1
|
Maxion A, Kutafina E, Dohrn MF, Sacré P, Lampert A, Tigerholm J, Namer B. A modelling study to dissect the potential role of voltage-gated ion channels in activity-dependent conduction velocity changes as identified in small fiber neuropathy patients. Front Comput Neurosci 2023; 17:1265958. [PMID: 38156040 PMCID: PMC10752960 DOI: 10.3389/fncom.2023.1265958] [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: 07/24/2023] [Accepted: 10/25/2023] [Indexed: 12/30/2023] Open
Abstract
Objective Patients with small fiber neuropathy (SFN) suffer from neuropathic pain, which is still a therapeutic problem. Changed activation patterns of mechano-insensitive peripheral nerve fibers (CMi) could cause neuropathic pain. However, there is sparse knowledge about mechanisms leading to CMi dysfunction since it is difficult to dissect specific molecular mechanisms in humans. We used an in-silico model to elucidate molecular causes of CMi dysfunction as observed in single nerve fiber recordings (microneurography) of SFN patients. Approach We analyzed microneurography data from 97 CMi-fibers from healthy individuals and 34 of SFN patients to identify activity-dependent changes in conduction velocity. Using the NEURON environment, we adapted a biophysical realistic preexisting CMi-fiber model with ion channels described by Hodgkin-Huxley dynamics for identifying molecular mechanisms leading to those changes. Via a grid search optimization, we assessed the interplay between different ion channels, Na-K-pump, and resting membrane potential. Main results Changing a single ion channel conductance, Na-K-pump or membrane potential individually is not sufficient to reproduce in-silico CMi-fiber dysfunction of unchanged activity-dependent conduction velocity slowing and quicker normalization of conduction velocity after stimulation as observed in microneurography. We identified the best combination of mechanisms: increased conductance of potassium delayed-rectifier and decreased conductance of Na-K-pump and depolarized membrane potential. When the membrane potential is unchanged, opposite changes in Na-K-pump and ion channels generate the same effect. Significance Our study suggests that not one single mechanism accounts for pain-relevant changes in CMi-fibers, but a combination of mechanisms. A depolarized membrane potential, as previously observed in patients with neuropathic pain, leads to changes in the contribution of ion channels and the Na-K-pump. Thus, when searching for targets for the treatment of neuropathic pain, combinations of several molecules in interplay with the membrane potential should be regarded.
Collapse
Affiliation(s)
- Anna Maxion
- Research Group Neuroscience, Interdisciplinary Centre for Clinical Research within the Faculty of Medicine at the RWTH Aachen University, Aachen, Germany
| | - Ekaterina Kutafina
- Institute of Medical Informatics, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Maike F. Dohrn
- Department of Neurology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Pierre Sacré
- Department of Electrical Engineering and Computer Science, University of Liège, Liège, Belgium
| | - Angelika Lampert
- Institute of Neurophysiology, Uniklinik RWTH Aachen University Aachen, Aachen, Germany
| | - Jenny Tigerholm
- Joint Research Center for Computational Biomedicine, RWTH Aachen, Aachen, Germany
| | - Barbara Namer
- Research Group Neuroscience, Interdisciplinary Centre for Clinical Research within the Faculty of Medicine at the RWTH Aachen University, Aachen, Germany
- Institute of Neurophysiology, RWTH Aachen University, Aachen, Germany
- Institute of Physiology and Pathophysiology, University of Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
2
|
Yaksh TL, Santos GGD, Borges Paes Lemes J, Malange K. Neuraxial drug delivery in pain management: An overview of past, present, and future. Best Pract Res Clin Anaesthesiol 2023; 37:243-265. [PMID: 37321769 DOI: 10.1016/j.bpa.2023.04.003] [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: 04/05/2023] [Accepted: 04/11/2023] [Indexed: 06/17/2023]
Abstract
Activation of neuraxial nociceptive linkages leads to a high level of encoding of the message that is transmitted to the brain and that can initiate a pain state with its attendant emotive covariates. As we review here, the encoding of this message is subject to a profound regulation by pharmacological targeting of dorsal root ganglion and dorsal horn systems. Though first shown with the robust and selective modulation by spinal opiates, subsequent work has revealed the pharmacological and biological complexity of these neuraxial systems and points to several regulatory targets. Novel therapeutic delivery platforms, such as viral transfection, antisense and targeted neurotoxins, point to disease-modifying approaches that can selectively address the acute and chronic pain phenotype. Further developments are called for in delivery devices to enhance local distribution and to minimize concentration gradients, as frequently occurs with the poorly mixed intrathecal space. The field has advanced remarkably since the mid-1970s, but these advances must always address the issues of safety and tolerability of neuraxial therapy.
Collapse
Affiliation(s)
- Tony L Yaksh
- Department of Anesthesiology University of California, San Diego, San Diego CA, 92103, USA.
| | | | | | - Kaue Malange
- Department of Anesthesiology University of California, San Diego, San Diego CA, 92103, USA
| |
Collapse
|
3
|
Almomani R, Sopacua M, Marchi M, Ślęczkowska M, Lindsey P, de Greef BTA, Hoeijmakers JGJ, Salvi E, Merkies ISJ, Ferdousi M, Malik RA, Ziegler D, Derks KWJ, Boenhof G, Martinelli-Boneschi F, Cazzato D, Lombardi R, Dib-Hajj S, Waxman SG, Smeets HJM, Gerrits MM, Faber CG, Lauria G. Genetic Profiling of Sodium Channels in Diabetic Painful and Painless and Idiopathic Painful and Painless Neuropathies. Int J Mol Sci 2023; 24:ijms24098278. [PMID: 37175987 PMCID: PMC10179245 DOI: 10.3390/ijms24098278] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 04/15/2023] [Accepted: 04/28/2023] [Indexed: 05/15/2023] Open
Abstract
Neuropathic pain is a frequent feature of diabetic peripheral neuropathy (DPN) and small fiber neuropathy (SFN). Resolving the genetic architecture of these painful neuropathies will lead to better disease management strategies, counselling and intervention. Our aims were to profile ten sodium channel genes (SCG) expressed in a nociceptive pathway in painful and painless DPN and painful and painless SFN patients, and to provide a perspective for clinicians who assess patients with painful peripheral neuropathy. Between June 2014 and September 2016, 1125 patients with painful-DPN (n = 237), painless-DPN (n = 309), painful-SFN (n = 547) and painless-SFN (n = 32), recruited in four different centers, were analyzed for SCN3A, SCN7A-SCN11A and SCN1B-SCN4B variants by single molecule Molecular inversion probes-Next Generation Sequence. Patients were grouped based on phenotype and the presence of SCG variants. Screening of SCN3A, SCN7A-SCN11A, and SCN1B-SCN4B revealed 125 different (potential) pathogenic variants in 194 patients (17.2%, n = 194/1125). A potential pathogenic variant was present in 18.1% (n = 142/784) of painful neuropathy patients vs. 15.2% (n = 52/341) of painless neuropathy patients (17.3% (n = 41/237) for painful-DPN patients, 14.9% (n = 46/309) for painless-DPN patients, 18.5% (n = 101/547) for painful-SFN patients, and 18.8% (n = 6/32) for painless-SFN patients). Of the variants detected, 70% were in SCN7A, SCN9A, SCN10A and SCN11A. The frequency of SCN9A and SCN11A variants was the highest in painful-SFN patients, SCN7A variants in painful-DPN patients, and SCN10A variants in painless-DPN patients. Our findings suggest that rare SCG genetic variants may contribute to the development of painful neuropathy. Genetic profiling and SCG variant identification should aid in a better understanding of the genetic variability in patients with painful and painless neuropathy, and may lead to better risk stratification and the development of more targeted and personalized pain treatments.
