| | - Simone Carradori
- Department of Pharmacy, "G. D'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Antonella Capperucci
- University of Florence, Department of Chemistry "Ugo Schiff", Via Della Lastruccia 3-13, I-50019, Sesto Fiorentino, Italy
| | - Lucrezia Lupori
- University of Florence, Department of Chemistry "Ugo Schiff", Via Della Lastruccia 3-13, I-50019, Sesto Fiorentino, Italy
| | - Susi Zara
- Department of Pharmacy, "G. D'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Marta Ferraroni
- University of Florence, Department of Chemistry "Ugo Schiff", Via Della Lastruccia 3-13, I-50019, Sesto Fiorentino, Italy
| | - Carla Ghelardini
- Pharmacology and Toxicology Section, Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Firenze, Firenze, Italy
| | - Ldc Mannelli
- Pharmacology and Toxicology Section, Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Firenze, Firenze, Italy
| | - Laura Micheli
- Pharmacology and Toxicology Section, Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Firenze, Firenze, Italy
| | - Elena Lucarini
- Pharmacology and Toxicology Section, Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Firenze, Firenze, Italy
| | - Fabrizio Carta
- NEUROFARBA Department, Sezione di Scienze Farmaceutiche, University of Florence, Via Ugo Schiff 6, 50019, Sesto Fiorentino, Florence, Italy
| | - Andrea Angeli
- NEUROFARBA Department, Sezione di Scienze Farmaceutiche, University of Florence, Via Ugo Schiff 6, 50019, Sesto Fiorentino, Florence, Italy; Centre of Advanced Research in Bionanoconjugates and Biopolymers Department, "Petru Poni" Institute of Macromolecular Chemistry, 707410, Iasi, Romania.
| | - Claudiu T Supuran
- NEUROFARBA Department, Sezione di Scienze Farmaceutiche, University of Florence, Via Ugo Schiff 6, 50019, Sesto Fiorentino, Florence, Italy
| |
Collapse
|
33
|
Gajewiak J, Christensen S, Dowell C, Hararah F, Fisher F, Huynh PN, Olivera B, McIntosh JM. Selective Penicillamine Substitution Enables Development of a Potent Analgesic Peptide that Acts through a Non-Opioid-Based Mechanism. J Med Chem 2021; 64:9271-9278. [PMID: 34142837 PMCID: PMC8360267 DOI: 10.1021/acs.jmedchem.1c00512] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Venom-derived compounds are of broad interest in neuropharmacology and drug development. α-Conotoxins are small disulfide-containing peptides from Conus snails that target nicotinic acetylcholine receptors (nAChRs) and are in clinical development for non-opioid-based treatment of intractable pain. Although refined by evolution for interaction with target prey receptors, enhancements of pharmacological properties are needed for use in mammalian systems. Therefore, we synthesized analogues of α-conotoxin RgIA using a combination of selective penicillamine substitutions together with natural and non-natural amino acid replacements. This approach resulted in a peptide with 9000-fold increased potency on the human α9α10 nAChR and improved resistance to disulfide shuffling compared to the native peptide. The lead analogue, RgIA-5474, potently blocked α9α10 nAChRs, but not opioid- or other pain-related targets. In addition, RgIA-5474 effectively reversed chemotherapy-induced neuropathic pain.
Collapse
Affiliation(s)
- Joanna Gajewiak
- School of Biological Sciences, University of Utah, 257 South 1400 East, Salt Lake City, UT 84112-0840
| | - Sean Christensen
- School of Biological Sciences, University of Utah, 257 South 1400 East, Salt Lake City, UT 84112-0840
| | - Cheryl Dowell
- School of Biological Sciences, University of Utah, 257 South 1400 East, Salt Lake City, UT 84112-0840
| | - Fuaad Hararah
- School of Biological Sciences, University of Utah, 257 South 1400 East, Salt Lake City, UT 84112-0840
| | - Fernando Fisher
- School of Biological Sciences, University of Utah, 257 South 1400 East, Salt Lake City, UT 84112-0840
| | - Peter N. Huynh
- School of Biological Sciences, University of Utah, 257 South 1400 East, Salt Lake City, UT 84112-0840
| | - Baldomero Olivera
- School of Biological Sciences, University of Utah, 257 South 1400 East, Salt Lake City, UT 84112-0840
| | - J. Michael McIntosh
- School of Biological Sciences, University of Utah, 257 South 1400 East, Salt Lake City, UT 84112-0840
- George E. Wahlen Department of Veterans Affairs Medical Center, 500 Foothill Dr, Salt Lake City, UT 84148
- Department of Psychiatry, University of Utah, 501 Chipeta Way, Salt Lake City, UT 84108
| |
Collapse
|
34
|
Wu Y, Qiang Y, Zhang G, Zhou M. Acute toxicity and micronucleus test of conotoxin lt14a in mice. Basic Clin Pharmacol Toxicol 2021; 129:52-60. [PMID: 33742558 DOI: 10.1111/bcpt.13582] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 03/09/2021] [Accepted: 03/15/2021] [Indexed: 01/09/2023]
Abstract
Conotoxins, which target ion channels or neurotransmitter receptors with high specificity, are valuable in drug development for pain, epilepsy and other neurological diseases. However, the toxicology of conotoxins is rarely reported. In this study, we primarily researched parts of the pharmacological and toxicological properties of an analgesic conotoxin lt14a. Three doses of lt14a (1, 5 and 10 mg/kg) could prolong the pentobarbital-induced sleep time of mice and showed no significant effect on the spontaneous locomotor activity of mice. Three doses of lt14a (50, 100 and 200 mg/kg) did not increase micronucleus rate in the micronucleus test. In addition, three doses of lt14a (200, 500 and 1000 mg/kg) showed no pathological change on the heart or brain of mice in the acute toxicity test. The high dose of lt14a (1000 times the effective analgesic dose) had a certain damaging effect on the liver and lung according to serological detection and histopathology. As part of the preclinical studies, our results provide acute toxicity and mutagenicity evaluation of the promising analgesic conotoxin lt14a.
