1
|
Le Franc A, Da Silva A, Lepetre-Mouelhi S. Nanomedicine and voltage-gated sodium channel blockers in pain management: a game changer or a lost cause? Drug Deliv Transl Res 2024; 14:2112-2145. [PMID: 38861139 DOI: 10.1007/s13346-024-01615-9] [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] [Accepted: 04/25/2024] [Indexed: 06/12/2024]
Abstract
Pain, a complex and debilitating condition affecting millions globally, is a significant concern, especially in the context of post-operative recovery. This comprehensive review explores the complexity of pain and its global impact, emphasizing the modulation of voltage-gated sodium channels (VGSC or NaV channels) as a promising avenue for pain management with the aim of reducing reliance on opioids. The article delves into the role of specific NaV isoforms, particularly NaV 1.7, NaV 1.8, and NaV 1.9, in pain process and discusses the development of sodium channel blockers to target these isoforms precisely. Traditional local anesthetics and selective NaV isoform inhibitors, despite showing varying efficacy in pain management, face challenges in systemic distribution and potential side effects. The review highlights the potential of nanomedicine in improving the delivery of local anesthetics, toxins and selective NaV isoform inhibitors for a targeted and sustained release at the site of pain. This innovative strategy seeks to improve drug bioavailability, minimize systemic exposure, and optimize therapeutic outcomes, holding significant promise for secure pain management and enhancing the quality of life for individuals recovering from surgical procedures or suffering from chronic pain.
Collapse
Affiliation(s)
- Adélaïde Le Franc
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400, Orsay, France
| | - Alexandre Da Silva
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400, Orsay, France
| | | |
Collapse
|
2
|
Harris BJ, Nguyen PT, Zhou G, Wulff H, DiMaio F, Yarov-Yarovoy V. Toward high-resolution modeling of small molecule-ion channel interactions. Front Pharmacol 2024; 15:1411428. [PMID: 38919257 PMCID: PMC11196768 DOI: 10.3389/fphar.2024.1411428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 05/13/2024] [Indexed: 06/27/2024] Open
Abstract
Ion channels are critical drug targets for a range of pathologies, such as epilepsy, pain, itch, autoimmunity, and cardiac arrhythmias. To develop effective and safe therapeutics, it is necessary to design small molecules with high potency and selectivity for specific ion channel subtypes. There has been increasing implementation of structure-guided drug design for the development of small molecules targeting ion channels. We evaluated the performance of two RosettaLigand docking methods, RosettaLigand and GALigandDock, on the structures of known ligand-cation channel complexes. Ligands were docked to voltage-gated sodium (NaV), voltage-gated calcium (CaV), and transient receptor potential vanilloid (TRPV) channel families. For each test case, RosettaLigand and GALigandDock methods frequently sampled a ligand-binding pose within a root mean square deviation (RMSD) of 1-2 Å relative to the experimental ligand coordinates. However, RosettaLigand and GALigandDock scoring functions cannot consistently identify experimental ligand coordinates as top-scoring models. Our study reveals that the proper scoring criteria for RosettaLigand and GALigandDock modeling of ligand-ion channel complexes should be assessed on a case-by-case basis using sufficient ligand and receptor interface sampling, knowledge about state-specific interactions of the ion channel, and inherent receptor site flexibility that could influence ligand binding.
Collapse
Affiliation(s)
- Brandon J. Harris
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, United States
- Biophysics Graduate Group, University of California, Davis, Davis, CA, United States
| | - Phuong T. Nguyen
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, United States
| | - Guangfeng Zhou
- Department of Biochemistry, University of Washington, Seattle, WA, United States
- Institute for Protein Design, University of Washington, Seattle, WA, United States
| | - Heike Wulff
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Frank DiMaio
- Department of Biochemistry, University of Washington, Seattle, WA, United States
| | - Vladimir Yarov-Yarovoy
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA, United States
- Biophysics Graduate Group, University of California, Davis, Davis, CA, United States
- Department of Anesthesiology and Pain Medicine, University of California, Davis, Davis, CA, United States
| |
Collapse
|
3
|
Rao R, Mohammed C, Alschuler L, Pomeranz Krummel DA, Sengupta S. Phytochemical Modulation of Ion Channels in Oncologic Symptomatology and Treatment. Cancers (Basel) 2024; 16:1786. [PMID: 38730738 PMCID: PMC11083444 DOI: 10.3390/cancers16091786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 04/24/2024] [Accepted: 05/04/2024] [Indexed: 05/13/2024] Open
Abstract
Modern chemotherapies offer a broad approach to cancer treatment but eliminate both cancer and non-cancer cells indiscriminately and, thus, are associated with a host of side effects. Advances in precision oncology have brought about new targeted therapeutics, albeit mostly limited to a subset of patients with an actionable mutation. They too come with side effects and, ultimately, 'self-resistance' to the treatment. There is recent interest in the modulation of ion channels, transmembrane proteins that regulate the flow of electrically charged molecules in and out of cells, as an approach to aid treatment of cancer. Phytochemicals have been shown to act on ion channels with high specificity regardless of the tumor's genetic profile. This paper explores the use of phytochemicals in cancer symptom management and treatment.
Collapse
Affiliation(s)
- Rohan Rao
- Department of Neurology & Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Caroline Mohammed
- Department of Neurology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Lise Alschuler
- Andrew Weil Center for Integrative Medicine, University of Arizona College of Medicine, Tucson, AZ 85719, USA
| | - Daniel A. Pomeranz Krummel
- Department of Neurosurgery, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Soma Sengupta
- Department of Neurology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
- Department of Neurosurgery, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| |
Collapse
|
4
|
Antoniazzi CTDD, Ruviaro NA, Peres DS, Rodrigues P, Viero FT, Trevisan G. Targeting TRPV4 Channels for Cancer Pain Relief. Cancers (Basel) 2024; 16:1703. [PMID: 38730655 PMCID: PMC11083562 DOI: 10.3390/cancers16091703] [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: 03/29/2024] [Revised: 04/25/2024] [Accepted: 04/26/2024] [Indexed: 05/13/2024] Open
Abstract
Despite the unique and complex nature of cancer pain, the activation of different ion channels can be related to the initiation and maintenance of pain. The transient receptor potential vanilloid 4 (TRPV4) is a cation channel broadly expressed in sensory afferent neurons. This channel is activated by multiple stimuli to mediate pain perception associated with inflammatory and neuropathic pain. Here, we focused on summarizing the role of TRPV4 in cancer etiology and cancer-induced pain mechanisms. Many studies revealed that the administration of a TRPV4 antagonist and TRPV4 knockdown diminishes nociception in chemotherapy-induced peripheral neuropathy (CIPN). Although the evidence on TRPV4 channels' involvement in cancer pain is scarce, the expression of these receptors was reportedly enhanced in cancer-induced bone pain (CIBP), perineural, and orofacial cancer models following the inoculation of tumor cells to the bone marrow cavity, sciatic nerve, and tongue, respectively. Effective pain management is a continuous problem for patients diagnosed with cancer, and current guidelines fail to address a mechanism-based treatment. Therefore, examining new molecules with potential antinociceptive properties targeting TRPV4 modulation would be interesting. Identifying such agents could lead to the development of treatment strategies with improved pain-relieving effects and fewer adverse effects than the currently available analgesics.
Collapse
Affiliation(s)
- Caren Tatiane de David Antoniazzi
- Graduate Program in Pharmacology, Federal University of Santa Maria (UFSM), Santa Maria 97105-900, Brazil; (C.T.d.D.A.); (D.S.P.); (P.R.); (F.T.V.)
| | - Náthaly Andrighetto Ruviaro
- Graduate Program in Toxicological Biochemistry, Federal University of Santa Maria (UFSM), Santa Maria 97105-900, Brazil;
| | - Diulle Spat Peres
- Graduate Program in Pharmacology, Federal University of Santa Maria (UFSM), Santa Maria 97105-900, Brazil; (C.T.d.D.A.); (D.S.P.); (P.R.); (F.T.V.)
| | - Patrícia Rodrigues
- Graduate Program in Pharmacology, Federal University of Santa Maria (UFSM), Santa Maria 97105-900, Brazil; (C.T.d.D.A.); (D.S.P.); (P.R.); (F.T.V.)
| | - Fernanda Tibolla Viero
- Graduate Program in Pharmacology, Federal University of Santa Maria (UFSM), Santa Maria 97105-900, Brazil; (C.T.d.D.A.); (D.S.P.); (P.R.); (F.T.V.)
| | - Gabriela Trevisan
- Graduate Program in Pharmacology, Federal University of Santa Maria (UFSM), Santa Maria 97105-900, Brazil; (C.T.d.D.A.); (D.S.P.); (P.R.); (F.T.V.)
- Graduate Program in Toxicological Biochemistry, Federal University of Santa Maria (UFSM), Santa Maria 97105-900, Brazil;
| |
Collapse
|
5
|
Chen P, Wang J, Zhang S, Wang Y, Sun Y, Bai S, Wu Q, Cheng X, Cao P, Qi X. Total syntheses of Tetrodotoxin and 9-epiTetrodotoxin. Nat Commun 2024; 15:679. [PMID: 38263179 PMCID: PMC10806222 DOI: 10.1038/s41467-024-45037-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 01/09/2024] [Indexed: 01/25/2024] Open
Abstract
Tetrodotoxin and congeners are specific voltage-gated sodium channel blockers that exhibit remarkable anesthetic and analgesic effects. Here, we present a scalable asymmetric syntheses of Tetrodotoxin and 9-epiTetrodotoxin from the abundant chemical feedstock furfuryl alcohol. The optically pure cyclohexane skeleton is assembled via a stereoselective Diels-Alder reaction. The dense heteroatom substituents are established sequentially by a series of functional group interconversions on highly oxygenated cyclohexane frameworks, including a chemoselective cyclic anhydride opening, and a decarboxylative hydroxylation. An innovative SmI2-mediated concurrent fragmentation, an oxo-bridge ring opening and ester reduction followed by an Upjohn dihydroxylation deliver the highly oxidized skeleton. Ruthenium-catalyzed oxidative alkyne cleavage and formation of the hemiaminal and orthoester under acidic conditions enable the rapid assembly of Tetrodotoxin, anhydro-Tetrodotoxin, 9-epiTetrodotoxin, and 9-epi lactone-Tetrodotoxin.
Collapse
Affiliation(s)
- Peihao Chen
- School of Life Sciences, Peking University, Beijing, 100871, China
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, Beijing, 102206, China
| | - Jing Wang
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, Beijing, 102206, China
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, 100084, China
| | - Shuangfeng Zhang
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, Beijing, 102206, China
| | - Yan Wang
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, Beijing, 102206, China
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, 100084, China
| | - Yuze Sun
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, Beijing, 102206, China
| | - Songlin Bai
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, Beijing, 102206, China
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, 100084, China
| | - Qingcui Wu
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, Beijing, 102206, China
| | - Xinyu Cheng
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, Beijing, 102206, China
- National Institute of Biological Sciences, Chinese Academy of Medical Sciences&Peking Union Medical College, Beijing, 100730, China
| | - Peng Cao
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, Beijing, 102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, 100084, China
| | - Xiangbing Qi
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, Beijing, 102206, China.