Collapse
Affiliation(s)
- Rowida Almomani
- Department of Medical Laboratory Sciences, Jordan University of Science and Technology, Irbid 22110, Jordan
- Clinical Genomics Unit, Department of Genetics and Cell Biology, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Maurice Sopacua
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Centre+, 6229 HX Maastricht, The Netherlands
| | - Margherita Marchi
- Neuroalgology Unit, IRCCS Foundation "Carlo Besta" Neurological Institute, 20133 Milan, Italy
| | - Milena Ślęczkowska
- Clinical Genomics Unit, Department of Genetics and Cell Biology, Maastricht University, 6229 ER Maastricht, The Netherlands
- Department of Toxicogenomics, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Patrick Lindsey
- Clinical Genomics Unit, Department of Genetics and Cell Biology, Maastricht University, 6229 ER Maastricht, The Netherlands
- Department of Toxicogenomics, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Bianca T A de Greef
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Centre+, 6229 HX Maastricht, The Netherlands
| | - Janneke G J Hoeijmakers
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Centre+, 6229 HX Maastricht, The Netherlands
| | - Erika Salvi
- Neuroalgology Unit, IRCCS Foundation "Carlo Besta" Neurological Institute, 20133 Milan, Italy
| | - Ingemar S J Merkies
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Centre+, 6229 HX Maastricht, The Netherlands
- Department of Neurology, Curaçao Medical Center, 4365+37Q, J. H. J. Hamelbergweg, Willemstad, Curacao
| | - Maryam Ferdousi
- Institute of Cardiovascular Sciences, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester M13 9P, UK
| | - Rayaz A Malik
- Institute of Cardiovascular Sciences, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester M13 9P, UK
- Weill Cornell Medicine-Qatar, Doha P.O. Box 24144, Qatar
| | - Dan Ziegler
- German Diabetes Centre, 40225 Düsseldorf, Germany
| | - Kasper W J Derks
- Department of Clinical Genetics, Maastricht University Medical Centre+, 6229 HX Maastricht, The Netherlands
| | - Gidon Boenhof
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, 40225 Düsseldorf, Germany
| | - Filippo Martinelli-Boneschi
- Laboratory of Human Genetics of Neurological Disorders, Institute of Experimental Neurology (INSPE), Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Daniele Cazzato
- Neuroalgology Unit, IRCCS Foundation "Carlo Besta" Neurological Institute, 20133 Milan, Italy
| | - Raffaella Lombardi
- Neuroalgology Unit, IRCCS Foundation "Carlo Besta" Neurological Institute, 20133 Milan, Italy
| | - Sulayman Dib-Hajj
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Hubert J M Smeets
- Clinical Genomics Unit, Department of Genetics and Cell Biology, Maastricht University, 6229 ER Maastricht, The Netherlands
- Department of Toxicogenomics, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Monique M Gerrits
- Department of Clinical Genetics, Maastricht University Medical Centre+, 6229 HX Maastricht, The Netherlands
| | - Catharina G Faber
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Centre+, 6229 HX Maastricht, The Netherlands
| | - Giuseppe Lauria
- Neuroalgology Unit, IRCCS Foundation "Carlo Besta" Neurological Institute, 20133 Milan, Italy
- Department of Biomedical and Clinical Sciences "Luigi Sacco", University of Milan, 20157 Milan, Italy
| |
Collapse
|
4
|
Dormer A, Narayanan M, Schentag J, Achinko D, Norman E, Kerrigan J, Jay G, Heydorn W. A Review of the Therapeutic Targeting of SCN9A and Nav1.7 for Pain Relief in Current Human Clinical Trials. J Pain Res 2023; 16:1487-1498. [PMID: 37168847 PMCID: PMC10166096 DOI: 10.2147/jpr.s388896] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 03/14/2023] [Indexed: 05/13/2023] Open
Abstract
Introduction There is a great need to find alternative treatments for chronic pain which have become a healthcare problem. We discuss current therapeutic targeting Nav1.7. Areas Covered Nav1.7 is a sodium ion channel protein that is associated with several human pain genetic syndromes. It has been found that mutations associated with Nav1.7 lead to the loss of the ability to perceive pain in individuals that are otherwise normal. Several therapeutic interventions are presently undergoing preclinical and research using the methodology of damping Nav1.7 expressions as a methodology to decrease the sensation of pain leading to analgesia. Expert Opinion It is our strong belief that there is a viable future in the targeting of protein of Nav1.7 for the relief of chronic pain in humans. The review will look at the genomics associated with SCN1A and proteomic of Nav1.7 as a foundation to explain the mechanism of the therapeutic interventions targeting Nav1.7, the human disease that are associated with Nav1.7, and the current development of treatment for chronic pain whether in preclinical or clinical trials targeting Nav1.7 expressions. The development of therapeutic antagonists targeting Nav1.7 could be a viable alternative to the current treatments which have led to the opioid crisis. Therefore, Nav1.7 targeted treatment has a major clinical significance that will have positive consequences as it relates to chronic pain interventions.
Collapse
Affiliation(s)
- Anton Dormer
- Research and Development, Pepvax, Inc, Silver Spring, MD, USA
- Correspondence: Anton Dormer, Research and Development, PepVax, Inc, 8720 Georgia Ave #1000, Silver Spring, MD, 20910, USA, Email
| | | | - Jerome Schentag
- Research and Development, Pepvax, Inc, Silver Spring, MD, USA
| | - Daniel Achinko
- Research and Development, Pepvax, Inc, Silver Spring, MD, USA
| | - Elton Norman
- Research and Development, Pepvax, Inc, Silver Spring, MD, USA
| | - James Kerrigan
- Research and Development, Navintus, Inc, Princeton, NJ, USA
| | - Gary Jay
- Research and Development, Navintus, Inc, Princeton, NJ, USA
| | | |
Collapse
|
5
|
Le Cann K, Meents JE, Sudha Bhagavath Eswaran V, Dohrn MF, Bott R, Maier A, Bialer M, Hautvast P, Erickson A, Rolke R, Rothermel M, Körner J, Kurth I, Lampert A. Assessing the impact of pain-linked Nav1.7 variants: An example of two variants with no biophysical effect. Channels (Austin) 2021; 15:208-228. [PMID: 33487118 PMCID: PMC7833769 DOI: 10.1080/19336950.2020.1870087] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/23/2020] [Accepted: 12/23/2020] [Indexed: 11/30/2022] Open
Abstract
Mutations in the voltage-gated sodium channel Nav1.7 are linked to human pain. The Nav1.7/N1245S variant was described before in several patients suffering from primary erythromelalgia and/or olfactory hypersensitivity. We have identified this variant in a pain patient and a patient suffering from severe and life-threatening orthostatic hypotension. In addition, we report a female patient suffering from muscle pain and carrying the Nav1.7/E1139K variant. We tested both Nav1.7 variants by whole-cell voltage-clamp recordings in HEK293 cells, revealing a slightly enhanced current density for the N1245S variant when co-expressed with the β1 subunit. This effect was counteracted by an enhanced slow inactivation. Both variants showed similar voltage dependence of activation and steady-state fast inactivation, as well as kinetics of fast inactivation, deactivation, and use-dependency compared to WT Nav1.7. Finally, homology modeling revealed that the N1245S substitution results in different intramolecular interaction partners. Taken together, these experiments do not point to a clear pathogenic effect of either the N1245S or E1139K variant and suggest they may not be solely responsible for the patients' pain symptoms. As discussed previously for other variants, investigations in heterologous expression systems may not sufficiently mimic the pathophysiological situation in pain patients, and single nucleotide variants in other genes or modulatory proteins are necessary for these specific variants to show their effect. Our findings stress that biophysical investigations of ion channel mutations need to be evaluated with care and should preferably be supplemented with studies investigating the mutations in their context, ideally in human sensory neurons.