Collapse
Affiliation(s)
- Yun Wu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, China.,National Engineering Research Center of South China Sea Marine Biotechnology, Guangzhou, China
| | - Yuanyuan Qiang
- National Engineering Research Center of South China Sea Marine Biotechnology, Guangzhou, China.,Ningxia Key Laboratory for Cerebrocranical Disease, Ningxia Medical University, Yinchuan, China
| | - Guangxian Zhang
- National Engineering Research Center of South China Sea Marine Biotechnology, Guangzhou, China.,China School of Fundamental Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Maojun Zhou
- National Engineering Research Center of South China Sea Marine Biotechnology, Guangzhou, China.,Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
35
|
Fumagalli G, Monza L, Cavaletti G, Rigolio R, Meregalli C. Neuroinflammatory Process Involved in Different Preclinical Models of Chemotherapy-Induced Peripheral Neuropathy. Front Immunol 2021; 11:626687. [PMID: 33613570 PMCID: PMC7890072 DOI: 10.3389/fimmu.2020.626687] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 12/21/2020] [Indexed: 12/12/2022] Open
Abstract
Peripheral neuropathies are characterized by nerves damage and axonal loss, and they could be classified in hereditary or acquired forms. Acquired peripheral neuropathies are associated with several causes, including toxic agent exposure, among which the antineoplastic compounds are responsible for the so called Chemotherapy-Induced Peripheral Neuropathy (CIPN). Several clinical features are related to the use of anticancer drugs which exert their action by affecting different mechanisms and structures of the peripheral nervous system: the axons (axonopathy) or the dorsal root ganglia (DRG) neurons cell body (neuronopathy/ganglionopathy). In addition, antineoplastic treatments may affect the blood brain barrier integrity, leading to cognitive impairment that may be severe and long-lasting. CIPN may affect patient quality of life leading to modification or discontinuation of the anticancer therapy. Although the mechanisms of the damage are not completely understood, several hypotheses have been proposed, among which neuroinflammation is now emerging to be relevant in CIPN pathophysiology. In this review, we consider different aspects of neuro-immune interactions in several CIPN preclinical studies which suggest a critical connection between chemotherapeutic agents and neurotoxicity. The features of the neuroinflammatory processes may be different depending on the type of drug (platinum derivatives, taxanes, vinca alkaloids and proteasome inhibitors). In particular, recent studies have demonstrated an involvement of the immune response (both innate and adaptive) and the stimulation and secretion of mediators (cytokines and chemokines) that may be responsible for the painful symptoms, whereas glial cells such as satellite and Schwann cells might contribute to the maintenance of the neuroinflammatory process in DRG and axons respectively. Moreover, neuroinflammatory components have also been shown in the spinal cord with microglia and astrocytes playing an important role in CIPN development. Taking together, better understanding of these aspects would permit the development of possible strategies in order to improve the management of CIPN.