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
6
|
Montuori E, De Luca D, Penna A, Stalberga D, Lauritano C. Alexandrium spp.: From Toxicity to Potential Biotechnological Benefits. Mar Drugs 2023; 22:31. [PMID: 38248656 PMCID: PMC10821459 DOI: 10.3390/md22010031] [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: 11/10/2023] [Revised: 12/27/2023] [Accepted: 12/28/2023] [Indexed: 01/23/2024] Open
Abstract
Many dinoflagellates of the genus Alexandrium are well known for being responsible for harmful algal blooms (HABs), producing potent toxins that cause damages to other marine organisms, aquaculture, fishery, tourism, as well as induce human intoxications and even death after consumption of contaminated shellfish or fish. In this review, we summarize potential bioprospecting associated to the genus Alexandrium, including which Alexandrium spp. produce metabolites with anticancer, antimicrobial, antiviral, as well as anti-Alzheimer applications. When available, we report their mechanisms of action and targets. We also discuss recent progress on the identification of secondary metabolites with biological properties favorable to human health and aquaculture. Altogether, this information highlights the importance of studying which culturing conditions induce the activation of enzymatic pathways responsible for the synthesis of bioactive metabolites. It also suggests considering and comparing clones collected in different locations for toxin monitoring and marine bioprospecting. This review can be of interest not only for the scientific community, but also for the entire population and industries.
Collapse
Affiliation(s)
- Eleonora Montuori
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy;
- Department of Ecosustainable Marine Biotechnology, Stazione Zoologica Anton Dohrn, Via Acton 55, 80133 Napoli, Italy
| | - Daniele De Luca
- Research Infrastructure for Marine Biological Resources Department, Stazione Zoologica Anton Dohrn, Villa Comunale, 80121 Napoli, Italy;
| | - Antonella Penna
- Department of Biomolecular Sciences, University of Urbino, Campus E. Mattei, 61029 Urbino, Italy;
| | - Darta Stalberga
- Department of Biomedical and Clinical Sciences, Division of Clinical Chemistry and Pharmacology, Linköping University, SE-58183 Linköping, Sweden;
| | - Chiara Lauritano
- Department of Ecosustainable Marine Biotechnology, Stazione Zoologica Anton Dohrn, Via Acton 55, 80133 Napoli, Italy
| |
Collapse
|
7
|
Wright K, Jiang H, Xia W, Murphy MB, Boronina TN, Nwafor JN, Kim H, Iheanacho AM, Azurmendi PA, Cole RN, Cole PA, Gabelli SB. The C-Terminal of Na V1.7 Is Ubiquitinated by NEDD4L. ACS BIO & MED CHEM AU 2023; 3:516-527. [PMID: 38144259 PMCID: PMC10739247 DOI: 10.1021/acsbiomedchemau.3c00031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 09/10/2023] [Accepted: 09/25/2023] [Indexed: 12/26/2023]
Abstract
NaV1.7, the neuronal voltage-gated sodium channel isoform, plays an important role in the human body's ability to feel pain. Mutations within NaV1.7 have been linked to pain-related syndromes, such as insensitivity to pain. To date, the regulation and internalization mechanisms of the NaV1.7 channel are not well known at a biochemical level. In this study, we perform biochemical and biophysical analyses that establish that the HECT-type E3 ligase, NEDD4L, ubiquitinates the cytoplasmic C-terminal (CT) region of NaV1.7. Through in vitro ubiquitination and mass spectrometry experiments, we identify, for the first time, the lysine residues of NaV1.7 within the CT region that get ubiquitinated. Furthermore, binding studies with an NEDD4L E3 ligase modulator (ubiquitin variant) highlight the dynamic partnership between NEDD4L and NaV1.7. These investigations provide a framework for understanding how NEDD4L-dependent regulation of the channel can influence the NaV1.7 function.
Collapse
Affiliation(s)
- Katharine
M. Wright
- Department
of Biophysics and Biophysical Chemistry, The Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Hanjie Jiang
- Division
of Genetics, Department of Medicine, Brigham
and Women’s Hospital, Boston, Massachusetts 02115, United States
- Department
of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, United States
- Department
of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Wendy Xia
- Department
of Biophysics and Biophysical Chemistry, The Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | | | - Tatiana N. Boronina
- Mass
Spectrometry
and Proteomics Facility, Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Justin N. Nwafor
- Department
of Biophysics and Biophysical Chemistry, The Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - HyoJeon Kim
- Division
of Genetics, Department of Medicine, Brigham
and Women’s Hospital, Boston, Massachusetts 02115, United States
- Department
of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Akunna M. Iheanacho
- Department
of Biophysics and Biophysical Chemistry, The Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
- Department
of Physiology, The Johns Hopkins School
of Medicine, Baltimore, Maryland 21205, United States
| | - P. Aitana Azurmendi
- Department
of Biophysics and Biophysical Chemistry, The Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Robert N. Cole
- Mass
Spectrometry
and Proteomics Facility, Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Philip A. Cole
- Division
of Genetics, Department of Medicine, Brigham
and Women’s Hospital, Boston, Massachusetts 02115, United States
- Department
of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Sandra B. Gabelli
- Department
of Biophysics and Biophysical Chemistry, The Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
- Department
of Medicine, The Johns Hopkins University
School of Medicine, Baltimore, Maryland 21205, United States
- Department
of Oncology, The Johns Hopkins University
School of Medicine, Baltimore, Maryland 21287, United States
| |
Collapse
|
8
|
Anastasiou TI, Kagiampaki E, Kondylatos G, Tselepides A, Peristeraki P, Mandalakis M. Assessing the Toxicity of Lagocephalus sceleratus Pufferfish from the Southeastern Aegean Sea and the Relationship of Tetrodotoxin with Gonadal Hormones. Mar Drugs 2023; 21:520. [PMID: 37888455 PMCID: PMC10608560 DOI: 10.3390/md21100520] [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: 09/03/2023] [Revised: 09/26/2023] [Accepted: 09/27/2023] [Indexed: 10/28/2023] Open
Abstract
Given the dramatic increase in the L. sceleratus population in the southeastern Aegean Sea, there is growing interest in assessing the toxicity of this pufferfish and the factors controlling its tetrodotoxin (TTX) content. In the present study, liver, gonads, muscle and skin of 37 L. sceleratus specimens collected during May and June 2021 from the island of Rhodes, Greece, were subjected to multi-analyte profiling using liquid chromatography-tandem mass spectrometry (LC-MS/MS) in order to quantitate TTX and evaluate whether this biotoxin interrelates with hormones. TTX and its analogues 4-epiTTX, 11-deoxyTTX, 11-norTTX-6-ol, 4,9-anhydroTTX and 5,11/6,11-dideoxyTTX were detected in all tissue types. Liver and gonads were the most toxic tissues, with the highest TTX concentrations being observed in the ovaries of female specimens. Only 22% of the analyzed muscle samples were non-toxic according to the Japanese toxicity threshold (2.2 μg TTX eq g-1), confirming the high poisoning risk from the inadvertent consumption of this species. Four steroid hormones (i.e., cortisol, testosterone, androstenedione and β-estradiol) and the gonadotropin-releasing hormone (GnRH) were detected in the gonads. Androstenedione dominated in female specimens, while GnRH was more abundant in males. A positive correlation of TTX and its analogues with β-estradiol was observed. However, a model incorporating sex rather than β-estradiol as the independent variable proven to be more efficient in predicting TTX concentration, implying that other sex-related characteristics are more important than specific hormone-regulated processes.
Collapse
Affiliation(s)
- Thekla I Anastasiou
- Hellenic Centre for Marine Research (HCMR), Institute of Marine Biology, Biotechnology and Aquaculture, 71500 Heraklion, Greece
- Department of Biology, University of Crete, 70013 Heraklion, Greece
| | - Eirini Kagiampaki
- Hellenic Centre for Marine Research (HCMR), Institute of Marine Biology, Biotechnology and Aquaculture, 71500 Heraklion, Greece
| | - Gerasimos Kondylatos
- Hellenic Centre for Marine Research (HCMR), Hydrobiological Station of Rhodes, 85131 Rhodes, Greece
| | | | - Panagiota Peristeraki
- Hellenic Centre for Marine Research (HCMR), Institute of Marine Biological Resources and Inland Waters, 71500 Heraklion, Greece
| | - Manolis Mandalakis
- Hellenic Centre for Marine Research (HCMR), Institute of Marine Biology, Biotechnology and Aquaculture, 71500 Heraklion, Greece
| |
Collapse
|
9
|
Huerta MÁ, de la Nava J, Artacho-Cordón A, Nieto FR. Efficacy and Security of Tetrodotoxin in the Treatment of Cancer-Related Pain: Systematic Review and Meta-Analysis. Mar Drugs 2023; 21:md21050316. [PMID: 37233510 DOI: 10.3390/md21050316] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/18/2023] [Accepted: 05/19/2023] [Indexed: 05/27/2023] Open
Abstract
The pharmacological treatment of cancer-related pain is unsatisfactory. Tetrodotoxin (TTX) has shown analgesia in preclinical models and clinical trials, but its clinical efficacy and safety have not been quantified. For this reason, our aim was to perform a systematic review and meta-analysis of the clinical evidence that was available. A systematic literature search was conducted in four electronic databases (Medline, Web of Science, Scopus, and ClinicalTrials.gov) up to 1 March 2023 in order to identify published clinical studies evaluating the efficacy and security of TTX in patients with cancer-related pain, including chemotherapy-induced neuropathic pain. Five articles were selected, three of which were randomized controlled trials (RCTs). The number of responders to the primary outcome (≥30% improvement in the mean pain intensity) and those suffering adverse events in the intervention and placebo groups were used to calculate effect sizes using the log odds ratio. The meta-analysis showed that TTX significantly increased the number of responders (mean = 0.68; 95% CI: 0.19-1.16, p = 0.0065) and the number of patients suffering non-severe adverse events (mean = 1.13; 95% CI: 0.31-1.95, p = 0.0068). However, TTX did not increase the risk of suffering serious adverse events (mean = 0.75; 95% CI: -0.43-1.93, p = 0.2154). In conclusion, TTX showed robust analgesic efficacy but also increased the risk of suffering non-severe adverse events. These results should be confirmed in further clinical trials with higher numbers of patients.
Collapse
Affiliation(s)
- Miguel Á Huerta
- Department of Pharmacology, University of Granada, 18016 Granada, Spain
- Institute of Neuroscience, Biomedical Research Center, University of Granada, 18016 Granada, Spain
- Biosanitary Research Institute ibs. Granada, 18012 Granada, Spain
| | - Javier de la Nava
- Department of Pharmacology, University of Granada, 18016 Granada, Spain
- Institute of Neuroscience, Biomedical Research Center, University of Granada, 18016 Granada, Spain
| | - Antonia Artacho-Cordón
- Department of Pharmacology, University of Granada, 18016 Granada, Spain
- Institute of Neuroscience, Biomedical Research Center, University of Granada, 18016 Granada, Spain
- Biosanitary Research Institute ibs. Granada, 18012 Granada, Spain
| | - Francisco R Nieto
- Department of Pharmacology, University of Granada, 18016 Granada, Spain
- Institute of Neuroscience, Biomedical Research Center, University of Granada, 18016 Granada, Spain
- Biosanitary Research Institute ibs. Granada, 18012 Granada, Spain
| |
Collapse
|
10
|
Rodrigues-Filho JL, Macêdo RL, Sarmento H, Pimenta VRA, Alonso C, Teixeira CR, Pagliosa PR, Netto SA, Santos NCL, Daura-Jorge FG, Rocha O, Horta P, Branco JO, Sartor R, Muller J, Cionek VM. From ecological functions to ecosystem services: linking coastal lagoons biodiversity with human well-being. HYDROBIOLOGIA 2023; 850:2611-2653. [PMID: 37323646 PMCID: PMC10000397 DOI: 10.1007/s10750-023-05171-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 02/03/2023] [Accepted: 02/09/2023] [Indexed: 06/17/2023]
Abstract
In this review we highlight the relevance of biodiversity that inhabit coastal lagoons, emphasizing how species functions foster processes and services associated with this ecosystem. We identified 26 ecosystem services underpinned by ecological functions performed by bacteria and other microbial organisms, zooplankton, polychaetae worms, mollusks, macro-crustaceans, fishes, birds, and aquatic mammals. These groups present high functional redundancy but perform complementary functions that result in distinct ecosystem processes. Because coastal lagoons are located in the interface between freshwater, marine and terrestrial ecosystems, the ecosystem services provided by the biodiversity surpass the lagoon itself and benefit society in a wider spatial and historical context. The species loss in coastal lagoons due to multiple human-driven impacts affects the ecosystem functioning, influencing negatively the provision of all categories of services (i.e., supporting, regulating, provisioning and cultural). Because animals' assemblages have unequal spatial and temporal distribution in coastal lagoons, it is necessary to adopt ecosystem-level management plans to protect habitat heterogeneity and its biodiversity, ensuring the provision of services for human well-being to multi-actors in the coastal zone.