Collapse
Affiliation(s)
- Kim Le Cann
- Institute of Physiology, RWTH Aachen University Hospital, Aachen, Germany
| | - Jannis E. Meents
- Institute of Physiology, RWTH Aachen University Hospital, Aachen, Germany
| | | | - Maike F. Dohrn
- Department of Neurology, Medical Faculty, RWTH Aachen University Hospital, Aachen, Germany
| | - Raya Bott
- Institute of Physiology, RWTH Aachen University Hospital, Aachen, Germany
| | - Andrea Maier
- Department of Neurology, Medical Faculty, RWTH Aachen University Hospital, Aachen, Germany
| | - Martin Bialer
- Division of Clinical Metabolism of Medical Genetics and Human Genomics at Northwell Health System, New-York, United States
| | - Petra Hautvast
- Institute of Physiology, RWTH Aachen University Hospital, Aachen, Germany
| | - Andelain Erickson
- Institute of Physiology, RWTH Aachen University Hospital, Aachen, Germany
| | - Roman Rolke
- Department for Palliative Care, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Markus Rothermel
- Department of Chemosensation, AG Neuromodulation, Institute for Biology II, RWTH Aachen University, Aachen, 52074, Germany
| | - Jannis Körner
- Institute of Physiology, RWTH Aachen University Hospital, Aachen, Germany
- Department of Anaesthesiology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Ingo Kurth
- Institute of Human Genetics, Medical Faculty, RWTH Aachen University Hospital, Aachen, Germany
| | - Angelika Lampert
- Institute of Physiology, RWTH Aachen University Hospital, Aachen, Germany
| |
Collapse
|
6
|
Malhotra A, Weaver J. Approach to Neuropathic Pain. Semin Neurol 2021; 41:744-759. [PMID: 34826876 DOI: 10.1055/s-0041-1726361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Neuropathic pain is a common chief complaint encountered by neurologists and primary care providers. It is caused by disorders involving the somatosensory nervous system. The clinical evaluation of neuropathic pain is challenging and requires a multifaceted systematic approach with an emphasis on a thorough history and physical examination to identify characteristic signs and symptoms. Ancillary laboratory investigations, targeted imaging, and electrodiagnostic studies further help identify underlying etiologies to guide specific treatments. Management of neuropathic pain encompasses treating the underlying pathology as well as symptomatic control with nonpharmacological, pharmacological, and interventional therapies. Here, we present an approach to help evaluate patients with neuropathic pain.
Collapse
Affiliation(s)
- Ashwin Malhotra
- Department of Neurology, NewYork-Presbyterian Hospital/Weill Cornell Medical Center, New York, New York.,Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Joshua Weaver
- Department of Neurology, NewYork-Presbyterian Hospital/Weill Cornell Medical Center, New York, New York
| |
Collapse
|
7
|
Williams A, Villamor L, Fussell J, Loveless R, Smeyne D, Philp J, Shaikh A, Sittaramane V. Discovery of Quinoline-Derived Trifluoromethyl Alcohols as Antiepileptic and Analgesic Agents That Block Sodium Channels. ChemMedChem 2021; 17:e202100547. [PMID: 34632703 DOI: 10.1002/cmdc.202100547] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/30/2021] [Indexed: 11/08/2022]
Abstract
The discovery of novel analgesic agents with high potency, low toxicity and low addictive properties remain a priority. This study aims to identify the analgesic potential of quinoline derived α-trifluoromethylated alcohols (QTA) and their mechanism of action. We synthesized and characterized several compounds of QTAs and screened them for antiepileptic and analgesic activity using zebrafish larvae in high thorough-put behavior analyses system. Toxicity and behavioral screening of 9 compounds (C1-C9) identified four candidates (C2, C3, C7 and C9) with antiepileptic properties that induces specific and reversible reduction in photomotor activity. Importantly, compounds C2 and C3 relieved the thermal pain response in zebrafish larvae indicating analgesic property. Further, using novel in vivo CoroNa green assay, we show that compounds C2 and C3 block sodium channels and reduce inflammatory sodium signals released by peripheral nerve and tissue damage. Thus, we have identified novel QTA compounds with antiepileptic and analgesic properties which could alleviate neuropathic pain.
Collapse
Affiliation(s)
- Ashley Williams
- Department of Biology, Georgia Southern University, Statesboro, GA, 30460, USA
| | - Laurie Villamor
- Department of Biology, Georgia Southern University, Statesboro, GA, 30460, USA
| | - Jake Fussell
- Department of Biology, Georgia Southern University, Statesboro, GA, 30460, USA
| | - Reid Loveless
- Department of Biology, Georgia Southern University, Statesboro, GA, 30460, USA
| | - Dylan Smeyne
- Department of Chemistry and Biochemistry, Georgia Southern University, Statesboro, GA30460, USA
| | - Jack Philp
- Department of Biology, Georgia Southern University, Statesboro, GA, 30460, USA
| | - Abid Shaikh
- Department of Chemistry and Biochemistry, Georgia Southern University, Statesboro, GA30460, USA
| | - Vinoth Sittaramane
- Department of Biology, Georgia Southern University, Statesboro, GA, 30460, USA
| |
Collapse
|
8
|
Monza L, Fumagalli G, Chiorazzi A, Alberti P. Translating morphology from bench side to bed side via neurophysiology: 8-min protocol for peripheral neuropathy research. J Neurosci Methods 2021; 363:109323. [PMID: 34391792 DOI: 10.1016/j.jneumeth.2021.109323] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 08/03/2021] [Accepted: 08/10/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND Peripheral neuropathy treatment is not always satisfactory. To fill this gap, inferences from bench side are warranted, where morphological and pathogenetic determinations can be performed. Nerve conduction studies (NCS) are ideal to translate results from preclinical to clinical setting. NEW METHODS We propose a comprehensive 8-minute protocol for sensory-motor neurophysiological assessment, similar to routine clinical practice: sensory proximal and distal caudal nerves, motor caudal nerve, and sensory digital nerve recordings were used and tested in 2 different experimental settings. In Experiment 1 we compared control (CTRL) animals to a severe sensory-motor polyneuropathy (animals treated with vincristine [VCR]), and in Experiment 2 CTRL animals were compared to a mild sensory polyneuropathy (animals treated with oxaliplatin [OHP]). NCS were performed after 1-month of chemotherapy and matched with confirmatory neuropathological analyses. RESULTS VCR treated animals showed, at NCS, a relevant sensory-motor polyneuropathy ensued at the end of treatment; whereas, OHP animals showed a mild distal sensory neuropathy. These patterns were confirmed by neuropathological analysis. COMPARISON WITH EXISTING METHODS In literature, the majority of proposed neurophysiological protocols relies mainly on a single nerve testing, rather than a combination of them, and only a few studies tested both caudal and sciatic nerve branches, nevertheless not aiming at fully reproduce clinical protocols (e.g., seeking for length-dependency); to provide evidence of appropriateness of our protocol we applied a gold standard: neuropathology. CONCLUSION The simple and rapid protocol here presented can be suggested as a good translation outcome measure in preclinical setting.