Collapse
Affiliation(s)
- Giulia Fumagalli
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.,NeuroMI (Milan Center for Neuroscience), University of Milano-Bicocca, Monza, Italy
| | - Laura Monza
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.,NeuroMI (Milan Center for Neuroscience), University of Milano-Bicocca, Monza, Italy
| | - Guido Cavaletti
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.,NeuroMI (Milan Center for Neuroscience), University of Milano-Bicocca, Monza, Italy
| | - Roberta Rigolio
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.,NeuroMI (Milan Center for Neuroscience), University of Milano-Bicocca, Monza, Italy
| | - Cristina Meregalli
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.,NeuroMI (Milan Center for Neuroscience), University of Milano-Bicocca, Monza, Italy
| |
Collapse
|
36
|
Therapeutic Agents for Oxaliplatin-Induced Peripheral Neuropathy; Experimental and Clinical Evidence. Int J Mol Sci 2021; 22:1393. [PMID: 33573316 PMCID: PMC7866815 DOI: 10.3390/ijms22031393&set/a 813269399+839900579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Oxaliplatin is an essential drug in the chemotherapy of colorectal, gastric, and pancreatic cancers, but it frequently causes peripheral neuropathy as a dose-limiting factor. So far, animal models of oxaliplatin-induced peripheral neuropathy have been established. The mechanisms of development of neuropathy induced by oxaliplatin have been elucidated, and many drugs and agents have been proven to have neuroprotective effects in basic studies. In addition, some of these drugs have been validated in clinical studies for their inhibitory effects on neuropathy. In this review, we summarize the basic and clinical evidence for the therapeutic effects of oxaliplatin. In basic research, there are many reports of neuropathy inhibitors that target oxidative stress, inflammatory response, sodium channel, transient receptor potential (TRP) channel, glutamate nervous system, and monoamine nervous system. Alternatively, very few drugs have clearly demonstrated the efficacy for oxaliplatin-induced peripheral neuropathy in clinical trials. It is important to activate translational research in order to translate basic research into clinical research.
Collapse
|
37
|
Therapeutic Agents for Oxaliplatin-Induced Peripheral Neuropathy; Experimental and Clinical Evidence. Int J Mol Sci 2021. [DOI: 10.3390/ijms22031393
expr 945913974 + 948698388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Oxaliplatin is an essential drug in the chemotherapy of colorectal, gastric, and pancreatic cancers, but it frequently causes peripheral neuropathy as a dose-limiting factor. So far, animal models of oxaliplatin-induced peripheral neuropathy have been established. The mechanisms of development of neuropathy induced by oxaliplatin have been elucidated, and many drugs and agents have been proven to have neuroprotective effects in basic studies. In addition, some of these drugs have been validated in clinical studies for their inhibitory effects on neuropathy. In this review, we summarize the basic and clinical evidence for the therapeutic effects of oxaliplatin. In basic research, there are many reports of neuropathy inhibitors that target oxidative stress, inflammatory response, sodium channel, transient receptor potential (TRP) channel, glutamate nervous system, and monoamine nervous system. Alternatively, very few drugs have clearly demonstrated the efficacy for oxaliplatin-induced peripheral neuropathy in clinical trials. It is important to activate translational research in order to translate basic research into clinical research.
Collapse
|
38
|
Kawashiri T, Mine K, Kobayashi D, Inoue M, Ushio S, Uchida M, Egashira N, Shimazoe T. Therapeutic Agents for Oxaliplatin-Induced Peripheral Neuropathy; Experimental and Clinical Evidence. Int J Mol Sci 2021; 22:ijms22031393. [PMID: 33573316 PMCID: PMC7866815 DOI: 10.3390/ijms22031393] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/20/2021] [Accepted: 01/27/2021] [Indexed: 02/07/2023] Open
Abstract
Oxaliplatin is an essential drug in the chemotherapy of colorectal, gastric, and pancreatic cancers, but it frequently causes peripheral neuropathy as a dose-limiting factor. So far, animal models of oxaliplatin-induced peripheral neuropathy have been established. The mechanisms of development of neuropathy induced by oxaliplatin have been elucidated, and many drugs and agents have been proven to have neuroprotective effects in basic studies. In addition, some of these drugs have been validated in clinical studies for their inhibitory effects on neuropathy. In this review, we summarize the basic and clinical evidence for the therapeutic effects of oxaliplatin. In basic research, there are many reports of neuropathy inhibitors that target oxidative stress, inflammatory response, sodium channel, transient receptor potential (TRP) channel, glutamate nervous system, and monoamine nervous system. Alternatively, very few drugs have clearly demonstrated the efficacy for oxaliplatin-induced peripheral neuropathy in clinical trials. It is important to activate translational research in order to translate basic research into clinical research.
Collapse
Affiliation(s)
- Takehiro Kawashiri
- Department of Clinical Pharmacy and Pharmaceutical Care, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (K.M.); (D.K.); (M.I.); (T.S.)