Collapse
Affiliation(s)
- Jorge L. Rodrigues-Filho
- Laboratório de Ecologia Aplicada e Conservação, Departamento de Engenharia de Pesca e Ciências Biológicas, Universidade Do Estado de Santa Catarina, Laguna, SC Brazil
- Programa de Pós-Graduação em Planejamento Territorial e Desenvolvimento Socioambiental (PPGPLAN)/UDESC/FAED, Universidade do Estado de Santa Catarina, Florianópolis, SC Brazil
| | - Rafael L. Macêdo
- Graduate Program in Ecology and Natural Resources, and Department of Ecology and Evolutionary Biology, Federal University of São Carlos - UFSCar, São Carlos, Brazil
| | - Hugo Sarmento
- Graduate Program in Ecology and Natural Resources, and Department of Ecology and Evolutionary Biology, Federal University of São Carlos - UFSCar, São Carlos, Brazil
- Graduate Program in Ecology of Inland Water Ecosystems (PEA), State University of Maringá (UEM), Centre of Research in Limnology, Ichthyology and Aquaculture (Nupélia), Maringá, Paraná, Brazil
| | - Victor R. A. Pimenta
- Laboratório de Ecologia Aplicada e Conservação, Departamento de Engenharia de Pesca e Ciências Biológicas, Universidade Do Estado de Santa Catarina, Laguna, SC Brazil
- Graduate Program in Ecology and Natural Resources, and Department of Ecology and Evolutionary Biology, Federal University of São Carlos - UFSCar, São Carlos, Brazil
| | - Cecilia Alonso
- Microbial Ecology of Aquatic Systems Research Group, Centro Universitario Regional del Este, Universidad de la República, Rocha, Uruguay
| | - Clarissa R. Teixeira
- Laboratório de Mamíferos Aquáticos (LAMAQ), Departamento de Ecologia e Zoologia, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
- Whale Habitat, Ecology & Telemetry Laboratory (WHET), Oregon State University (OSU), Newport, OR USA
| | - Paulo R. Pagliosa
- Laboratório de Biodiversidade Costeira, Coordenadoria Especial de Oceanografia, Universidade Federal de Santa Catarina, Florianópolis, SC Brazil
| | - Sérgio A. Netto
- Universidade do Sul de Santa Catarina, UNISUL, Tubarão, Santa Catarina, Brazil
| | - Natália C. L. Santos
- Instituto de Biologia, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ Brazil
| | - Fábio G. Daura-Jorge
- Laboratório de Mamíferos Aquáticos (LAMAQ), Departamento de Ecologia e Zoologia, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
- Programa de Pós-Graduação em Ecologia (POSECO), Universidade Federal de Santa Catarina (UFSC), Trindade, Florianópolis, Brazil
| | - Odete Rocha
- Graduate Program in Ecology and Natural Resources, and Department of Ecology and Evolutionary Biology, Federal University of São Carlos - UFSCar, São Carlos, Brazil
| | - Paulo Horta
- Laboratório de Ficologia, Departamento de Botânica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC Brazil
| | - Joaquim O. Branco
- Graduate Program in Ecology and Natural Resources, and Department of Ecology and Evolutionary Biology, Federal University of São Carlos - UFSCar, São Carlos, Brazil
- Programa de Pós-Graduação em Ciência e Tecnologia Ambiental, Universidade do Vale do Itajaí, Itajaí, SC Brazil
| | - Rodrigo Sartor
- Universidade do Sul de Santa Catarina, UNISUL, Tubarão, Santa Catarina, Brazil
| | - Jean Muller
- Universidade do Sul de Santa Catarina, UNISUL, Tubarão, Santa Catarina, Brazil
| | - Vivian M. Cionek
- Programa de Pós-Graduação em Ciência e Tecnologia Ambiental, Universidade do Vale do Itajaí, Itajaí, SC Brazil
| |
Collapse
|
11
|
Virgen CG, Kelkar N, Tran A, Rosa CM, Cruz-Topete D, Amatya S, Cornett EM, Urits I, Viswanath O, Kaye AD. Pharmacological management of cancer pain: Novel therapeutics. Biomed Pharmacother 2022; 156:113871. [DOI: 10.1016/j.biopha.2022.113871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 10/10/2022] [Accepted: 10/13/2022] [Indexed: 12/24/2022] Open
|
12
|
Shkodra M, Caraceni A. Treatment of Neuropathic Pain Directly Due to Cancer: An Update. Cancers (Basel) 2022; 14:cancers14081992. [PMID: 35454894 PMCID: PMC9031615 DOI: 10.3390/cancers14081992] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/10/2022] [Accepted: 04/12/2022] [Indexed: 02/06/2023] Open
Abstract
Simple Summary This review discusses treatment approaches for providing pain relief to oncological patients affected by pain caused by nerve damage due to the tumor, also known as neuropathic cancer pain. Although being encountered often and causing a relevant burden to these patients, neuropathic cancer pain remains still difficult to diagnose and treat. Strong evidence about the best drugs to be used remain limited, as do therapeutic choices. Abstract Neuropathic pain can be defined as pain related to abnormal somatosensory processing in either the peripheral or central nervous system. In this review article, with neuropathic cancer pain (NCP), we refer to pain due to nervous tissue lesions caused by the tumor or its metastases. Nervous tissue damage is the cause of cancer pain in approximately 40% of those experiencing cancer pain. Recognizing a neuropathic pathophysiology in these cases may be difficult and requires specific criteria that are not homogenously applied in clinical practice. The management of this type of pain can be challenging, requiring the use of specific non-opioid adjuvant drugs. The majority of the criteria for NCP diagnosis and management have been based mainly on results from the noncancer population, risking the failure of addressing the specific needs of this population of patients. In this review, we summarize current management options available for NCP and provide some insights on new promising treatments.
Collapse
Affiliation(s)
- Morena Shkodra
- Palliative Care, Pain Therapy and Rehabilitation Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milano, Italy;
- Institute of Clinical Medicine, University of Oslo, 0318 Oslo, Norway
- Correspondence:
| | - Augusto Caraceni
- Palliative Care, Pain Therapy and Rehabilitation Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milano, Italy;
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milano, Italy
| |
Collapse
|
13
|
Wisedchaisri G, Gamal El-Din TM. Druggability of Voltage-Gated Sodium Channels-Exploring Old and New Drug Receptor Sites. Front Pharmacol 2022; 13:858348. [PMID: 35370700 PMCID: PMC8968173 DOI: 10.3389/fphar.2022.858348] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 03/01/2022] [Indexed: 01/12/2023] Open
Abstract
Voltage-gated ion channels are important drug targets because they play crucial physiological roles in both excitable and non-excitable cells. About 15% of clinical drugs used for treating human diseases target ion channels. However, most of these drugs do not provide sufficient specificity to a single subtype of the channels and their off-target side effects can be serious and sometimes fatal. Recent advancements in imaging techniques have enabled us for the first time to visualize unique and hidden parts of voltage-gated sodium channels in different structural conformations, and to develop drugs that further target a selected functional state in each channel subtype with the potential for high precision and low toxicity. In this review we describe the druggability of voltage-gated sodium channels in distinct functional states, which could potentially be used to selectively target the channels. We review classical drug receptors in the channels that have recently been structurally characterized by cryo-electron microscopy with natural neurotoxins and clinical drugs. We further examine recent drug discoveries for voltage-gated sodium channels and discuss opportunities to use distinct, state-dependent receptor sites in the voltage sensors as unique drug targets. Finally, we explore potential new receptor sites that are currently unknown for sodium channels but may be valuable for future drug discovery. The advancement presented here will help pave the way for drug development that selectively targets voltage-gated sodium channels.
Collapse
Affiliation(s)
- Goragot Wisedchaisri
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Tamer M Gamal El-Din
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| |
Collapse
|
14
|
Louzao MC, Vilariño N, Vale C, Costas C, Cao A, Raposo-Garcia S, Vieytes MR, Botana LM. Current Trends and New Challenges in Marine Phycotoxins. Mar Drugs 2022; 20:md20030198. [PMID: 35323497 PMCID: PMC8950113 DOI: 10.3390/md20030198] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/04/2022] [Accepted: 03/05/2022] [Indexed: 02/04/2023] Open
Abstract
Marine phycotoxins are a multiplicity of bioactive compounds which are produced by microalgae and bioaccumulate in the marine food web. Phycotoxins affect the ecosystem, pose a threat to human health, and have important economic effects on aquaculture and tourism worldwide. However, human health and food safety have been the primary concerns when considering the impacts of phycotoxins. Phycotoxins toxicity information, often used to set regulatory limits for these toxins in shellfish, lacks traceability of toxicity values highlighting the need for predefined toxicological criteria. Toxicity data together with adequate detection methods for monitoring procedures are crucial to protect human health. However, despite technological advances, there are still methodological uncertainties and high demand for universal phycotoxin detectors. This review focuses on these topics, including uncertainties of climate change, providing an overview of the current information as well as future perspectives.
Collapse
Affiliation(s)
- Maria Carmen Louzao
- Departamento de Farmacologia, Facultad de Veterinaria, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (N.V.); (C.V.); (C.C.); (A.C.); (S.R.-G.)
- Correspondence: (M.C.L.); (L.M.B.)
| | - Natalia Vilariño
- Departamento de Farmacologia, Facultad de Veterinaria, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (N.V.); (C.V.); (C.C.); (A.C.); (S.R.-G.)
| | - Carmen Vale
- Departamento de Farmacologia, Facultad de Veterinaria, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (N.V.); (C.V.); (C.C.); (A.C.); (S.R.-G.)
| | - Celia Costas
- Departamento de Farmacologia, Facultad de Veterinaria, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (N.V.); (C.V.); (C.C.); (A.C.); (S.R.-G.)
| | - Alejandro Cao
- Departamento de Farmacologia, Facultad de Veterinaria, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (N.V.); (C.V.); (C.C.); (A.C.); (S.R.-G.)
| | - Sandra Raposo-Garcia
- Departamento de Farmacologia, Facultad de Veterinaria, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (N.V.); (C.V.); (C.C.); (A.C.); (S.R.-G.)
| | - Mercedes R. Vieytes
- Departamento de Fisiologia, Facultad de Veterinaria, Universidade de Santiago de Compostela, 27002 Lugo, Spain;
| | - Luis M. Botana
- Departamento de Farmacologia, Facultad de Veterinaria, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (N.V.); (C.V.); (C.C.); (A.C.); (S.R.-G.)