Collapse
Affiliation(s)
- Laura Monza
- Experimental Neurology Unit, School of Medicine and Surgery, Monza, Italy; NeuroMI (Milan Center for Neuroscience), Milan, Italy
| | - Giulia Fumagalli
- Experimental Neurology Unit, School of Medicine and Surgery, Monza, Italy; NeuroMI (Milan Center for Neuroscience), Milan, Italy
| | - Alessia Chiorazzi
- Experimental Neurology Unit, School of Medicine and Surgery, Monza, Italy; NeuroMI (Milan Center for Neuroscience), Milan, Italy
| | - Paola Alberti
- Experimental Neurology Unit, School of Medicine and Surgery, Monza, Italy; NeuroMI (Milan Center for Neuroscience), Milan, Italy.
| |
Collapse
|
9
|
Alsaloum M, Labau JIR, Sosniak D, Zhao P, Almomani R, Gerrits M, Hoeijmakers JGJ, Lauria G, Faber CG, Waxman SG, Dib-Hajj S. A novel gain-of-function sodium channel β2 subunit mutation in idiopathic small fiber neuropathy. J Neurophysiol 2021; 126:827-839. [PMID: 34320850 DOI: 10.1152/jn.00184.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Small fiber neuropathy (SFN) is a common condition affecting thinly myelinated Aδ and unmyelinated C fibers, often resulting in excruciating pain and dysautonomia. SFN has been associated with several conditions, but a significant number of cases have no discernible cause. Recent genetic studies have identified potentially pathogenic gain-of-function mutations in several the pore-forming voltage-gated sodium channel α subunits (NaVs) in a subset of patients with SFN, but the auxiliary sodium channel β subunits have been less implicated in the development of the disease. β subunits modulate NaV trafficking and gating, and several mutations have been linked to epilepsy and cardiac dysfunction. Recently, we provided the first evidence for the contribution of a mutation in the β2-subunit to pain in human painful diabetic neuropathy. Here, we provide the first evidence for the involvement of a sodium channel β subunit mutation in the pathogenesis of SFN with no other known causes. We show, through current-clamp analysis, that the newly-identified Y69H variant of the β2 subunit induces neuronal hyperexcitability in dorsal root ganglion neurons, lowering the threshold for action potential firing and allowing for increased repetitive action potential spiking. Underlying the hyperexcitability induced by the β2-Y69H variant, we demonstrate an upregulation in tetrodotoxin-sensitive, but not tetrodotoxin-resistant sodium currents. This provides the first evidence for the involvement of β2 subunits in SFN and strengthens the link between sodium channel β subunits and the development of neuropathic pain in humans.
Collapse
Affiliation(s)
- Matthew Alsaloum
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States.,Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT, United States.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, United States.,Yale Medical Scientist Training Program, Yale School of Medicine, New Haven, CT, United States.,Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT, United States
| | - Julie I R Labau
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States.,Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT, United States.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, United States.,Department of Genetics and Cell Biology, Clinical Genomics Unit, Maastricht University, Maastricht, the Netherlands.,Department of Neurology, Maastricht University Medical Centre+, Maastricht, the Netherlands
| | - Daniel Sosniak
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States.,Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT, United States.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, United States
| | - Peng Zhao
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States.,Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT, United States.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, United States
| | - Rowida Almomani
- Department of Genetics and Cell Biology, Clinical Genomics Unit, Maastricht University, Maastricht, the Netherlands.,Department of Medical Laboratory Sciences, Jordan University of Science and Technology, Irbid, Jordan
| | - Monique Gerrits
- Department of Clinical Genetics, Maastricht University Medical Centre+, Maastricht, the Netherlands
| | | | - Giuseppe Lauria
- Neuroalgology Unit, IRCCS Foundation "Carlo Besta" Neurological Institute, Milan, Italy.,Department of Biomedical and Clinical Sciences "Luigi Sacco," University of Milan, Milan, Italy
| | - Catherina G Faber
- Department of Neurology, Maastricht University Medical Centre+, Maastricht, the Netherlands
| | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States.,Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT, United States.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, United States
| | - Sulayman Dib-Hajj
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States.,Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT, United States.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, United States
| |
Collapse
|
10
|
Yang M, Li Y, Liu L, Zhou M. A novel proline-rich M-superfamily conotoxin that can simultaneously affect sodium, potassium and calcium currents. J Venom Anim Toxins Incl Trop Dis 2021; 27:e20200164. [PMID: 34234819 PMCID: PMC8230863 DOI: 10.1590/1678-9199-jvatitd-2020-0164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 03/01/2021] [Indexed: 12/19/2022] Open
Abstract
Background Conotoxins have become a research hotspot in the neuropharmacology field for their high activity and specificity in targeting ion channels and neurotransmitter receptors. There have been reports of a conotoxin acting on two ion channels, but rare reports of a conotoxin acting on three ion channels. Methods Vr3a, a proline-rich M-superfamily conotoxin from a worm-hunting Conus varius, was obtained by solid-phase synthesis and identified by mass spectrometry. The effects of synthesized Vr3a on sodium, potassium and calcium currents were tested on rat DRG cells by patch clamp experiments. The further effects of Vr3a on human Cav1.2 and Cav2.2 currents were tested on HEK293 cells. Results About 10 μM Vr3a has no effects on the peak sodium currents, but can induce a ~10 mV shift in a polarizing direction in the current-voltage relationship. In addition, 10 μM Vr3a can increase 19.61 ± 5.12% of the peak potassium currents and do not induce a shift in the current-voltage relationship. An amount of 10 μM Vr3a can inhibit 31.26% ± 4.53% of the peak calcium currents and do not induce a shift in the current-voltage relationship. The IC50 value of Vr3a on calcium channel currents in rat DRG neurons is 19.28 ± 4.32 μM. Moreover, 10 μM Vr3a can inhibit 15.32% ± 5.41% of the human Cav1.2 currents and 12.86% ± 4.93% of the human Cav2.2 currents. Conclusions Vr3a can simultaneously affect sodium, potassium and calcium currents. This novel triple-target conotoxin Vr3a expands understanding of conotoxin functions.
Collapse
Affiliation(s)
- Manyi Yang
- Department of Hepatobiliary and Pancreatic Surgery, NHC Key Laboratory of Nanobiological Technology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yubin Li
- Department of Oncology, State Local Joint Engineering Laboratory for Anticancer Drugs, NHC Key Laboratory of Cancer Proteomics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Longfei Liu
- Department of Urology, National Clinical Research Center for Geriatric Disorder, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Maojun Zhou
- Department of Oncology, State Local Joint Engineering Laboratory for Anticancer Drugs, NHC Key Laboratory of Cancer Proteomics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
11
|
Egenolf N, Zu Altenschildesche CM, Kreß L, Eggermann K, Namer B, Gross F, Klitsch A, Malzacher T, Kampik D, Malik RA, Kurth I, Sommer C, Üçeyler N. Diagnosing small fiber neuropathy in clinical practice: a deep phenotyping study. Ther Adv Neurol Disord 2021; 14:17562864211004318. [PMID: 34335876 PMCID: PMC8283814 DOI: 10.1177/17562864211004318] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 03/02/2021] [Indexed: 11/16/2022] Open
Abstract
Background and aims Small fiber neuropathy (SFN) is increasingly suspected in patients with pain of uncertain origin, and making the diagnosis remains a challenge lacking a diagnostic gold standard. Methods In this case-control study, we prospectively recruited 86 patients with a medical history and clinical phenotype suggestive of SFN. Patients underwent neurological examination, quantitative sensory testing (QST), and distal and proximal skin punch biopsy, and were tested for pain-associated gene loci. Fifty-five of these patients additionally underwent pain-related evoked potentials (PREP), corneal confocal microscopy (CCM), and a quantitative sudomotor axon reflex test (QSART). Results Abnormal distal intraepidermal nerve fiber density (IENFD) (60/86, 70%) and neurological examination (53/86, 62%) most frequently reflected small fiber disease. Adding CCM and/or PREP further increased the number of patients with small fiber impairment to 47/55 (85%). Genetic testing revealed potentially pathogenic gene variants in 14/86 (16%) index patients. QST, QSART, and proximal IENFD were of lower impact. Conclusion We propose to diagnose SFN primarily based on the results of neurological examination and distal IENFD, with more detailed phenotyping in specialized centers.