- Correspondence: ; Tel.: +81-92-642-6573
| | - Keisuke Mine
- Department of Clinical Pharmacy and Pharmaceutical Care, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (K.M.); (D.K.); (M.I.); (T.S.)
| | - Daisuke Kobayashi
- Department of Clinical Pharmacy and Pharmaceutical Care, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (K.M.); (D.K.); (M.I.); (T.S.)
| | - Mizuki Inoue
- Department of Clinical Pharmacy and Pharmaceutical Care, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (K.M.); (D.K.); (M.I.); (T.S.)
| | - Soichiro Ushio
- Department of Pharmacy, Okayama University Hospital, Okayama 700-8558, Japan;
| | - Mayako Uchida
- Education and Research Center for Clinical Pharmacy, Osaka University of Pharmaceutical Sciences, Osaka 569-1094, Japan;
| | - Nobuaki Egashira
- Department of Pharmacy, Kyushu University Hospital, Fukuoka 812-8582, Japan;
| | - Takao Shimazoe
- Department of Clinical Pharmacy and Pharmaceutical Care, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (K.M.); (D.K.); (M.I.); (T.S.)
| |
Collapse
|
39
|
Nicotinic Acetylcholine Receptor Involvement in Inflammatory Bowel Disease and Interactions with Gut Microbiota. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18031189. [PMID: 33572734 PMCID: PMC7908252 DOI: 10.3390/ijerph18031189] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/22/2021] [Accepted: 01/25/2021] [Indexed: 12/12/2022]
Abstract
The gut-brain axis describes a complex interplay between the central nervous system and organs of the gastrointestinal tract. Sensory neurons of dorsal root and nodose ganglia, neurons of the autonomic nervous system, and immune cells collect and relay information about the status of the gut to the brain. A critical component in this bi-directional communication system is the vagus nerve which is essential for coordinating the immune system’s response to the activities of commensal bacteria in the gut and to pathogenic strains and their toxins. Local control of gut function is provided by networks of neurons in the enteric nervous system also called the ‘gut-brain’. One element common to all of these gut-brain systems is the expression of nicotinic acetylcholine receptors. These ligand-gated ion channels serve myriad roles in the gut-brain axis including mediating fast synaptic transmission between autonomic pre- and postganglionic neurons, modulation of neurotransmitter release from peripheral sensory and enteric neurons, and modulation of cytokine release from immune cells. Here we review the role of nicotinic receptors in the gut-brain axis with a focus on the interplay of these receptors with the gut microbiome and their involvement in dysregulation of gut function and inflammatory bowel diseases.
Collapse
|
40
|
Li X, Tae HS, Chu Y, Jiang T, Adams DJ, Yu R. Medicinal chemistry, pharmacology, and therapeutic potential of α-conotoxins antagonizing the α9α10 nicotinic acetylcholine receptor. Pharmacol Ther 2020; 222:107792. [PMID: 33309557 DOI: 10.1016/j.pharmthera.2020.107792] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 12/07/2020] [Indexed: 12/20/2022]
Abstract
α-Conotoxins are disulfide-rich and well-structured peptides, most of which can block nicotinic acetylcholine receptors (nAChRs) with exquisite selectivity and potency. There are various nAChR subtypes, of which the α9α10 nAChR functions as a heteromeric ionotropic receptor in the mammalian cochlea and mediates postsynaptic transmission from the medial olivocochlear. The α9α10 nAChR subtype has also been proposed as a target for the treatment of neuropathic pain and the suppression of breast cancer cell proliferation. Therefore, α-conotoxins targeting the α9α10 nAChR are potentially useful in the development of specific therapeutic drugs and pharmacological tools. Despite dissimilarities in their amino acid sequence and structures, these conopeptides are potent antagonists of the α9α10 nAChR subtype. Consequently, the activity and stability of these peptides have been subjected to chemical modifications. The resulting synthetic analogues have not only functioned as molecular probes to explore ligand binding sites of the α9α10 nAChR, but also have the potential to become candidates for drug development. From the perspectives of medicinal chemistry and pharmacology, we highlight the structure and function of the α9α10 nAChR and review studies of α-conotoxins targeting it, including their three-dimensional structures, structure optimization strategies, and binding modes at the α9α10 nAChR, as well as their therapeutic potential.
Collapse
Affiliation(s)
- Xiao Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China; Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266003, China
| | - Han-Shen Tae
- Illawarra Health and Medical Research Institute (IHMRI), University of Wollongong, Wollongong, New South Wales 2522, Australia
| | - Yanyan Chu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China; Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266003, China; Innovation Platform of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, Shandong 266100, China
| | - Tao Jiang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China; Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266003, China
| | - David J Adams
- Illawarra Health and Medical Research Institute (IHMRI), University of Wollongong, Wollongong, New South Wales 2522, Australia.
| | - Rilei Yu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China; Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266003, China; Innovation Platform of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, Shandong 266100, China.