- Correspondence: (M.C.L.); (L.M.B.)
| |
Collapse
|
15
|
Katikou P, Gokbulut C, Kosker AR, Campàs M, Ozogul F. An Updated Review of Tetrodotoxin and Its Peculiarities. Mar Drugs 2022; 20:md20010047. [PMID: 35049902 PMCID: PMC8780202 DOI: 10.3390/md20010047] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/28/2021] [Accepted: 12/30/2021] [Indexed: 12/19/2022] Open
Abstract
Tetrodotoxin (TTX) is a crystalline, weakly basic, colorless organic substance and is one of the most potent marine toxins known. Although TTX was first isolated from pufferfish, it has been found in numerous other marine organisms and a few terrestrial species. Moreover, tetrodotoxication is still an important health problem today, as TTX has no known antidote. TTX poisonings were most commonly reported from Japan, Thailand, and China, but today the risk of TTX poisoning is spreading around the world. Recent studies have shown that TTX-containing fish are being found in other regions of the Pacific and in the Indian Ocean, as well as the Mediterranean Sea. This review aims to summarize pertinent information available to date on the structure, origin, distribution, mechanism of action of TTX and analytical methods used for the detection of TTX, as well as on TTX-containing organisms, symptoms of TTX poisoning, and incidence worldwide.
Collapse
Affiliation(s)
- Panagiota Katikou
- Ministry of Rural Development and Food, Directorate of Research, Innovation and Education, Hapsa & Karatasou 1, 54626 Thessaloniki, Greece
- Correspondence: (P.K.); (F.O.)
| | - Cengiz Gokbulut
- Department of Pharmacology, Faculty of Medicine, Balikesir University, Balikesir 10145, Turkey;
| | - Ali Rıza Kosker
- Department of Seafood Processing Technology, Faculty of Fisheries, Cukurova University, Adana 01330, Turkey;
| | - Mònica Campàs
- IRTA, Ctra Poble Nou km 5.5, 43540 Sant Carles de la Ràpita, Spain;
| | - Fatih Ozogul
- Department of Seafood Processing Technology, Faculty of Fisheries, Cukurova University, Adana 01330, Turkey;
- Correspondence: (P.K.); (F.O.)
| |
Collapse
|
16
|
Ulman A, Yildiz T, Demirel N, Canak O, Yemişken E, Pauly D. The biology and ecology of the invasive silver-cheeked toadfish (Lagocephalus sceleratus), with emphasis on the Eastern Mediterranean. NEOBIOTA 2021. [DOI: 10.3897/neobiota.68.71767] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Invasive species pose threats to either human health or inflict ecological and/or economic damage. The silver-cheeked toadfish (Lagocephalus sceleratus), a Lessepsian species, is one of the most harmful species in the Mediterranean Sea, because of its potent neurotoxin, impacts on marine biodiversity, and the increased costs and labor they inflict on fishers. Since the catch and consumption of this pufferfish is prohibited by almost all countries bordering the Mediterranean, they have now expanded into the entire Mediterranean and Black Sea. We performed a comprehensive study of L. sceleratus covering ecological aspects, growth, reproduction, diet and trophic level based on samples from southwestern coasts of Turkey. The estimated growth parameters were L∞ = 88.7 cm, K = 0.27 year-1, C = 0.6 and WP = 0.1. Their sex-ratio was M/F = 1:0.69. Lagocephalus sceleratus appears to be a batch spawner with discontinuous oocyte recruitment and has different spawning seasons in the Eastern Mediterranean which seem to be based on temperature cues which get shorter in duration as one moves north from the Suez. We also report their first positive ecological trait, that they are controlling some other invasive species through their diets, such as lionfish, Red Sea goatfish, rabbitfish and longspine sea urchins, in addition to controlling themselves through cannibalism, which appears to be density-dependent. They are indeed a top predator in the region with a trophic level of 4.1. We suggest that targeted fishing using improved gear-types to reduce fishing gear damages are initiated, and that finding commercial markets for pufferfish could help to naturally fund ongoing control efforts.
Collapse
|
17
|
Voltage-Gated Sodium Channels: A Prominent Target of Marine Toxins. Mar Drugs 2021; 19:md19100562. [PMID: 34677461 PMCID: PMC8537899 DOI: 10.3390/md19100562] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 09/29/2021] [Accepted: 10/02/2021] [Indexed: 12/19/2022] Open
Abstract
Voltage-gated sodium channels (VGSCs) are considered to be one of the most important ion channels given their remarkable physiological role. VGSCs constitute a family of large transmembrane proteins that allow transmission, generation, and propagation of action potentials. This occurs by conducting Na+ ions through the membrane, supporting cell excitability and communication signals in various systems. As a result, a wide range of coordination and physiological functions, from locomotion to cognition, can be accomplished. Drugs that target and alter the molecular mechanism of VGSCs’ function have highly contributed to the discovery and perception of the function and the structure of this channel. Among those drugs are various marine toxins produced by harmful microorganisms or venomous animals. These toxins have played a key role in understanding the mode of action of VGSCs and in mapping their various allosteric binding sites. Furthermore, marine toxins appear to be an emerging source of therapeutic tools that can relieve pain or treat VGSC-related human channelopathies. Several studies documented the effect of marine toxins on VGSCs as well as their pharmaceutical applications, but none of them underlined the principal marine toxins and their effect on VGSCs. Therefore, this review aims to highlight the neurotoxins produced by marine animals such as pufferfish, shellfish, sea anemone, and cone snail that are active on VGSCs and discuss their pharmaceutical values.
Collapse
|
18
|
Fong MF, Duffy KR, Leet MP, Candler CT, Bear MF. Correction of amblyopia in cats and mice after the critical period. eLife 2021; 10:e70023. [PMID: 34464258 PMCID: PMC8456712 DOI: 10.7554/elife.70023] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 08/20/2021] [Indexed: 11/25/2022] Open
Abstract
Monocular deprivation early in development causes amblyopia, a severe visual impairment. Prognosis is poor if therapy is initiated after an early critical period. However, clinical observations have shown that recovery from amblyopia can occur later in life when the non-deprived (fellow) eye is removed. The traditional interpretation of this finding is that vision is improved simply by the elimination of interocular suppression in primary visual cortex, revealing responses to previously subthreshold input. However, an alternative explanation is that silencing activity in the fellow eye establishes conditions in visual cortex that enable the weak connections from the amblyopic eye to gain strength, in which case the recovery would persist even if vision is restored in the fellow eye. Consistent with this idea, we show here in cats and mice that temporary inactivation of the fellow eye is sufficient to promote a full and enduring recovery from amblyopia at ages when conventional treatments fail. Thus, connections serving the amblyopic eye are capable of substantial plasticity beyond the critical period, and this potential is unleashed by reversibly silencing the fellow eye.
Collapse
Affiliation(s)
- Ming-fai Fong
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of TechnologyCambridgeUnited States
| | - Kevin R Duffy
- Department of Psychology and Neuroscience, Dalhousie UniversityHalifaxCanada
| | - Madison P Leet
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of TechnologyCambridgeUnited States
| | - Christian T Candler
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of TechnologyCambridgeUnited States
| | - Mark F Bear
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of TechnologyCambridgeUnited States
| |
Collapse
|
19
|
Bucciarelli GM, Lechner M, Fontes A, Kats LB, Eisthen HL, Shaffer HB. From Poison to Promise: The Evolution of Tetrodotoxin and Its Potential as a Therapeutic. Toxins (Basel) 2021; 13:toxins13080517. [PMID: 34437388 PMCID: PMC8402337 DOI: 10.3390/toxins13080517] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/17/2021] [Accepted: 07/20/2021] [Indexed: 11/16/2022] Open
Abstract
Tetrodotoxin (TTX) is a potent neurotoxin that was first identified in pufferfish but has since been isolated from an array of taxa that host TTX-producing bacteria. However, determining its origin, ecosystem roles, and biomedical applications has challenged researchers for decades. Recognized as a poison and for its lethal effects on humans when ingested, TTX is primarily a powerful sodium channel inhibitor that targets voltage-gated sodium channels, including six of the nine mammalian isoforms. Although lethal doses for humans range from 1.5-2.0 mg TTX (blood level 9 ng/mL), when it is administered at levels far below LD50, TTX exhibits therapeutic properties, especially to treat cancer-related pain, neuropathic pain, and visceral pain. Furthermore, TTX can potentially treat a variety of medical ailments, including heroin and cocaine withdrawal symptoms, spinal cord injuries, brain trauma, and some kinds of tumors. Here, we (i) describe the perplexing evolution and ecology of tetrodotoxin, (ii) review its mechanisms and modes of action, and (iii) offer an overview of the numerous ways it may be applied as a therapeutic. There is much to be explored in these three areas, and we offer ideas for future research that combine evolutionary biology with therapeutics. The TTX system holds great promise as a therapeutic and understanding the origin and chemical ecology of TTX as a poison will only improve its general benefit to humanity.
Collapse
Affiliation(s)
- Gary M. Bucciarelli
- Department of Ecology and Evolutionary Biology & UCLA La Kretz Center for California Conservation Science, Institute of the Environment and Sustainability, University of California, Los Angeles, CA 90095, USA; (M.L.); (H.B.S.)
- Correspondence:
| | - Maren Lechner
- Department of Ecology and Evolutionary Biology & UCLA La Kretz Center for California Conservation Science, Institute of the Environment and Sustainability, University of California, Los Angeles, CA 90095, USA; (M.L.); (H.B.S.)
| | - Audrey Fontes
- Natural Science Division, Pepperdine University, Malibu, CA 90263, USA; (A.F.); (L.B.K.)
| | - Lee B. Kats
- Natural Science Division, Pepperdine University, Malibu, CA 90263, USA; (A.F.); (L.B.K.)
| | - Heather L. Eisthen
- Department of Integrative Biology, Michigan State University, East Lansing, MI 48824, USA;
| | - H. Bradley Shaffer
- Department of Ecology and Evolutionary Biology & UCLA La Kretz Center for California Conservation Science, Institute of the Environment and Sustainability, University of California, Los Angeles, CA 90095, USA; (M.L.); (H.B.S.)
| |
Collapse
|
20
|
Tetrodotoxin: A New Strategy to Treat Visceral Pain? Toxins (Basel) 2021; 13:toxins13070496. [PMID: 34357968 PMCID: PMC8310099 DOI: 10.3390/toxins13070496] [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: 05/21/2021] [Revised: 07/08/2021] [Accepted: 07/13/2021] [Indexed: 12/13/2022] Open
Abstract
Visceral pain is one of the most common symptoms associated with functional gastrointestinal (GI) disorders. Although the origin of these symptoms has not been clearly defined, the implication of both the central and peripheral nervous systems in visceral hypersensitivity is well established. The role of several pathways in visceral nociception has been explored, as well as the influence of specific receptors on afferent neurons, such as voltage-gated sodium channels (VGSCs). VGSCs initiate action potentials and dysfunction of these channels has recently been associated with painful GI conditions. Current treatments for visceral pain generally involve opioid based drugs, which are associated with important side-effects and a loss of effectiveness or tolerance. Hence, efforts have been intensified to find new, more effective and longer-lasting therapies. The implication of VGSCs in visceral hypersensitivity has drawn attention to tetrodotoxin (TTX), a relatively selective sodium channel blocker, as a possible and promising molecule to treat visceral pain and related diseases. As such, here we will review the latest information regarding this toxin that is relevant to the treatment of visceral pain and the possible advantages that it may offer relative to other treatments, alone or in combination.