Collapse
Affiliation(s)
- Nadine Egenolf
- Department of Neurology, University of Würzburg, Germany
| | | | - Luisa Kreß
- Department of Neurology, University of Würzburg, Germany
| | - Katja Eggermann
- Institute of Human Genetics, Medical Faculty, RWTH Aachen University, Aachen, Nordrhein-Westfalen, Germany
| | - Barbara Namer
- Institute of Physiology, University of Erlangen, Bayern, Germany
| | | | | | | | - Daniel Kampik
- Department of Ophthalmology, University of Würzburg, Bayern, Germany
| | - Rayaz A Malik
- Weill Cornell Medicine-Qatar, Qatar Foundation, Education City, Doha, Qatar
| | - Ingo Kurth
- Institute of Human Genetics, Medical Faculty, RWTH Aachen University, Aachen, Nordrhein-Westfalen, Germany
| | - Claudia Sommer
- Department of Neurology, University of Würzburg, Germany
| | - Nurcan Üçeyler
- Department of Neurology, University of Würzburg, Josef-Schneider-Str. 11, Würzburg, 97080, Germany
| |
Collapse
|
12
|
Addressing the Need of a Translational Approach in Peripheral Neuropathy Research: Morphology Meets Function. Brain Sci 2021; 11:brainsci11020139. [PMID: 33499072 PMCID: PMC7911498 DOI: 10.3390/brainsci11020139] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 01/08/2021] [Accepted: 01/19/2021] [Indexed: 12/12/2022] Open
Abstract
Peripheral neuropathies (PNs) are a type of common disease that hampers the quality of life of affected people. Treatment, in most cases, is just symptomatic and often ineffective. To improve drug discovery in this field, preclinical evidence is warranted. In vivo rodent models allow a multiparametric approach to test new therapeutic strategies, since they can allow pathogenetic and morphological studies different from the clinical setting. However, human readouts are warranted to promptly translate data from the bench to the bedside. A feasible solution would be neurophysiology, performed similarly at both sides. We describe a simple protocol that reproduces the standard clinical protocol of a neurophysiology hospital department. We devised the optimal montage for sensory and motor recordings (neurography) in mice, and we also implemented F wave testing and a short electromyography (EMG) protocol at rest. We challenged this algorithm by comparing control animals (BALB/c mice) with a model of mild neuropathy to grasp even subtle changes. The neurophysiological results were confirmed with neuropathology. The treatment group showed all expected alterations. Moreover, the neurophysiology matched the neuropathological analyses. Therefore, our protocol can be suggested to promptly translate data from the bench to the bedside and vice versa.
Collapse
|
13
|
Kerth CM, Hautvast P, Körner J, Lampert A, Meents JE. Phosphorylation of a chronic pain mutation in the voltage-gated sodium channel Nav1.7 increases voltage sensitivity. J Biol Chem 2021; 296:100227. [PMID: 33361158 PMCID: PMC7948457 DOI: 10.1074/jbc.ra120.014288] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 12/17/2020] [Accepted: 12/22/2020] [Indexed: 12/19/2022] Open
Abstract
Mutations in voltage-gated sodium channels (Navs) can cause alterations in pain sensation, such as chronic pain diseases like inherited erythromelalgia. The mutation causing inherited erythromelalgia, Nav1.7 p.I848T, is known to induce a hyperpolarized shift in the voltage dependence of activation in Nav1.7. So far, however, the mechanism to explain this increase in voltage sensitivity remains unknown. In the present study, we show that phosphorylation of the newly introduced Thr residue explains the functional change. We expressed wildtype human Nav1.7, the I848T mutant, or other mutations in HEK293T cells and performed whole-cell patch-clamp electrophysiology. As the insertion of a Thr residue potentially creates a novel phosphorylation site for Ser/Thr kinases and because Nav1.7 had been shown in Xenopus oocytes to be affected by protein kinases C and A, we used different nonselective and selective kinase inhibitors and activators to test the effect of phosphorylation on Nav1.7 in a human system. We identify protein kinase C, but not protein kinase A, to be responsible for the phosphorylation of T848 and thereby for the shift in voltage sensitivity. Introducing a negatively charged amino acid instead of the putative phosphorylation site mimics the effect on voltage gating to a lesser extent. 3D modeling using the published cryo-EM structure of human Nav1.7 showed that introduction of this negatively charged site seems to alter the interaction of this residue with the surrounding amino acids and thus to influence channel function. These results could provide new opportunities for the development of novel treatment options for patients with chronic pain.
Collapse
Affiliation(s)
- Clara M Kerth
- Institute of Physiology, Uniklinik RWTH Aachen University, Aachen, Germany
| | - Petra Hautvast
- Institute of Physiology, Uniklinik RWTH Aachen University, Aachen, Germany
| | - Jannis Körner
- Institute of Physiology, Uniklinik RWTH Aachen University, Aachen, Germany; Department of Anesthesiology, Uniklinik RWTH Aachen University, Aachen, Germany
| | - Angelika Lampert
- Institute of Physiology, Uniklinik RWTH Aachen University, Aachen, Germany
| | - Jannis E Meents
- Institute of Physiology, Uniklinik RWTH Aachen University, Aachen, Germany.
| |
Collapse
|
14
|
Solé L, Tamkun MM. Trafficking mechanisms underlying Na v channel subcellular localization in neurons. Channels (Austin) 2020; 14:1-17. [PMID: 31841065 PMCID: PMC7039628 DOI: 10.1080/19336950.2019.1700082] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 11/13/2019] [Indexed: 01/06/2023] Open
Abstract
Voltage gated sodium channels (Nav) play a crucial role in action potential initiation and propagation. Although the discovery of Nav channels dates back more than 65 years, and great advances in understanding their localization, biophysical properties, and links to disease have been made, there are still many questions to be answered regarding the cellular and molecular mechanisms involved in Nav channel trafficking, localization and regulation. This review summarizes the different trafficking mechanisms underlying the polarized Nav channel localization in neurons, with an emphasis on the axon initial segment (AIS), as well as discussing the latest advances regarding how neurons regulate their excitability by modifying AIS length and location. The importance of Nav channel localization is emphasized by the relationship between mutations, impaired trafficking and disease. While this review focuses on Nav1.6, other Nav isoforms are also discussed.
Collapse
Affiliation(s)
- Laura Solé
- Molecular, Cellular and Integrative Neurosciences Graduate Program, Colorado State University, Fort Collins, CO, USA
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Michael M. Tamkun
- Molecular, Cellular and Integrative Neurosciences Graduate Program, Colorado State University, Fort Collins, CO, USA
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO, USA
| |
Collapse
|
15
|
A Systematic Review of Pharmacologic and Rehabilitative Treatment of Small Fiber Neuropathies. Diagnostics (Basel) 2020; 10:diagnostics10121022. [PMID: 33260566 PMCID: PMC7761307 DOI: 10.3390/diagnostics10121022] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 11/08/2020] [Accepted: 11/25/2020] [Indexed: 01/16/2023] Open
Abstract
The aim of this systematic review is to guide the physician in defining the pharmacologic and rehabilitative therapeutic approaches for adopting the best strategies described in the current literature. The search was conducted in PubMed, EMBASE, Cochrane Library and Web of Science to identify the treatment of small fiber neuropathies. Two reviewers independently reviewed and came to a consensus on which articles met inclusion/exclusion criteria. The authors excluded the duplicates, animal studies and included the English articles in which the treatment of patients with small fiber neuropathies was described. The search identified a total of 975 articles with the keywords “small fiber neuropathy” AND “rehabilitation” OR “therapy” OR “treatment”. Seventy-eight selected full-text were analyzed by the reviewers. Forty-two publications met the inclusion criteria and were included in the systematic review to describe the rehabilitative and pharmacologic treatment of small fiber neuropathies. Despite the range of different protocols of treatment for small fiber neuropathy, other robust trials are needed. In addition, always different therapeutic approaches are used; a unique protocol could be important for the clinicians. More research is needed to build evidence for the best strategy and to delineate a definitive therapeutic protocol.
Collapse
|
16
|
Abstract
Small fiber neuropathy (SFN) is a prevalent neurologic syndrome. Testing methods have emerged in recent years to better diagnose it, including autonomic tests and skin punch biopsy. SFN can present in a non-length-dependent fashion and can be mistaken for syndromes such as fibromyalgia and complex regional pain syndrome. SFN is caused by a variety of metabolic, infectious, genetic, and inflammatory diseases. Recently treatments have emerged for TTR amyloid neuropathy and Fabry disease, and novel biomarkers have been found both in genetic and inflammatory SFN syndromes. Ongoing trials attempt to establish the efficacy of intravenous immunoglobulin in inflammatory SFN syndromes.