| |
Collapse
|
41
|
Huynh PN, Harvey PJ, Gajewiak J, Craik DJ, Michael McIntosh J. Critical residue properties for potency and selectivity of α-Conotoxin RgIA towards α9α10 nicotinic acetylcholine receptors. Biochem Pharmacol 2020; 181:114124. [PMID: 32593612 PMCID: PMC7572646 DOI: 10.1016/j.bcp.2020.114124] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/20/2020] [Accepted: 06/23/2020] [Indexed: 12/24/2022]
Abstract
The α9α10 nicotinic acetylcholine receptor (nAChR) has been characterized as an effective anti-pain target that functions through a non-opioid mechanism. However, as a pentameric ion channel comprised of two different subunits, the specific targeting of α9α10 nAChRs has proven challenging. Previously the 13-amino-acid peptide, RgIA, was shown to block α9α10 nAChRs with high potency and specificity. This peptide, characterized from the venom of the carnivorous marine snail, Conus regius, produced analgesia in several rodent models of chronic pain. Despite promising pre-clinical data in behavioral assays, the number of specific α9α10 nAChR antagonists remains small and the physiological mechanisms of analgesia remain cryptic. In this study, we implement amino-acid substitutions to definitively characterize the chemical properties of RgIA that contribute to its activity against α9α10 nAChRs. Using this mutational approach, we determined the vital role of biochemical side-chain properties and amino acids in the second loop that are amenable to substitutions to further engineer next-generation analogs for the blockade of α9α10 nAChRs.
Collapse
Affiliation(s)
- Peter N Huynh
- School of Biological Sciences, University of Utah, Salt Lake City, UT 84112, USA.
| | - Peta J Harvey
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Joanna Gajewiak
- School of Biological Sciences, University of Utah, Salt Lake City, UT 84112, USA
| | - David J Craik
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - J Michael McIntosh
- School of Biological Sciences, University of Utah, Salt Lake City, UT 84112, USA; George E. Whalen Veterans Affairs Medical Center, Salt Lake City, UT 84112, USA; Department of Psychiatry, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
42
|
Lee JH, Kim W. The Role of Satellite Glial Cells, Astrocytes, and Microglia in Oxaliplatin-Induced Neuropathic Pain. Biomedicines 2020; 8:E324. [PMID: 32887259 PMCID: PMC7554902 DOI: 10.3390/biomedicines8090324] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 08/30/2020] [Accepted: 08/31/2020] [Indexed: 12/17/2022] Open
Abstract
Oxaliplatin is a third-generation platinum-based chemotherapeutic drug. Although its efficacy against colorectal cancer is well known, peripheral neuropathy that develops during and after infusion of the agents could decrease the quality of life of the patients. Various pathways have been reported to be the cause of the oxaliplatin-induced paresthesia and dysesthesia; however, its mechanism of action has not been fully understood yet. In recent years, researchers have investigated the function of glia in pain, and demonstrated that glia in the peripheral and central nervous system could play a critical role in the development and maintenance of neuropathic pain. These results suggest that targeting the glia may be an effective therapeutic option. In the past ten years, 20 more papers focused on the role of glia in oxaliplatin-induced thermal and mechanical hypersensitivity. However, to date no review has been written to summarize and discuss their results. Thus, in this study, by reviewing 23 studies that conducted in vivo experiments in rodents, the change of satellite glial cells, astrocytes, and microglia activation in the dorsal root ganglia, spinal cord, and the brain of oxaliplatin-induced neuropathic pain animals is discussed.
Collapse
Affiliation(s)
| | - Woojin Kim
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul 02453, Korea;
| |
Collapse
|
43
|
Lucarini E, Parisio C, Branca JJV, Segnani C, Ippolito C, Pellegrini C, Antonioli L, Fornai M, Micheli L, Pacini A, Bernardini N, Blandizzi C, Ghelardini C, Di Cesare Mannelli L. Deepening the Mechanisms of Visceral Pain Persistence: An Evaluation of the Gut-Spinal Cord Relationship. Cells 2020; 9:cells9081772. [PMID: 32722246 PMCID: PMC7464824 DOI: 10.3390/cells9081772] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/11/2020] [Accepted: 07/20/2020] [Indexed: 12/11/2022] Open
Abstract
The management of visceral pain is a major clinical problem in patients affected by gastrointestinal disorders. The poor knowledge about pain chronicization mechanisms prompted us to study the functional and morphological alterations of the gut and nervous system in the animal model of persistent visceral pain caused by 2,4-dinitrobenzenesulfonic acid (DNBS). This agent, injected intrarectally, induced a colonic inflammation peaking on day 3 and remitting progressively from day 7. In concomitance with bowel inflammation, the animals developed visceral hypersensitivity, which persisted after colitis remission for up to three months. On day 14, the administration of pain-relieving drugs (injected intraperitoneally and intrathecally) revealed a mixed nociceptive, inflammatory and neuropathic pain originating from both the peripheral and central nervous system. At this time point, the colonic histological analysis highlighted a partial restitution of the tunica mucosa, transmural collagen deposition, infiltration of mast cells and eosinophils, and upregulation of substance P (SP)-positive nerve fibers, which were surrounded by eosinophils and MHC-II-positive macrophages. A significant activation of microglia and astrocytes was observed in the dorsal and ventral horns of spinal cord. These results suggest that the persistence of visceral pain induced by colitis results from maladaptive plasticity of the enteric, peripheral and central nervous systems.