Collapse
|
21
|
González-Cano R, Ruiz-Cantero MC, Santos-Caballero M, Gómez-Navas C, Tejada MÁ, Nieto FR. Tetrodotoxin, a Potential Drug for Neuropathic and Cancer Pain Relief? Toxins (Basel) 2021; 13:toxins13070483. [PMID: 34357955 PMCID: PMC8310002 DOI: 10.3390/toxins13070483] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 07/08/2021] [Accepted: 07/09/2021] [Indexed: 12/20/2022] Open
Abstract
Tetrodotoxin (TTX) is a potent neurotoxin found mainly in puffer fish and other marine and terrestrial animals. TTX blocks voltage-gated sodium channels (VGSCs) which are typically classified as TTX-sensitive or TTX-resistant channels. VGSCs play a key role in pain signaling and some TTX-sensitive VGSCs are highly expressed by adult primary sensory neurons. During pathological pain conditions, such as neuropathic pain, upregulation of some TTX-sensitive VGSCs, including the massive re-expression of the embryonic VGSC subtype NaV1.3 in adult primary sensory neurons, contribute to painful hypersensitization. In addition, people with loss-of-function mutations in the VGSC subtype NaV1.7 present congenital insensitive to pain. TTX displays a prominent analgesic effect in several models of neuropathic pain in rodents. According to this promising preclinical evidence, TTX is currently under clinical development for chemo-therapy-induced neuropathic pain and cancer-related pain. This review focuses primarily on the preclinical and clinical evidence that support a potential analgesic role for TTX in these pain states. In addition, we also analyze the main toxic effects that this neurotoxin produces when it is administered at therapeutic doses, and the therapeutic potential to alleviate neuropathic pain of other natural toxins that selectively block TTX-sensitive VGSCs.
Collapse
Affiliation(s)
- Rafael González-Cano
- Department of Pharmacology, and Neurosciences Institute (Biomedical Research Center), University of Granada, 18016 Granada, Spain; (R.G.-C.); (M.C.R.-C.); (M.S.-C.); (C.G.-N.)
- Biosanitary Research Institute ibs.GRANADA, 18012 Granada, Spain
| | - M. Carmen Ruiz-Cantero
- Department of Pharmacology, and Neurosciences Institute (Biomedical Research Center), University of Granada, 18016 Granada, Spain; (R.G.-C.); (M.C.R.-C.); (M.S.-C.); (C.G.-N.)
- Biosanitary Research Institute ibs.GRANADA, 18012 Granada, Spain
| | - Miriam Santos-Caballero
- Department of Pharmacology, and Neurosciences Institute (Biomedical Research Center), University of Granada, 18016 Granada, Spain; (R.G.-C.); (M.C.R.-C.); (M.S.-C.); (C.G.-N.)
- Biosanitary Research Institute ibs.GRANADA, 18012 Granada, Spain
| | - Carlos Gómez-Navas
- Department of Pharmacology, and Neurosciences Institute (Biomedical Research Center), University of Granada, 18016 Granada, Spain; (R.G.-C.); (M.C.R.-C.); (M.S.-C.); (C.G.-N.)
| | | | - Francisco R. Nieto
- Department of Pharmacology, and Neurosciences Institute (Biomedical Research Center), University of Granada, 18016 Granada, Spain; (R.G.-C.); (M.C.R.-C.); (M.S.-C.); (C.G.-N.)
- Biosanitary Research Institute ibs.GRANADA, 18012 Granada, Spain
- Correspondence: ; Tel.: +34-958-242-056
| |
Collapse
|
22
|
Unlocking the Health Potential of Microalgae as Sustainable Sources of Bioactive Compounds. Int J Mol Sci 2021; 22:ijms22094383. [PMID: 33922258 PMCID: PMC8122763 DOI: 10.3390/ijms22094383] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/14/2021] [Accepted: 04/18/2021] [Indexed: 12/15/2022] Open
Abstract
Microalgae are known to produce a plethora of compounds derived from the primary and secondary metabolism. Different studies have shown that these compounds may have allelopathic, antimicrobial, and antipredator activities. In addition, in vitro and in vivo screenings have shown that several compounds have interesting bioactivities (such as antioxidant, anti-inflammatory, anticancer, and antimicrobial) for the possible prevention and treatment of human pathologies. Additionally, the enzymatic pathways responsible for the synthesis of these compounds, and the targets and mechanisms of their action have also been investigated for a few species. However, further research is necessary for their full exploitation and possible pharmaceutical and other industrial applications. Here, we review the current knowledge on the chemical characteristics, biological activities, mechanism of action, and the enzymes involved in the synthesis of microalgal metabolites with potential benefits for human health.
Collapse
|
23
|
Tetrodotoxin for Chemotherapy-Induced Neuropathic Pain: A Randomized, Double-Blind, Placebo-Controlled, Parallel-Dose Finding Trial. Toxins (Basel) 2021; 13:toxins13040235. [PMID: 33805908 PMCID: PMC8064362 DOI: 10.3390/toxins13040235] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/20/2021] [Accepted: 03/23/2021] [Indexed: 12/15/2022] Open
Abstract
Tetrodotoxin (TTX) has emerged as a potentially efficacious agent for chemotherapy-induced neuropathic pain (CINP), a prevalent, debilitating condition often resistant to analgesics. This randomized, double-blind, dose-finding study was undertaken to explore safety and trends in efficacy of four TTX doses and to identify a dose for further study. One hundred and twenty-five patients with taxane- or platinum-related CINP received subcutaneous placebo or TTX (7.5 µg twice daily (BID), 15 µg BID, 30 µg once daily (QD), 30 µg BID) for four consecutive days. Primary outcome measure was average patient-reported Numeric Pain Rating Scale (NPRS) score during Days 21–28 post-treatment. Changes in mean NPRS score were not statistically different between cohorts, due to small trial size and influence of a few robust placebo responders. Cumulative responder analysis showed significant difference from placebo with 30 µg BID cohort using the maximum response at any timepoint (p = 0.072), 5-day (p = 0.059), 10-day (p = 0.027), and 20-day (p = 0.071) rolling averages. In secondary quality of life (QOL) outcomes, 30 µg BID cohort also differed significantly from placebo in a number of SF-36 and CIPN20 subscales. Most adverse events (AE) were mild or moderate with oral paresthesia (29.6%) and oral hypoesthesia (24.8%) as most common.
Collapse
|
24
|
New opportunities and challenges of venom-based and bacteria-derived molecules for anticancer targeted therapy. Semin Cancer Biol 2020; 80:356-369. [PMID: 32846203 DOI: 10.1016/j.semcancer.2020.08.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 08/14/2020] [Accepted: 08/15/2020] [Indexed: 12/24/2022]
Abstract
Due to advances in detection and treatment of cancer, especially the rise in the targeted therapy, the five-year relative survival rate of all cancers has increased significantly. However, according to the analysis of the survival rate of cancer patients in 2019, the survival rate of most cancers is still less than five years. Therefore, to combat complex cancer and further improve the 5-year survival rate of cancer patients, it is necessary to develop some new anticancer drugs. Because of the adaptive evolution of toxic species for millions of years, the venom sac is a "treasure bank", which has millions of biomolecules with high affinity and stability awaiting further development. Complete utilization of venom-based and bacteria-derived drugs in the market is still staggering because of incomplete understanding regarding their mode of action. In this review, we focused on the currently identified targets for anticancer effects based on venomous and bacterial biomolecules, such as ion channels, membrane non-receptor molecules, integrins, and other related target molecules. This review will serve as the key for exploring the molecular mechanisms behind the anticancer potential of venom-based and bacteria-derived drugs and will also lay the path for the development of anticancer targeted therapy.
Collapse
|
25
|
Kavoosi M, O’Reilly TE, Kavoosi M, Chai P, Engel C, Korz W, Gallen CC, Lester RM. Safety, Tolerability, Pharmacokinetics, and Concentration-QTc Analysis of Tetrodotoxin: A Randomized, Dose Escalation Study in Healthy Adults. Toxins (Basel) 2020; 12:toxins12080511. [PMID: 32784930 PMCID: PMC7472037 DOI: 10.3390/toxins12080511] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 08/05/2020] [Accepted: 08/05/2020] [Indexed: 12/19/2022] Open
Abstract
Tetrodotoxin (TTX) is a highly specific voltage-gated sodium channel (VGSC) blocker in clinical evaluation as a peripheral-acting analgesic for chronic pain. This study presents the first published results of the safety including cardiac liability of TTX at therapeutic-relevant concentrations in twenty-five healthy adults. Randomized, double-blind, placebo-, and positive- (moxifloxacin) controlled study evaluated single ascending doses of 15 µg, 30 µg, and 45 µg TTX over 3 periods with a 7-day washout between each period. Subcutaneous injections of TTX were readily absorbed, reaching maximum plasma concentration (Cmax) within 1.5 h. Both extent of exposure (AUC) and Cmax increased in proportion to dose. No QT prolongation was identified by concentration-QTc analysis and the upper bounds of the two-sided 90% confidence interval of predicted maximum baseline and placebo corrected QTcF (ΔΔQTcF) value did not exceed 10 ms for all tetrodotoxin doses, thereby meeting the criteria of a negative QT study. Safety assessments showed no clinically relevant changes with values similar between all groups and no subject withdrawing due to adverse events. Paresthesia, oral-paresthesia, headache, dizziness, nausea, and myalgia were the most common TEAEs (overall occurrence ≥5%) in the TTX treatment groups. TTX doses investigated in this study are safe, well-tolerated, and lack proarrhythmic proclivity.
Collapse
Affiliation(s)
- Mojgan Kavoosi
- WEX Pharmaceuticals Inc., Vancouver, BC V6E-4A6, Canada; (M.K.); (W.K.); (C.C.G.)
- Correspondence:
| | | | - Mehran Kavoosi
- WEX Pharmaceuticals Inc., Vancouver, BC V6E-4A6, Canada; (M.K.); (W.K.); (C.C.G.)
| | - Peng Chai
- Celerion Inc., Lincoln, NE 68502, USA;
| | | | - Walter Korz
- WEX Pharmaceuticals Inc., Vancouver, BC V6E-4A6, Canada; (M.K.); (W.K.); (C.C.G.)
| | | | | |
Collapse
|
26
|
Kushnarev M, Pirvulescu IP, Candido KD, Knezevic NN. Neuropathic pain: preclinical and early clinical progress with voltage-gated sodium channel blockers. Expert Opin Investig Drugs 2020; 29:259-271. [PMID: 32070160 DOI: 10.1080/13543784.2020.1728254] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Introduction: Neuropathic pain is a chronic condition that significantly affects the quality of life of millions of people globally. Most of the pharmacologic treatments currently in use demonstrate modest efficacy and over half of all patients do not respond to medical management. Hence, there is a need for new, efficacious drugs. Evidence points toward voltage-gated sodium channels as a key target for novel analgesics.Area covered: The role of voltage-gated sodium channels in pain pathophysiology is illuminated and the preclinical and clinical data for new sodium channel blockers and toxin-derived lead compounds are examined. The expansion of approved sodium channel blockers is discussed along with the limitations of current research, trends in drug development, and the potential of personalized medicine.Expert opinion: The transition from preclinical to clinical studies can be difficult because of the inherent inability of animal models to express the complexities of pain states. Pain pathways are notoriously intricate and may be pharmacologically modulated at a variety of targets; it is unlikely that action at a single target could completely abolish a pain response because pain is rarely unifactorial. Combination therapy may be necessary and this could further confound the discovery of novel agents.