Collapse
Affiliation(s)
- Lawrence A Zeidman
- Neuromuscular-EMG Division, Department of Neurology, Loyola University Chicago, Loyola University Medical Center, Stritch School of Medicine, 2160 South First Avenue, Maguire Building - Room 2700, Maywood, IL 60153-3328, USA.
| |
Collapse
|
17
|
Yang M, Zhou M. μ-conotoxin TsIIIA, a peptide inhibitor of human voltage-gated sodium channel hNa v1.8. Toxicon 2020; 186:29-34. [PMID: 32758497 DOI: 10.1016/j.toxicon.2020.07.024] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 07/19/2020] [Accepted: 07/22/2020] [Indexed: 10/23/2022]
Abstract
TsIIIA, the first μ-conotoxin from Conus tessulatus, can selectively inhibit rat tetrodotoxin-resistant sodium channels. TsIIIA also shows potent analgesic activity in a mice hotplate analgesic assay, but its effect on human sodium channels remains unknown. In this study, eight human sodium channel subtypes, hNav1.1- hNav1.8, were expressed in HEK293 or ND7/23 cells and tested on the chemically synthesized TsIIIA. Patch clamp experiments showed that 10 μM TsIIIA had no effects on the tetrodotoxin-sensitive hNav1.1, hNav1.2, hNav1.3, hNav1.4, hNav1.6 and hNav1.7, as well as tetrodotoxin-resistant hNav1.5. For tetrodotoxin-resistant hNav1.8, concentrations of 1, 5 and 10 μM TsIIIA reduced the hNav1.8 currents to 59.26%, 36.21% and 24.93% respectively. Further detailed dose-effect experiments showed that TsIIIA inhibited hNav1.8 currents with an IC50 value of 2.11 μM. In addition, 2 μM TsIIIA did not induce a shift in the current-voltage relationship of hNav1.8. Taken together, the hNav1.8 peptide inhibitor TsIIIA provides a pharmacological probe for sodium channels and a potential therapeutic agent for pain.
Collapse
Affiliation(s)
- Manyi Yang
- Department of Hepatobiliary and Pancreatic Surgery, NHC Key Laboratory of Nanobiological Technology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Maojun Zhou
- Department of Oncology, State Local Joint Engineering Laboratory for Anticancer Drugs, NHC Key Laboratory of Cancer Proteomics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| |
Collapse
|
18
|
Devigili G, Cazzato D, Lauria G. Clinical diagnosis and management of small fiber neuropathy: an update on best practice. Expert Rev Neurother 2020; 20:967-980. [PMID: 32654574 DOI: 10.1080/14737175.2020.1794825] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Small fiber neuropathy (SFN) is a heterogeneous group of disorders affecting thin myelinated Aδ and unmyelinated C fibers. Common symptoms include neuropathic pain and autonomic disturbances, and the typical clinical presentation is that of a length-dependent polyneuropathy, although other distributions could be present. AREA COVERED This review focuses on several aspects of SFN including etiology, clinical presentation, diagnostic criteria and tests, management, and future perspectives. Diagnostic challenges are discussed, encompassing the role of accurate and standardized assessment of symptoms and signs and providing clues for the clinical practice. The authors discuss the evidence in support of skin biopsy and quantitative sensory testing as diagnostic tests and present an overview of other diagnostic techniques to assess sensory and autonomic fibers dysfunction. The authors also suggest a systematic approach to the etiology including a set of laboratory tests and genetic examinations of sodium channelopathies and other rare conditions that might drive the therapeutic approach based on underlying cause or symptoms treatment. EXPERT OPINION SFN provides a useful model for neuropathic pain whose known mechanisms and cause could pave the way toward personalized treatments.
Collapse
Affiliation(s)
- Grazia Devigili
- Movement Disorders Unit, Fondazione IRCCS Istituto Neurologico "Carlo Besta" , Milan, Italy
| | - Daniele Cazzato
- Neurophysiology Unit, Fondazione IRCCS Istituto Neurologico "Carlo Besta" , Milan, Italy
| | - Giuseppe Lauria
- Neuroalgology Unit, Fondazione IRCCS Istituto Neurologico "Carlo Besta" , Milan, Italy.,Department of Biomedical and Clinical Sciences "Luigi Sacco", University of Milan , Milan, Italy
| |
Collapse
|
19
|
Labau JIR, Estacion M, Tanaka BS, de Greef BTA, Hoeijmakers JGJ, Geerts M, Gerrits MM, Smeets HJM, Faber CG, Merkies ISJ, Lauria G, Dib-Hajj SD, Waxman SG. Differential effect of lacosamide on Nav1.7 variants from responsive and non-responsive patients with small fibre neuropathy. Brain 2020; 143:771-782. [PMID: 32011655 PMCID: PMC7089662 DOI: 10.1093/brain/awaa016] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 11/13/2019] [Accepted: 12/06/2019] [Indexed: 12/20/2022] Open
Abstract
Small fibre neuropathy is a common pain disorder, which in many cases fails to respond to treatment with existing medications. Gain-of-function mutations of voltage-gated sodium channel Nav1.7 underlie dorsal root ganglion neuronal hyperexcitability and pain in a subset of patients with small fibre neuropathy. Recent clinical studies have demonstrated that lacosamide, which blocks sodium channels in a use-dependent manner, attenuates pain in some patients with Nav1.7 mutations; however, only a subgroup of these patients responded to the drug. Here, we used voltage-clamp recordings to evaluate the effects of lacosamide on five Nav1.7 variants from patients who were responsive or non-responsive to treatment. We show that, at the clinically achievable concentration of 30 μM, lacosamide acts as a potent sodium channel inhibitor of Nav1.7 variants carried by responsive patients, via a hyperpolarizing shift of voltage-dependence of both fast and slow inactivation and enhancement of use-dependent inhibition. By contrast, the effects of lacosamide on slow inactivation and use-dependence in Nav1.7 variants from non-responsive patients were less robust. Importantly, we found that lacosamide selectively enhances fast inactivation only in variants from responders. Taken together, these findings begin to unravel biophysical underpinnings that contribute to responsiveness to lacosamide in patients with small fibre neuropathy carrying select Nav1.7 variants.
Collapse
Affiliation(s)
- Julie I R Labau
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA.,Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA.,Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA.,Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, The Netherlands.,Department of Genetics and Cell Biology, Clinical Genomics Unit, Maastricht University, Maastricht, The Netherlands
| | - Mark Estacion
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA.,Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA.,Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Brian S Tanaka
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA.,Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA.,Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Bianca T A de Greef
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, The Netherlands.,Department of Clinical Epidemiology and Medical Technology Assessment (KEMTA), Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Janneke G J Hoeijmakers
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Margot Geerts
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Monique M Gerrits
- Department of Clinical Genetics, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Hubert J M Smeets
- Department of Genetics and Cell Biology, Clinical Genomics Unit, Maastricht University, Maastricht, The Netherlands
| | - Catharina G Faber
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Ingemar S J Merkies
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, The Netherlands.,Department of Neurology, St. Elisabeth Hospital, Willemstad, Curaçao
| | - Giuseppe Lauria
- Neuroalgology Unit, IRCCS Foundation, "Carlo Besta" Neurological Institute, Milan, Italy.,Department of Biomedical and Clinical Sciences "Luigi Sacco", University of Milan, Italy
| | - Sulayman D Dib-Hajj
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA.,Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA.,Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA.,Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06510, USA.,Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| |
Collapse
|
20
|
Painful and painless mutations of SCN9A and SCN11A voltage-gated sodium channels. Pflugers Arch 2020; 472:865-880. [PMID: 32601768 PMCID: PMC7351857 DOI: 10.1007/s00424-020-02419-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/25/2020] [Accepted: 06/10/2020] [Indexed: 12/11/2022]
Abstract
Chronic pain is a global problem affecting up to 20% of the world’s population and has a significant economic, social and personal cost to society. Sensory neurons of the dorsal root ganglia (DRG) detect noxious stimuli and transmit this sensory information to regions of the central nervous system (CNS) where activity is perceived as pain. DRG neurons express multiple voltage-gated sodium channels that underlie their excitability. Research over the last 20 years has provided valuable insights into the critical roles that two channels, NaV1.7 and NaV1.9, play in pain signalling in man. Gain of function mutations in NaV1.7 cause painful conditions while loss of function mutations cause complete insensitivity to pain. Only gain of function mutations have been reported for NaV1.9. However, while most NaV1.9 mutations lead to painful conditions, a few are reported to cause insensitivity to pain. The critical roles these channels play in pain along with their low expression in the CNS and heart muscle suggest they are valid targets for novel analgesic drugs.