Collapse
Affiliation(s)
- Elena Lucarini
- Department of Neuroscience, Psychology, Drug Research and Child Health, Neurofarba, Pharmacology and Toxicology Section, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (E.L.); (C.P.); (L.M.); (C.G.)
| | - Carmen Parisio
- Department of Neuroscience, Psychology, Drug Research and Child Health, Neurofarba, Pharmacology and Toxicology Section, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (E.L.); (C.P.); (L.M.); (C.G.)
| | - Jacopo J. V. Branca
- Department of Experimental and Clinical Medicine—DMSC, Anatomy and Histology Section, University of Florence, L. go Brambilla 3, 50134 Florence, Italy; (J.J.V.B.); (A.P.)
| | - Cristina Segnani
- Department of Clinical and Experimental Medicine, Unit of Histology, University of Pisa, 56126 Pisa, Italy; (C.S.); (C.I.); (N.B.)
| | - Chiara Ippolito
- Department of Clinical and Experimental Medicine, Unit of Histology, University of Pisa, 56126 Pisa, Italy; (C.S.); (C.I.); (N.B.)
| | - Carolina Pellegrini
- Department of Pharmacy, Unit of Pharmacology, University of Pisa, 56126 Pisa, Italy;
| | - Luca Antonioli
- Department of Clinical and Experimental Medicine, Unit of Pharmacology and Pharmacovigilance, University of Pisa, 56126 Pisa, Italy; (L.A.); (M.F.); (C.B.)
| | - Matteo Fornai
- Department of Clinical and Experimental Medicine, Unit of Pharmacology and Pharmacovigilance, University of Pisa, 56126 Pisa, Italy; (L.A.); (M.F.); (C.B.)
| | - Laura Micheli
- Department of Neuroscience, Psychology, Drug Research and Child Health, Neurofarba, Pharmacology and Toxicology Section, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (E.L.); (C.P.); (L.M.); (C.G.)
| | - Alessandra Pacini
- Department of Experimental and Clinical Medicine—DMSC, Anatomy and Histology Section, University of Florence, L. go Brambilla 3, 50134 Florence, Italy; (J.J.V.B.); (A.P.)
| | - Nunzia Bernardini
- Department of Clinical and Experimental Medicine, Unit of Histology, University of Pisa, 56126 Pisa, Italy; (C.S.); (C.I.); (N.B.)
- Interdepartmental Research Center “Nutraceuticals and Food for Health”, University of Pisa, 56126 Pisa, Italy
| | - Corrado Blandizzi
- Department of Clinical and Experimental Medicine, Unit of Pharmacology and Pharmacovigilance, University of Pisa, 56126 Pisa, Italy; (L.A.); (M.F.); (C.B.)
| | - Carla Ghelardini
- Department of Neuroscience, Psychology, Drug Research and Child Health, Neurofarba, Pharmacology and Toxicology Section, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (E.L.); (C.P.); (L.M.); (C.G.)
| | - Lorenzo Di Cesare Mannelli
- Department of Neuroscience, Psychology, Drug Research and Child Health, Neurofarba, Pharmacology and Toxicology Section, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (E.L.); (C.P.); (L.M.); (C.G.)
- Correspondence:
| |
Collapse
|
44
|
AlSharari SD, Toma W, Mahmood HM, Michael McIntosh J, Imad Damaj M. The α9α10 nicotinic acetylcholine receptors antagonist α-conotoxin RgIA reverses colitis signs in murine dextran sodium sulfate model. Eur J Pharmacol 2020; 883:173320. [PMID: 32645334 DOI: 10.1016/j.ejphar.2020.173320] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 06/24/2020] [Accepted: 06/24/2020] [Indexed: 01/03/2023]
Abstract
Nicotinic acetylcholine receptors can regulate inflammation primarily through the vagus nerve via the cholinergic anti-inflammatory pathway. α9α10 nicotinic receptors (nAChRs) are a new promising target for chronic pain and inflammation. Recently, α9α10 selective α-conotoxin antagonists were shown to have antinociception effect in neuropathic and tonic inflammatory pain animal models. However, limited data available on the role of α9α10 nAChRs in experimental colitis. In this study, we report for the first time, the role of α9α10 nAChRs in the dextran sodium sulfate (DSS) experimental animal colitis model. We determined the effect of the α9α10 nAChRs antagonist, α-conotoxin RgIA (α-RgIA) in DSS-induced colitis model in adult male and female C57BL/6 J mice. DSS solution was freely given in the drinking water for seven consecutive days, and tap water was given on the 8th day. We then sacrificed mice on day 8 to examine the entire colon. Disease severity, colon tissue histology, and tumor necrosis factor-α (TNF-α) were evaluated. The lower doses (0.02 and 0.1 nmol/mouse, s.c.) of α-RgIA treatment in DSS-treated mice were inactive, whereas the higher dose (0.2 nmol/mouse, s.c.) reversed the disease activity index (DAI) score, loss of body weight, total histological damage score, as well as the colonic level of TNF-α compared to the DSS-control group. Moreover, the highest dose of α-RgIA (0.2 nmol/mouse, s.c.) significantly rescued the colon length shortening in DSS-treated mice compared to the DSS-control mice. The availability of α9*-selective conotoxins has opened new avenues in pharmacology research and potential targets in inflammatory disorders.