Collapse
Affiliation(s)
- Mikhail Kushnarev
- Department of Anesthesiology, Advocate Illinois Masonic Medical Center, Chicago, IL, USA
| | - Iulia Paula Pirvulescu
- Department of Anesthesiology, Advocate Illinois Masonic Medical Center, Chicago, IL, USA
| | - Kenneth D Candido
- Department of Anesthesiology, Advocate Illinois Masonic Medical Center, Chicago, IL, USA.,Department of Anesthesiology, College of Medicine, University of Illinois, Chicago, IL, USA.,Department of Surgery, College of Medicine, University of Illinois, Chicago, IL, USA
| | - Nebojsa Nick Knezevic
- Department of Anesthesiology, Advocate Illinois Masonic Medical Center, Chicago, IL, USA.,Department of Anesthesiology, College of Medicine, University of Illinois, Chicago, IL, USA.,Department of Surgery, College of Medicine, University of Illinois, Chicago, IL, USA
| |
Collapse
|
27
|
Bajaj S, Ong ST, Chandy KG. Contributions of natural products to ion channel pharmacology. Nat Prod Rep 2020; 37:703-716. [PMID: 32065187 DOI: 10.1039/c9np00056a] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Covering: Up to 2020Ion channels are a vast super-family of membrane proteins that play critical physiological roles in excitable and non-excitable cells. Their biomedical importance makes them valuable and attractive drug targets for neurological, cardiovascular, gastrointestinal and metabolic diseases, and for cancer therapy and immune modulation. Current therapeutics target only a minor subset of ion channels, leaving a large unexploited space within the ion channel field. Natural products harnessed from the almost unlimited and diverse universe of compounds within the bioenvironment have been used to modulate channels for decades. In this review we highlight the impact made by natural products on ion channel pharmacology, specifically on K+, NaV and CaV channels, and use case studies to describe the development of ion channel-modulating drugs from natural sources for the treatment of pain, heart disease and autoimmune diseases.
Collapse
Affiliation(s)
- Saumya Bajaj
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Experimental Medicine Building, 59 Nanyang Drive, 636921, Singapore.
| | | | | |
Collapse
|
28
|
Analgesia Effect of Enteric Sustained-Release Tetrodotoxin Pellets in the Rat. Pharmaceutics 2020; 12:pharmaceutics12010032. [PMID: 31906313 PMCID: PMC7022972 DOI: 10.3390/pharmaceutics12010032] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 12/26/2019] [Accepted: 12/29/2019] [Indexed: 12/28/2022] Open
Abstract
Tetrodotoxin (TTX) was identified as a latent neurotoxin that has a significant analgesia effect. It was rapidly absorbed and excreted in rat after intramuscular (i.m.) injection. To maintain the effect, frequent injections were required. The enteric sustained-release TTX pellets with sucrose pellets as a drug carrier was prepared by fluidized bed spray irrigation, coated in sequence with Eudragit NE30D as a sustained-release layer, hydroxypropyl methylcellulose (HPMC) as a barrier layer and Eudragit L30D-55 as an enteric coating. TTX in the pellets could be sustained released for 12 h in dissolution test. In vivo, TTX pellets reached Cmax at 5 h, and t1/2 was 14.52 ± 2.37 h after intragastrically (i.g.) administration in rat. In acetic acid induced writhing test in rat, the pellets at the dosages of 20, 40, 60 and 80 μg·kg−1 produced analgesic effect at about 1.5 h to 9 h and the strongest effect was at about 3 h to 6 h. Simultaneously, the LD50 of the enteric sustained-release TTX pellets was 840.13 μg·kg−1, and the ED50 was about 30 μg·kg−1. Thus, the therapeutic index was about 25. The enteric sustained-release TTX pellets with absolute analgesia effect and greatly enhanced safety was prepared.
Collapse
|
29
|
Mulcahy JV, Pajouhesh H, Beckley JT, Delwig A, Bois JD, Hunter JC. Challenges and Opportunities for Therapeutics Targeting the Voltage-Gated Sodium Channel Isoform Na V1.7. J Med Chem 2019; 62:8695-8710. [PMID: 31012583 PMCID: PMC6786914 DOI: 10.1021/acs.jmedchem.8b01906] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Voltage-gated sodium ion channel subtype 1.7 (NaV1.7) is a high interest target for the discovery of non-opioid analgesics. Compelling evidence from human genetic data, particularly the finding that persons lacking functional NaV1.7 are insensitive to pain, has spurred considerable effort to develop selective inhibitors of this Na+ ion channel target as analgesic medicines. Recent clinical setbacks and disappointing performance of preclinical compounds in animal pain models, however, have led to skepticism around the potential of selective NaV1.7 inhibitors as human therapeutics. In this Perspective, we discuss the attributes and limitations of recently disclosed investigational drugs targeting NaV1.7 and review evidence that, by better understanding the requirements for selectivity and target engagement, the opportunity to deliver effective analgesic medicines targeting NaV1.7 endures.
Collapse
Affiliation(s)
- John V. Mulcahy
- SiteOne Therapeutics, 280 Utah Ave, Suite 250, South San Francisco, CA 94080
| | - Hassan Pajouhesh
- SiteOne Therapeutics, 280 Utah Ave, Suite 250, South San Francisco, CA 94080
| | - Jacob T. Beckley
- SiteOne Therapeutics, 351 Evergreen Drive, Suite B1, Bozeman, MT 59715
| | - Anton Delwig
- SiteOne Therapeutics, 280 Utah Ave, Suite 250, South San Francisco, CA 94080
| | - J. Du Bois
- Stanford University, Lokey Chemistry and Biology, 337 Campus Drive, Stanford, CA 94305
| | - John C. Hunter
- SiteOne Therapeutics, 280 Utah Ave, Suite 250, South San Francisco, CA 94080
| |
Collapse
|
30
|
Ma RSY, Kayani K, Whyte-Oshodi D, Whyte-Oshodi A, Nachiappan N, Gnanarajah S, Mohammed R. Voltage gated sodium channels as therapeutic targets for chronic pain. J Pain Res 2019; 12:2709-2722. [PMID: 31564962 PMCID: PMC6743634 DOI: 10.2147/jpr.s207610] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 08/02/2019] [Indexed: 01/23/2023] Open
Abstract
Being maladaptive and frequently unresponsive to pharmacotherapy, chronic pain presents a major unmet clinical need. While an intact central nervous system is required for conscious pain perception, nociceptor hyperexcitability induced by nerve injury in the peripheral nervous system (PNS) is sufficient and necessary to initiate and maintain neuropathic pain. The genesis and propagation of action potentials is dependent on voltage-gated sodium channels, in particular, Nav1.7, Nav1.8 and Nav1.9. However, nerve injury triggers changes in their distribution, expression and/or biophysical properties, leading to aberrant excitability. Most existing treatment for pain relief acts through non-selective, state-dependent sodium channel blockage and have narrow therapeutic windows. Natural toxins and developing subtype-specific and molecular-specific sodium channel blockers show promise for treatment of neuropathic pain with minimal side effects. New approaches to analgesia include combination therapy and gene therapy. Here, we review how individual sodium channel subtypes contribute to pain, and the attempts made to develop more effective analgesics for the treatment of chronic pain.
Collapse
Affiliation(s)
- Renee Siu Yu Ma
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Kayani Kayani
- Department of Medicine, University of Cambridge, Cambridge, UK
| | | | | | | | | | - Raihan Mohammed
- Department of Medicine, University of Cambridge, Cambridge, UK
| |
Collapse
|
31
|
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: 2.4] [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
|
32
|
Gonçalves TC, Benoit E, Kurz M, Lucarain L, Fouconnier S, Combemale S, Jaquillard L, Schombert B, Chambard JM, Boukaiba R, Hessler G, Bohme A, Bialy L, Hourcade S, Béroud R, De Waard M, Servent D, Partiseti M. From identification to functional characterization of cyriotoxin-1a, an antinociceptive toxin from the spider Cyriopagopus schioedtei. Br J Pharmacol 2019; 176:1298-1314. [PMID: 30784059 DOI: 10.1111/bph.14628] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 01/24/2019] [Accepted: 01/31/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND AND PURPOSE The NaV 1.7 channel is highly expressed in dorsal root ganglia of the sensory nervous system and plays a central role in the pain signalling process. We investigated a library prepared from original venoms of 117 different animals to identify new selective inhibitors of this target. EXPERIMENTAL APPROACH We used high throughput screening of a large venom collection using automated patch-clamp experiments on human voltage-gated sodium channel subtypes and then in vitro and in vivo electrophysiological experiments to characterize the active peptides that have been purified, sequenced, and chemically synthesized. Analgesic effects were evaluated in vivo in mice models. KEY RESULTS We identified cyriotoxin-1a (CyrTx-1a), a novel peptide isolated from Cyriopagopus schioedtei spider venom, as a candidate for further characterization. This 33 amino acids toxin belongs to the inhibitor cystine knot structural family and inhibits hNaV 1.1-1.3 and 1.6-1.7 channels in the low nanomolar range, compared to the micromolar range for hNaV 1.4-1.5 and 1.8 channels. CyrTx-1a was 920 times more efficient at inhibiting tetrodotoxin (TTX)-sensitive than TTX-resistant sodium currents recorded from adult mouse dorsal root ganglia neurons and in vivo electrophysiological experiments showed that CyrTx-1a was approximately 170 times less efficient than huwentoxin-IV at altering mouse skeletal neuromuscular excitability properties. CyrTx-1a exhibited an analgesic effect in mice by increasing reaction time in the hot-plate assay. CONCLUSIONS AND IMPLICATIONS The pharmacological profile of CyrTx-1a paves the way for further molecular engineering aimed to optimize the potential antinociceptive properties of this peptide.