Collapse
|
21
|
Pagliusi M, Bonet IJM, Lemes JBP, Oliveira ALL, Carvalho NS, Tambeli CH, Parada CA, Sartori CR. Social defeat stress-induced hyperalgesia is mediated by nav 1.8 + nociceptive fibers. Neurosci Lett 2020; 729:135006. [PMID: 32387758 DOI: 10.1016/j.neulet.2020.135006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 04/17/2020] [Accepted: 04/20/2020] [Indexed: 10/24/2022]
Abstract
Recently the voltage-gated sodium (Nav) channels began to be studied as possible targets for analgesic drugs. In addition, specific Nav 1.8 blockers are currently being used to treat some types of chronic pain pathologies such as neuropathies and fibromyalgia. Nav 1.8+ fibers convey nociceptive information to brain structures belonging to the limbic system, which is involved in the pathophysiology of major depressive disorders. From this, using a model of chronic social defeat stress (SDS) and intrathecal injections of Nav 1.8 antisense, this study investigated the possible involvement of Nav 1.8+ nociceptive fibers in SDS- induced hyperalgesia in C57/BL mice. Our results showed that SDS induced a depressive-like behavior of social avoidance and increased the sensitivity to mechanical (electronic von Frey test) and chemical (capsaicin test) nociceptive stimuli. We also showed that intrathecal injection of Nav 1.8 antisense reversed the SDS-induced hyperalgesia as demonstrated by both, mechanical and chemical nociceptive tests. We confirmed the antisense efficacy and specificity in a separate no-defeated cohort through real-time PCR, which showed a significant reduction of Nav 1.8 mRNA and no reduction of Nav 1.7 and Nav 1.9 in the L4, L5 and L6 dorsal root ganglia (DRG). The present study advances the understanding of SDS-induced hyperalgesia, which seems to be dependent on Nav 1.8+ nociceptive fibers.
Collapse
Affiliation(s)
- Marco Pagliusi
- Department of Structural and Functional Biology, State University of Campinas, Rua Monteiro Lobato, 255, Cidade Universitaria Zeferino Vaz, Box 6109, Campinas, SP 13083-865, Brazil
| | - Ivan José Magayewski Bonet
- Department of Oral and Maxillofacial Surgery,University of California San Francisco, 513 Parnassus Ave, Box 0440 S709, San Francisco, CA 94143, United States
| | - Júlia Borges Paes Lemes
- Department of Structural and Functional Biology, State University of Campinas, Rua Monteiro Lobato, 255, Cidade Universitaria Zeferino Vaz, Box 6109, Campinas, SP 13083-865, Brazil
| | - Anna Lethicia Lima Oliveira
- Department of Structural and Functional Biology, State University of Campinas, Rua Monteiro Lobato, 255, Cidade Universitaria Zeferino Vaz, Box 6109, Campinas, SP 13083-865, Brazil
| | - Nathalia Santos Carvalho
- Department of Structural and Functional Biology, State University of Campinas, Rua Monteiro Lobato, 255, Cidade Universitaria Zeferino Vaz, Box 6109, Campinas, SP 13083-865, Brazil
| | - Claudia Herrera Tambeli
- Department of Structural and Functional Biology, State University of Campinas, Rua Monteiro Lobato, 255, Cidade Universitaria Zeferino Vaz, Box 6109, Campinas, SP 13083-865, Brazil
| | - Carlos Amilcar Parada
- Department of Structural and Functional Biology, State University of Campinas, Rua Monteiro Lobato, 255, Cidade Universitaria Zeferino Vaz, Box 6109, Campinas, SP 13083-865, Brazil
| | - Cesar Renato Sartori
- Department of Structural and Functional Biology, State University of Campinas, Rua Monteiro Lobato, 255, Cidade Universitaria Zeferino Vaz, Box 6109, Campinas, SP 13083-865, Brazil.
| |
Collapse
|
22
|
|
23
|
Zhao Y, He J, Yu N, Jia C, Wang S. Mechanisms of Dexmedetomidine in Neuropathic Pain. Front Neurosci 2020; 14:330. [PMID: 32431587 PMCID: PMC7214625 DOI: 10.3389/fnins.2020.00330] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 03/20/2020] [Indexed: 12/23/2022] Open
Abstract
Dexmedetomidin is a new-generation, highly selective α2 adrenergic receptor agonist with a large number of advantages, including its sedative and analgesic properties, its ability to inhibit sympathetic nerves, its reduced anesthetic dosage, its hemodynamic stability, its mild respiratory depression abilities, and its ability to improve postoperative recognition. Its safety and effectiveness, as well as its ability to provide a certain degree of comfort to patients, make it a useful anesthetic adjuvant for a wide range of clinical applications. For example, dexmedetomidine is commonly used in patients undergoing general anesthesia, and it also exerts sedative effects during tracheal intubation or mechanical ventilation in intensive care unit patients. In recent years, with the deepening of clinical research on dexmedetomidine, the drug is still applied in the treatment of spastic pain, myofascial pain, neuropathic pain, complex pain syndrome, and chronic headache, as well as for multimodal analgesia. However, we must note that the appropriateness of patient and dose selection should be given attention when using this drug; furthermore, patients should be observed for adverse reactions such as hypotension and bradycardia. Therefore, the safety and effectiveness of this drug for long-term use remain to be studied. In addition, basic experimental studies have also found that dexmedetomidine can protect important organs, such as the brain, heart, kidney, liver, and lung, through various mechanisms, such as antisympathetic effects, the inhibition of apoptosis and oxidative stress, and a reduction in the inflammatory response. Moreover, the neuroprotective properties of dexmedetomidine have received the most attention from scholars. Hence, in this review, we mainly focus on the characteristics and clinical applications of dexmedetomidine, especially the role of dexmedetomidine in the nervous system and the use of dexmedetomidine in the relief of neuropathic pain.
Collapse
Affiliation(s)
- Yang Zhao
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jianshuai He
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ning Yu
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Changxin Jia
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shilei Wang
- Department of Anesthesiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
24
|
Tanabe Y, Shiraishi S, Hashimoto K, Ikeda K, Nishizawa D, Hasegawa J, Shimomura A, Ozaki Y, Tamura N, Yunokawa M, Yonemori K, Takano T, Kawabata H, Tamura K, Fujiwara Y, Shimizu C. Taxane-induced sensory peripheral neuropathy is associated with an SCN9A single nucleotide polymorphism in Japanese patients. BMC Cancer 2020; 20:325. [PMID: 32295642 PMCID: PMC7161266 DOI: 10.1186/s12885-020-06834-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 04/06/2020] [Indexed: 12/26/2022] Open
Abstract
Background Sodium channels located in the dorsal root ganglion, particularly Nav1.7 and Nav1.8, encoded by SCN9A and SCN10A, respectively, act as molecular gatekeepers for pain detection. Our aim was to determine the association between TIPN and SCN9A and SCN10A polymorphisms. Methods Three single nucleotide polymorphisms (SNPs) in SCN9A and two in SCN10A were investigated using whole-genome genotyping data from 186 Japanese breast or ovarian cancer patients classified into two groups as follows: cases that developed taxane-induced grade 2–3 neuropathy (N = 108) and controls (N = 78) with grade 0–1 neuropathy. Multiple logistic regression analyses were conducted to evaluate associations between TIPN and SNP genotypes. Results SCN9A-rs13017637 was a significant predictor of grade 2 or higher TIPN (odds ratio (OR) = 3.463; P = 0.0050) after correction for multiple comparisons, and precision was improved when only breast cancer patients were included (OR 5.053, P = 0.0029). Moreover, rs13017637 was a significant predictor of grade 2 or higher TIPN 1 year after treatment (OR 3.906, P = 0.037), indicating its contribution to TIPN duration. Conclusion SCN9A rs13017637 was associated with the severity and duration of TIPN. These findings are highly exploratory and require replication and validation prior to any consideration of clinical use.