Collapse
Affiliation(s)
- Shakir D AlSharari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia; Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Wisam Toma
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, 23298, USA.
| | - Hafiz M Mahmood
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - J Michael McIntosh
- George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, UT, 84108, USA; Departments of Psychiatry and Biology, University of Utah, Salt Lake City, UT, 84112, USA
| | - M Imad Damaj
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, 23298, USA
| |
Collapse
|
45
|
Isolation, Characterization and In-Silico Study of Conotoxin Protein from Conus loroisii and Its Anti-cancer Activity. Int J Pept Res Ther 2020. [DOI: 10.1007/s10989-020-10091-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
46
|
Mussina K, Toktarkhanova D, Filchakova O. Nicotinic Acetylcholine Receptors of PC12 Cells. Cell Mol Neurobiol 2020; 41:17-29. [PMID: 32335772 DOI: 10.1007/s10571-020-00846-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Accepted: 04/09/2020] [Indexed: 12/18/2022]
Abstract
Nicotinic acetylcholine receptors (nAChRs) have gained much attention in the scientific community since they play a significant role in multiple physiological and pathophysiological processes. Multiple approaches to study the receptors exist, with characterization of the receptors' functionality at a single cellular level using cell culturing being one of them. Derived from an adrenal medulla tumor, PC12 cells express nicotinic receptor subunits and form functional nicotinic receptors. Thus, the cells offer a convenient environment to address questions related to the functionality of the receptors. The review summarizes the findings on nicotinic receptors' expression and functions which were conducted using PC12 cells. Specific focus is given to α3-containing receptors as well as α7 receptor. Critical evaluation of findings is provided alongside insights into what can still be learned about nAChRs, using PC12 cells.
Collapse
Affiliation(s)
- Kamilla Mussina
- Biology Department, School of Sciences and Humanities, Nazarbayev University, NurSultan, Republic of Kazakhstan
| | - Dana Toktarkhanova
- Biology Department, School of Sciences and Humanities, Nazarbayev University, NurSultan, Republic of Kazakhstan
| | - Olena Filchakova
- Biology Department, School of Sciences and Humanities, Nazarbayev University, NurSultan, Republic of Kazakhstan.
| |
Collapse
|
47
|
Toma W, Ulker E, Alqasem M, AlSharari SD, McIntosh JM, Damaj MI. Behavioral and Molecular Basis of Cholinergic Modulation of Pain: Focus on Nicotinic Acetylcholine Receptors. Curr Top Behav Neurosci 2020; 45:153-166. [PMID: 32468494 DOI: 10.1007/7854_2020_135] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Nicotinic acetylcholine receptors (nAChRs) have emerged as a novel therapeutic strategy for pain and inflammatory disorders. In particular, α4β2∗, α7, and α9α10 nAChR subtypes have been investigated as potential targets to treat pain. The nAChRs are distributed on the pain transmission pathways, including central and peripheral nervous systems and immune cells as well. Several agonists for α4β2∗ nAChR subtypes have been investigated in multiple animal pain models with promising results. However, studies in human indicated a narrow therapeutic window for α4β2∗ agonists. Furthermore, animal studies suggest that using agonists for α7 nAChR subtype and antagonists for α9α10 nAChR subtypes are potential novel therapies for chronic pain management, including inflammatory and neuropathic pain. More recently, alternative nAChRs ligands such as positive allosteric modulators and silent agonists have shown potential to develop into new treatments for chronic pain.