Collapse
Affiliation(s)
- Tânia C Gonçalves
- Integrated Drug Discovery-High Content Biology, Sanofi R&D, Vitry-sur-Seine, France.,Service d'Ingénierie Moléculaire des Protéines (SIMOPRO), CEA, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Evelyne Benoit
- Service d'Ingénierie Moléculaire des Protéines (SIMOPRO), CEA, Université Paris-Saclay, Gif-sur-Yvette, France.,Institut des Neurosciences Paris-Saclay (Neuro-PSI), UMR CNRS/Université Paris-Sud 9197, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Michael Kurz
- Integrated Drug Discovery-Synthetic Molecular Design, Sanofi R&D, Frankfurt, Germany
| | - Laetitia Lucarain
- Integrated Drug Discovery-High Content Biology, Sanofi R&D, Vitry-sur-Seine, France
| | - Sophie Fouconnier
- Integrated Drug Discovery-High Content Biology, Sanofi R&D, Vitry-sur-Seine, France
| | | | | | - Brigitte Schombert
- Integrated Drug Discovery-High Content Biology, Sanofi R&D, Vitry-sur-Seine, France
| | - Jean-Marie Chambard
- Integrated Drug Discovery-High Content Biology, Sanofi R&D, Vitry-sur-Seine, France
| | - Rachid Boukaiba
- Integrated Drug Discovery-High Content Biology, Sanofi R&D, Vitry-sur-Seine, France
| | - Gerhard Hessler
- Integrated Drug Discovery-Synthetic Molecular Design, Sanofi R&D, Frankfurt, Germany
| | - Andrees Bohme
- Integrated Drug Discovery-High Content Biology, Sanofi R&D, Vitry-sur-Seine, France
| | - Laurent Bialy
- Integrated Drug Discovery-Synthetic Molecular Design, Sanofi R&D, Frankfurt, Germany
| | - Stéphane Hourcade
- Neuroscience Therapeutic Area, Neurodegeneration Research, Sanofi R&D, Chilly-Mazarin, France
| | | | - Michel De Waard
- Smartox Biotechnology, Saint-Egrève, France.,Institut du Thorax, Inserm UMR 1087/CNRS UMR 6291, LabEx "Ion Channels, Science and Therapeutics", Nantes, France
| | - Denis Servent
- Service d'Ingénierie Moléculaire des Protéines (SIMOPRO), CEA, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Michel Partiseti
- Integrated Drug Discovery-High Content Biology, Sanofi R&D, Vitry-sur-Seine, France
| |
Collapse
|
33
|
Maatuf Y, Geron M, Priel A. The Role of Toxins in the Pursuit for Novel Analgesics. Toxins (Basel) 2019; 11:toxins11020131. [PMID: 30813430 PMCID: PMC6409898 DOI: 10.3390/toxins11020131] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 02/17/2019] [Accepted: 02/20/2019] [Indexed: 12/19/2022] Open
Abstract
Chronic pain is a major medical issue which reduces the quality of life of millions and inflicts a significant burden on health authorities worldwide. Currently, management of chronic pain includes first-line pharmacological therapies that are inadequately effective, as in just a portion of patients pain relief is obtained. Furthermore, most analgesics in use produce severe or intolerable adverse effects that impose dose restrictions and reduce compliance. As the majority of analgesic agents act on the central nervous system (CNS), it is possible that blocking pain at its source by targeting nociceptors would prove more efficient with minimal CNS-related side effects. The development of such analgesics requires the identification of appropriate molecular targets and thorough understanding of their structural and functional features. To this end, plant and animal toxins can be employed as they affect ion channels with high potency and selectivity. Moreover, elucidation of the toxin-bound ion channel structure could generate pharmacophores for rational drug design while favorable safety and analgesic profiles could highlight toxins as leads or even as valuable therapeutic compounds themselves. Here, we discuss the use of plant and animal toxins in the characterization of peripherally expressed ion channels which are implicated in pain.
Collapse
Affiliation(s)
- Yossi Maatuf
- The Institute for Drug Research (IDR), School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel.
| | - Matan Geron
- The Institute for Drug Research (IDR), School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel.
| | - Avi Priel
- The Institute for Drug Research (IDR), School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel.
| |
Collapse
|
34
|
Boente-Juncal A, Vale C, Cifuentes M, Otero P, Camiña M, Rodriguez-Vieytes M, Botana LM. Chronic In Vivo Effects of Repeated Exposure to Low Oral Doses of Tetrodotoxin: Preliminary Evidence of Nephrotoxicity and Cardiotoxicity. Toxins (Basel) 2019; 11:E96. [PMID: 30736354 PMCID: PMC6410189 DOI: 10.3390/toxins11020096] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 01/22/2019] [Accepted: 02/01/2019] [Indexed: 11/24/2022] Open
Abstract
Tetrodotoxin (TTX) is one of the most potent naturally occurring neurotoxins. InitiallyTTX was associated with human food intoxications in Japan, but nowadays, concerns about thehuman health risks posed by TTX have increased in Europe after the identification of the toxin infish, marine gastropods, and bivalves captured in European waters. Even when TTX monitoring isnot currently performed in Europe, an acute oral no observable effect level (NOAEL) of 75 μg/kghas been recently established but, to date, no studies evaluating the chronic oral toxicity of TTXhave been released, even when EFSA has highlighted the need for them. Thus, in this work, thechronic effects of low oral TTX doses (below the acute lethal dose 50) were evaluated followinginternationally adopted guidelines. The results presented here demonstrate that low oral doses ofTTX have deleterious effects on renal and cardiac tissues. Moreover, alterations in bloodbiochemistry parameters, urine production, and urinalysis data were already detected at the oraldose of 75 μg/kg after the 28 days exposure. Thus, the data presented here constitute an initialapproach for the chronic evaluation of the in vivo toxicity of tetrodotoxin after its ingestion throughcontaminated fishery products.
Collapse
Affiliation(s)
- Andrea Boente-Juncal
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002 Lugo, Spain.
| | - Carmen Vale
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002 Lugo, Spain.
| | - Manuel Cifuentes
- Departamento de Anatomía, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002 Lugo, Spain.
| | - Paz Otero
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002 Lugo, Spain.
| | - Mercedes Camiña
- Departamento de Fisiología, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002 Lugo, Spain.
| | - Mercedes Rodriguez-Vieytes
- Departamento de Fisiología, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002 Lugo, Spain.
| | - Luis Miguel Botana
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002 Lugo, Spain.
| |
Collapse
|
35
|
Coates MD, Vrana KE, Ruiz-Velasco V. The influence of voltage-gated sodium channels on human gastrointestinal nociception. Neurogastroenterol Motil 2019; 31:e13460. [PMID: 30216585 DOI: 10.1111/nmo.13460] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 08/01/2018] [Accepted: 08/07/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND Abdominal pain is a frequent and persistent problem in the most common gastrointestinal disorders, including irritable bowel syndrome and inflammatory bowel disease. Pain adversely impacts quality of life, incurs significant healthcare expenditures, and remains a challenging issue to manage with few safe therapeutic options currently available. It is imperative that new methods are developed for identifying and treating this symptom. A variety of peripherally active neuroendocrine signaling elements have the capability to influence gastrointestinal pain perception. A large and growing body of evidence suggests that voltage-gated sodium channels (VGSCs) play a critical role in the development and modulation of nociceptive signaling associated with the gut. Several VGSC isoforms demonstrate significant promise as potential targets for improved diagnosis and treatment of gut-based disorders associated with hyper- and hyposensitivity to abdominal pain. PURPOSE In this article, we critically review key investigations that have evaluated the potential role that VGSCs play in visceral nociception and discuss recent advances related to this topic. Specifically, we discuss the following: (a) what is known about the structure and basic function of VGSCs, (b) the role that each VGSC plays in gut nociception, particularly as it relates to human physiology, and (c) potential diagnostic and therapeutic uses of VGSCs to manage disorders associated with chronic abdominal pain.
Collapse
Affiliation(s)
- Matthew D Coates
- Division of Gastroenterology & Hepatology, Department of Medicine, Penn State University College of Medicine, Hershey, Pennsylvania
| | - Kent E Vrana
- Department of Pharmacology, Penn State University College of Medicine, Hershey, Pennsylvania
| | - Victor Ruiz-Velasco
- Department of Anesthesiology and Perioperative Medicine, Penn State University College of Medicine, Hershey, Pennsylvania
| |
Collapse
|
36
|
Addressing the Issue of Tetrodotoxin Targeting. Mar Drugs 2018; 16:md16100352. [PMID: 30261623 PMCID: PMC6212850 DOI: 10.3390/md16100352] [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: 09/04/2018] [Revised: 09/21/2018] [Accepted: 09/24/2018] [Indexed: 12/27/2022] Open
Abstract
This review is devoted to the medical application of tetrodotoxin (TTX), a potent non-protein specific blocker of voltage-gated sodium (NaV) channels. The selectivity of action, lack of affinity with the heart muscle NaV channels, and the inability to penetrate the blood–brain barrier make this toxin an attractive candidate for anesthetic and analgesic drug design. The efficacy of TTX was shown in neuropathic, acute and inflammatory pain models. The main emphasis of the review is on studies focused on the improvement of TTX efficacy and safety in conjunction with additional substances and drug delivery systems. A significant improvement in the effectiveness of the toxin was demonstrated when used in tandem with vasoconstrictors, local anesthetics and chemical permeation enhancers, with the best results obtained with the encapsulation of TTX in microparticles and liposomes conjugated to gold nanorods.
Collapse
|
37
|
Gonçalves TC, Benoit E, Partiseti M, Servent D. The Na V1.7 Channel Subtype as an Antinociceptive Target for Spider Toxins in Adult Dorsal Root Ganglia Neurons. Front Pharmacol 2018; 9:1000. [PMID: 30233376 PMCID: PMC6131673 DOI: 10.3389/fphar.2018.01000] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 08/14/2018] [Indexed: 12/11/2022] Open
Abstract
Although necessary for human survival, pain may sometimes become pathologic if long-lasting and associated with alterations in its signaling pathway. Opioid painkillers are officially used to treat moderate to severe, and even mild, pain. However, the consequent strong and not so rare complications that occur, including addiction and overdose, combined with pain management costs, remain an important societal and economic concern. In this context, animal venom toxins represent an original source of antinociceptive peptides that mainly target ion channels (such as ASICs as well as TRP, CaV, KV and NaV channels) involved in pain transmission. The present review aims to highlight the NaV1.7 channel subtype as an antinociceptive target for spider toxins in adult dorsal root ganglia neurons. It will detail (i) the characteristics of these primary sensory neurons, the first ones in contact with pain stimulus and conveying the nociceptive message, (ii) the electrophysiological properties of the different NaV channel subtypes expressed in these neurons, with a particular attention on the NaV1.7 subtype, an antinociceptive target of choice that has been validated by human genetic evidence, and (iii) the features of spider venom toxins, shaped of inhibitory cysteine knot motif, that present high affinity for the NaV1.7 subtype associated with evidenced analgesic efficacy in animal models.
Collapse
Affiliation(s)
- Tânia C Gonçalves
- Sanofi R&D, Integrated Drug Discovery - High Content Biology, Paris, France.,Service d'Ingénierie Moléculaire des Protéines, CEA de Saclay, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Evelyne Benoit
- Service d'Ingénierie Moléculaire des Protéines, CEA de Saclay, Université Paris-Saclay, Gif-sur-Yvette, France.,Institut des Neurosciences Paris-Saclay, UMR CNRS/Université Paris-Sud 9197, Gif-sur-Yvette, France
| | - Michel Partiseti
- Sanofi R&D, Integrated Drug Discovery - High Content Biology, Paris, France
| | - Denis Servent
- Service d'Ingénierie Moléculaire des Protéines, CEA de Saclay, Université Paris-Saclay, Gif-sur-Yvette, France
| |
Collapse
|
38
|
Hong B, Sun J, Zheng H, Le Q, Wang C, Bai K, He J, He H, Dong Y. Effect of Tetrodotoxin Pellets in a Rat Model of Postherpetic Neuralgia. Mar Drugs 2018; 16:E195. [PMID: 29874779 PMCID: PMC6025269 DOI: 10.3390/md16060195] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 05/23/2018] [Accepted: 06/01/2018] [Indexed: 12/11/2022] Open
Abstract
Postherpetic neuralgia (PHN) is nerve pain caused by a reactivation of the varicella zoster virus. Medications are used to reduce PHN but their use is limited by serious side effects. Tetrodotoxin (TTX) is a latent neurotoxin that can block neuropathic pain, but its therapeutic index is only 3⁻5 times with intravenous or intramuscular injection. Therefore, we prepared oral TTX pellets and examined their effect in a rat model of PHN induced by resiniferatoxin (RTX). Oral TTX pellets were significantly effective at preventing RTX-induced mechanical and thermal allodynia, and similar to pregabalin. Moreover, oral administration of TTX pellets dose-dependently inhibited RTX-induced PHN compared with intramuscular administration of TTX injection. We also studied the pharmacokinetic profile of TTX pellets. Our results showed that the blood concentration of TTX reached a maximum plasma concentration (Cmax) at around 2 h, with an elimination half-life time (t1/2) of 3.23 ± 1.74 h after intragastric administration. The median lethal dose (LD50) of TTX pellets was 517.43 μg/kg via oral administration to rats, while the median effective dose (ED50) was approximately 5.85 μg/kg, and the therapeutic index was 88.45. Altogether, this has indicated that oral TTX pellets greatly enhance safety when compared with TTX injection.