Collapse
Affiliation(s)
- Wisam Toma
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Esad Ulker
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
| | - Mashael Alqasem
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Shakir D AlSharari
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - J Michael McIntosh
- Departments of Psychiatry and Biology, University of Utah, Salt Lake City, UT, USA
| | - M Imad Damaj
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
48
|
RgIA4 Accelerates Recovery from Paclitaxel-Induced Neuropathic Pain in Rats. Mar Drugs 2019; 18:md18010012. [PMID: 31877728 PMCID: PMC7024385 DOI: 10.3390/md18010012] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 12/16/2019] [Accepted: 12/18/2019] [Indexed: 02/06/2023] Open
Abstract
Chemotherapeutic drugs are widely utilized in the treatment of human cancers. Painful chemotherapy-induced neuropathy is a common, debilitating, and dose-limiting side effect for which there is currently no effective treatment. Previous studies have demonstrated the potential utility of peptides from the marine snail from the genus Conus for the treatment of neuropathic pain. α-Conotoxin RgIA and a potent analog, RgIA4, have previously been shown to prevent the development of neuropathy resulting from the administration of oxaliplatin, a platinum-based antineoplastic drug. Here, we have examined its efficacy against paclitaxel, a chemotherapeutic drug that works by a mechanism of action distinct from that of oxaliplatin. Paclitaxel was administered at 2 mg/kg (intraperitoneally (IP)) every other day for a total of 8 mg/kg. Sprague Dawley rats that were co-administered RgIA4 at 80 µg/kg (subcutaneously (SC)) once daily, five times per week, for three weeks showed significant recovery from mechanical allodynia by day 31. Notably, the therapeutic effects reached significance 12 days after the last administration of RgIA4, which is suggestive of a rescue mechanism. These findings support the effects of RgIA4 in multiple chemotherapeutic models and the investigation of α9α10 nicotinic acetylcholine receptors (nAChRs) as a non-opioid target in the treatment of chronic pain.
Collapse
|
49
|
Marine Toxins and Nociception: Potential Therapeutic Use in the Treatment of Visceral Pain Associated with Gastrointestinal Disorders. Toxins (Basel) 2019; 11:toxins11080449. [PMID: 31370176 PMCID: PMC6723473 DOI: 10.3390/toxins11080449] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 07/24/2019] [Accepted: 07/26/2019] [Indexed: 12/12/2022] Open
Abstract
Visceral pain, of which the pathogenic basis is currently largely unknown, is a hallmark symptom of both functional disorders, such as irritable bowel syndrome, and inflammatory bowel disease. Intrinsic sensory neurons in the enteric nervous system and afferent sensory neurons of the dorsal root ganglia, connecting with the central nervous system, represent the primary neuronal pathways transducing gut visceral pain. Current pharmacological therapies have several limitations, owing to their partial efficacy and the generation of severe adverse effects. Numerous cellular targets of visceral nociception have been recognized, including, among others, channels (i.e., voltage-gated sodium channels, VGSCs, voltage-gated calcium channels, VGCCs, Transient Receptor Potential, TRP, and Acid-sensing ion channels, ASICs) and neurotransmitter pathways (i.e., GABAergic pathways), which represent attractive targets for the discovery of novel drugs. Natural biologically active compounds, such as marine toxins, able to bind with high affinity and selectivity to different visceral pain molecular mediators, may represent a useful tool (1) to improve our knowledge of the physiological and pathological relevance of each nociceptive target, and (2) to discover therapeutically valuable molecules. In this review we report the most recent literature describing the effects of marine toxin on gastrointestinal visceral pain pathways and the possible clinical implications in the treatment of chronic pain associated with gut diseases.
Collapse
|
50
|
Hu S, Huang KM, Adams EJ, Loprinzi CL, Lustberg MB. Recent Developments of Novel Pharmacologic Therapeutics for Prevention of Chemotherapy-Induced Peripheral Neuropathy. Clin Cancer Res 2019; 25:6295-6301. [PMID: 31123053 DOI: 10.1158/1078-0432.ccr-18-2152] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 04/17/2019] [Accepted: 05/17/2019] [Indexed: 12/11/2022]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a common and dose-limiting toxicity, negatively affecting both quality of life and disease outcomes. To date, there is no proven preventative strategy for CIPN. Although multiple randomized trials have evaluated a variety of pharmacologic interventions for the treatment of CIPN, only duloxetine has shown clear efficacy in a phase III study. The National Cancer Institute's Symptom Management and Health-Related Quality of Life Steering Committee has identified CIPN as a priority for translational research in cancer care. Promising advances in preclinical research have identified several novel preventative and therapeutic targets, which have the potential to transform the care of patients with this debilitating neurotoxicity. Here, we provide an overarching view of emerging strategies and therapeutic targets that are currently being evaluated in CIPN.
Collapse
Affiliation(s)
- Shuiying Hu
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Kevin M Huang
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Elizabeth J Adams
- Department of Medical Oncology, The Ohio State University, Comprehensive Cancer Center, Columbus, Ohio
| | | | - Maryam B Lustberg
- Department of Medical Oncology, The Ohio State University, Comprehensive Cancer Center, Columbus, Ohio.
| |
Collapse
|