Collapse
Affiliation(s)
- Bihong Hong
- Department of Materials Science and Engineering, College of Materials, Xiamen University, Xiamen 361005, China.
- Engineering Research Center of Marine Biological Resource Comprehensive Utilization, Third Institute of Oceanography, State Oceanic Administration, Xiamen 361005, China.
| | - Jipeng Sun
- Engineering Research Center of Marine Biological Resource Comprehensive Utilization, Third Institute of Oceanography, State Oceanic Administration, Xiamen 361005, China.
| | - Hongzhi Zheng
- School of pharmaceutical Sciences, Xiamen University, Xiamen 361102, China.
| | - Qingqing Le
- Engineering Research Center of Marine Biological Resource Comprehensive Utilization, Third Institute of Oceanography, State Oceanic Administration, Xiamen 361005, China.
| | - Changsen Wang
- Engineering Research Center of Marine Biological Resource Comprehensive Utilization, Third Institute of Oceanography, State Oceanic Administration, Xiamen 361005, China.
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, China.
| | - Kaikai Bai
- Engineering Research Center of Marine Biological Resource Comprehensive Utilization, Third Institute of Oceanography, State Oceanic Administration, Xiamen 361005, China.
| | - Jianlin He
- Engineering Research Center of Marine Biological Resource Comprehensive Utilization, Third Institute of Oceanography, State Oceanic Administration, Xiamen 361005, China.
| | - Huanghuang He
- Engineering Research Center of Marine Biological Resource Comprehensive Utilization, Third Institute of Oceanography, State Oceanic Administration, Xiamen 361005, China.
| | - Yanming Dong
- Department of Materials Science and Engineering, College of Materials, Xiamen University, Xiamen 361005, China.
| |
Collapse
|
39
|
Gaps in Understanding Mechanism and Lack of Treatments: Potential Use of a Nonhuman Primate Model of Oxaliplatin-Induced Neuropathic Pain. Pain Res Manag 2018; 2018:1630709. [PMID: 29854035 PMCID: PMC5954874 DOI: 10.1155/2018/1630709] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 04/03/2018] [Indexed: 12/11/2022]
Abstract
The antineoplastic agent oxaliplatin induces an acute hypersensitivity evoked by cold that has been suggested to be due to sensitized central and peripheral neurons. Rodent-based preclinical studies have suggested numerous treatments for the alleviation of oxaliplatin-induced neuropathic pain, but few have demonstrated robust clinical efficacy. One issue is that current understanding of the pathophysiology of oxaliplatin-induced neuropathic pain is primarily based on rodent models, which might not entirely recapitulate the clinical pathophysiology. In addition, there is currently no objective physiological marker for pain that could be utilized to objectively indicate treatment efficacy. Nonhuman primates are phylogenetically and neuroanatomically similar to humans; thus, disease mechanism in nonhuman primates could reflect that of clinical oxaliplatin-induced neuropathy. Cold-activated pain-related brain areas in oxaliplatin-treated macaques were attenuated with duloxetine, the only drug that has demonstrated clinical efficacy for chemotherapy-induced neuropathic pain. By contrast, drugs that have not demonstrated clinical efficacy in oxaliplatin-induced neuropathic pain did not reduce brain activation. Thus, a nonhuman primate model could greatly enhance understanding of clinical pathophysiology beyond what has been obtained with rodent models and, furthermore, brain activation could serve as an objective marker of pain and therapeutic efficacy.
Collapse
|
40
|
Hamad MK, He K, Abdulrazeq HF, Mustafa AM, Luceri R, Kamal N, Ali M, Nakhla J, Herzallah MM, Mammis A. Potential Uses of Isolated Toxin Peptides in Neuropathic Pain Relief: A Literature Review. World Neurosurg 2018; 113:333-347.e5. [DOI: 10.1016/j.wneu.2018.01.116] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 01/15/2018] [Accepted: 01/16/2018] [Indexed: 01/31/2023]
|
41
|
Erickson A, Deiteren A, Harrington AM, Garcia‐Caraballo S, Castro J, Caldwell A, Grundy L, Brierley SM. Voltage-gated sodium channels: (Na V )igating the field to determine their contribution to visceral nociception. J Physiol 2018; 596:785-807. [PMID: 29318638 PMCID: PMC5830430 DOI: 10.1113/jp273461] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 01/02/2018] [Indexed: 12/19/2022] Open
Abstract
Chronic visceral pain, altered motility and bladder dysfunction are common, yet poorly managed symptoms of functional and inflammatory disorders of the gastrointestinal and urinary tracts. Recently, numerous human channelopathies of the voltage-gated sodium (NaV ) channel family have been identified, which induce either painful neuropathies, an insensitivity to pain, or alterations in smooth muscle function. The identification of these disorders, in addition to the recent utilisation of genetically modified NaV mice and specific NaV channel modulators, has shed new light on how NaV channels contribute to the function of neuronal and non-neuronal tissues within the gastrointestinal tract and bladder. Here we review the current pre-clinical and clinical evidence to reveal how the nine NaV channel family members (NaV 1.1-NaV 1.9) contribute to abdominal visceral function in normal and disease states.
Collapse
Affiliation(s)
- Andelain Erickson
- Visceral Pain Research Group, Human Physiology, Centre for Neuroscience, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth Australia5042Australia
- Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of AdelaideSouth Australian Health and Medical Research Institute (SAHMRI)North TerraceAdelaideSouth Australia 5000Australia
| | - Annemie Deiteren
- Visceral Pain Research Group, Human Physiology, Centre for Neuroscience, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth Australia5042Australia
- Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of AdelaideSouth Australian Health and Medical Research Institute (SAHMRI)North TerraceAdelaideSouth Australia 5000Australia
| | - Andrea M. Harrington
- Visceral Pain Research Group, Human Physiology, Centre for Neuroscience, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth Australia5042Australia
- Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of AdelaideSouth Australian Health and Medical Research Institute (SAHMRI)North TerraceAdelaideSouth Australia 5000Australia
| | - Sonia Garcia‐Caraballo
- Visceral Pain Research Group, Human Physiology, Centre for Neuroscience, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth Australia5042Australia
- Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of AdelaideSouth Australian Health and Medical Research Institute (SAHMRI)North TerraceAdelaideSouth Australia 5000Australia
| | - Joel Castro
- Visceral Pain Research Group, Human Physiology, Centre for Neuroscience, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth Australia5042Australia
- Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of AdelaideSouth Australian Health and Medical Research Institute (SAHMRI)North TerraceAdelaideSouth Australia 5000Australia
| | - Ashlee Caldwell
- Visceral Pain Research Group, Human Physiology, Centre for Neuroscience, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth Australia5042Australia
- Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of AdelaideSouth Australian Health and Medical Research Institute (SAHMRI)North TerraceAdelaideSouth Australia 5000Australia
| | - Luke Grundy
- Visceral Pain Research Group, Human Physiology, Centre for Neuroscience, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth Australia5042Australia
- Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of AdelaideSouth Australian Health and Medical Research Institute (SAHMRI)North TerraceAdelaideSouth Australia 5000Australia
| | - Stuart M. Brierley
- Visceral Pain Research Group, Human Physiology, Centre for Neuroscience, College of Medicine and Public HealthFlinders UniversityBedford ParkSouth Australia5042Australia
- Centre for Nutrition and Gastrointestinal Diseases, Discipline of Medicine, University of AdelaideSouth Australian Health and Medical Research Institute (SAHMRI)North TerraceAdelaideSouth Australia 5000Australia
| |
Collapse
|
42
|
Tetrodotoxin, a Candidate Drug for Nav1.1-Induced Mechanical Pain? Mar Drugs 2018; 16:md16020072. [PMID: 29470418 PMCID: PMC5852500 DOI: 10.3390/md16020072] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 02/09/2018] [Accepted: 02/20/2018] [Indexed: 12/28/2022] Open
Abstract
Tetrodotoxin (TTX), the mode of action of which has been known since the 1960s, is widely used in pharmacology as a specific inhibitor of voltage-gated sodium channels (Nav channels). This toxin has contributed to the characterization of the allosteric model of the Nav channel, and to discriminating TTX-sensitive and TTX-resistant subtypes. In addition to its role as a pharmacological tool, TTX is now considered a therapeutic molecule, and its development should lead to its use in certain pathologies involving Nav channels, particularly in the field of pain. Specifically, the blockade of Nav channels expressed in nociceptive fibres is one strategy for alleviating pain and its deleterious consequences on health. Recent work has identified, in addition to the Nav1.7, 1.8 and 1.9 channels, the Nav1.1 subtype on dorsal root ganglion (DRG) neurons as a crucial player in mechanical and non-thermal pain. The sensitivity of Nav1.1 to TTX could be exploited at the therapeutic level, especially in chronic pain conditions.
Collapse
|
43
|
Rwei AY, Paris JL, Wang B, Wang W, Axon CD, Vallet-Regí M, Langer R, Kohane DS. Ultrasound-triggered local anaesthesia. Nat Biomed Eng 2017; 1:644-653. [PMID: 29152410 PMCID: PMC5687284 DOI: 10.1038/s41551-017-0117-6] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 06/27/2017] [Indexed: 01/09/2023]
Abstract
On-demand relief of local pain would allow patients to control the timing, intensity and duration of nerve block in a safe and non-invasive manner. Ultrasound would be a suitable trigger for such a system, as it is in common clinical use and can penetrate deeply into the body. Here, we demonstrate that ultrasound-triggered delivery of an anaesthetic from liposomes allows the timing, intensity and duration of nerve block to be controlled by ultrasound parameters. On insonation, the encapsulated sonosensitizer protoporphyrin IX produces reactive oxygen species that react with the liposomal membrane, leading to the release of the potent local anaesthetic tetrodotoxin. We also show repeatable ultrasound-triggered nerve blocks in vivo, with nerve-block duration depending on the extent and intensity of insonation. We did not detect any systemic toxicity, and tissue reaction was benign in all groups. On-demand, personalized local anaesthesia could be beneficial for the managing of relatively localized pain states, and potentially minimize opioid use.
Collapse
Affiliation(s)
- Alina Y Rwei
- Department of Anaesthesiology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Juan L Paris
- Dpto. Química Inorgánica y Bioinorgánica, Facultad de Farmacia, UCM, Instituto de Investigación Sanitaria Hospital 12 de Octubre i+12, 28040, Madrid, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Avenida Monforte de Lemos, 3-5, 28029, Madrid, Spain
| | - Bruce Wang
- Department of Anaesthesiology, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Weiping Wang
- Dr Li Dak-Sum Research Centre, The University of Hong Kong-Karolinska Institutet Collaboration in Regenerative Medicine, The University of Hong Kong, Hong Kong, China
| | - Christopher D Axon
- Department of Anaesthesiology, Boston Children's Hospital, Boston, MA, 02115, USA
| | - María Vallet-Regí
- Dpto. Química Inorgánica y Bioinorgánica, Facultad de Farmacia, UCM, Instituto de Investigación Sanitaria Hospital 12 de Octubre i+12, 28040, Madrid, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Avenida Monforte de Lemos, 3-5, 28029, Madrid, Spain
| | - Robert Langer
- David H. Koch Institutes for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Daniel S Kohane
- Department of Anaesthesiology, Boston Children's Hospital, Boston, MA, 02115, USA.
- Laboratory for Biomaterials and Drug Delivery, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|