Collapse
Affiliation(s)
- Yuko Tanabe
- Department of Breast and Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan. .,Department of Medical Oncology, Toranomon Hospital, 2-2-2 Toranomon, Minato-ku, Tokyo, 105-8470, Japan.
| | - Seiji Shiraishi
- Department of Anesthesiology, Kohnodai Hospital, National Center for Global Health and Medicine, 1-7-1, Kohnodai, Ichikawa-shi, Chiba, 272-8516, Japan
| | - Kenji Hashimoto
- Department of Breast and Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Kazutaka Ikeda
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo, 156-8506, Japan
| | - Daisuke Nishizawa
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo, 156-8506, Japan
| | - Junko Hasegawa
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo, 156-8506, Japan
| | - Akihiko Shimomura
- Department of Breast and Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan.,Department of Medical Oncology, Toranomon Hospital, 2-2-2 Toranomon, Minato-ku, Tokyo, 105-8470, Japan
| | - Yukinori Ozaki
- Department of Medical Oncology, Toranomon Hospital, 2-2-2 Toranomon, Minato-ku, Tokyo, 105-8470, Japan
| | - Nobuko Tamura
- Department of Breast and Endocrine Surgery, Toranomon Hospital, 2-2-2 Toranomon, Minato-ku, Tokyo, 105-8470, Japan
| | - Mayu Yunokawa
- Department of Breast and Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Kan Yonemori
- Department of Breast and Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Toshimi Takano
- Department of Medical Oncology, Toranomon Hospital, 2-2-2 Toranomon, Minato-ku, Tokyo, 105-8470, Japan
| | - Hidetaka Kawabata
- Department of Breast and Endocrine Surgery, Toranomon Hospital, 2-2-2 Toranomon, Minato-ku, Tokyo, 105-8470, Japan
| | - Kenji Tamura
- Department of Breast and Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Yasuhiro Fujiwara
- Department of Breast and Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Chikako Shimizu
- Department of Breast Medical Oncology, Comprehensive Cancer Center, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, 162-8655, Japan
| |
Collapse
|
25
|
|
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: 29] [Impact Index Per Article: 7.3] [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
|
Kelley MA, Oaklander AL. Association of small-fiber polyneuropathy with three previously unassociated rare missense SCN9A variants. Can J Pain 2020; 4:19-29. [PMID: 32719824 PMCID: PMC7384751 DOI: 10.1080/24740527.2020.1712652] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 12/27/2019] [Accepted: 01/01/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND Small fiber polyneuropathy (SFN) involves ectopic firing and degeneration of small-diameter, somatic/autonomic peripheral axons. Causes include diabetes, inflammation and rare pathogenic mutations, including in SCN9-11 genes that encode small fiber sodium channels. AIMS The aim of this study is to associate a new phenotype-immunotherapy-responsive SFN-with rare amino acid-substituting SCN9A variants and present potential explanations. METHODS A retrospective chart review of two Caucasians with skin biopsy confirmed SFN and rare SCN9A single nucleotide polymorphisms not previously reported in neuropathy. RESULTS A 47-year-old with 4 years of disabling widespread neuropathic pain and exertional intolerance had nerve- and skin biopsy-confirmed SFN, with blood tests revealing only high-titer antinuclear antibodies and low complement C4 consistent with B cell dysimmunity. Six years of intravenous immunoglobulin (IVIg) therapy markedly improved sensory and autonomic symptoms and normalized his neurite density. After whole exome sequencing revealed a potentially pathogenic SCN9A-A3734G variant, sodium channel blockers were tried. Herpes zoster left a 32-year-old with disabling exertional intolerance ("chronic fatigue syndrome"), postural syncope and tachycardia, arm and leg paresthesias, reduced sweating, and distal hairloss. Screening revealed antinuclear and potassium channel autoantibodies, so prednisone and then IVIg were prescribed with great benefit. During 4 years of immunotherapy, his symptoms and function improved, and all abnormal biomarkers (autonomic testing and skin biopsies) normalized. Whole exome sequencing then revealed two nearby compound heterozygous SCN9A variants that were computer-predicted to be deleterious. CONCLUSIONS These cases newly associate three novel amino acid-substituting SCN9A variants with immunotherapy-responsive neuropathy. Only larger studies can determine whether these are contributory or coincidental, but they associate new variants with moderate or high likelihood of pathogenicity with a new highly related phenotype.
Collapse
Affiliation(s)
- Mary A. Kelley
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Neurology, Dell Medical School at the University of Texas, Austin, Texas, USA
| | - Anne Louise Oaklander
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Pathology (Neuropathology), Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
28
|
Marín M, Esteban FJ, Ramírez-Rodrigo H, Ros E, Sáez-Lara MJ. An integrative methodology based on protein-protein interaction networks for identification and functional annotation of disease-relevant genes applied to channelopathies. BMC Bioinformatics 2019; 20:565. [PMID: 31718537 PMCID: PMC6849233 DOI: 10.1186/s12859-019-3162-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 10/15/2019] [Indexed: 12/19/2022] Open
Abstract
Background Biologically data-driven networks have become powerful analytical tools that handle massive, heterogeneous datasets generated from biomedical fields. Protein-protein interaction networks can identify the most relevant structures directly tied to biological functions. Functional enrichments can then be performed based on these structural aspects of gene relationships for the study of channelopathies. Channelopathies refer to a complex group of disorders resulting from dysfunctional ion channels with distinct polygenic manifestations. This study presents a semi-automatic workflow using protein-protein interaction networks that can identify the most relevant genes and their biological processes and pathways in channelopathies to better understand their etiopathogenesis. In addition, the clinical manifestations that are strongly associated with these genes are also identified as the most characteristic in this complex group of diseases. Results In particular, a set of nine representative disease-related genes was detected, these being the most significant genes in relation to their roles in channelopathies. In this way we attested the implication of some voltage-gated sodium (SCN1A, SCN2A, SCN4A, SCN4B, SCN5A, SCN9A) and potassium (KCNQ2, KCNH2) channels in cardiovascular diseases, epilepsies, febrile seizures, headache disorders, neuromuscular, neurodegenerative diseases or neurobehavioral manifestations. We also revealed the role of Ankyrin-G (ANK3) in the neurodegenerative and neurobehavioral disorders as well as the implication of these genes in other systems, such as the immunological or endocrine systems. Conclusions This research provides a systems biology approach to extract information from interaction networks of gene expression. We show how large-scale computational integration of heterogeneous datasets, PPI network analyses, functional databases and published literature may support the detection and assessment of possible potential therapeutic targets in the disease. Applying our workflow makes it feasible to spot the most relevant genes and unknown relationships in channelopathies and shows its potential as a first-step approach to identify both genes and functional interactions in clinical-knowledge scenarios of target diseases. Methods An initial gene pool is previously defined by searching general databases under a specific semantic framework. From the resulting interaction network, a subset of genes are identified as the most relevant through the workflow that includes centrality measures and other filtering and enrichment databases.
Collapse
Affiliation(s)
- Milagros Marín
- Department of Computer Architecture and Technology - CITIC, University of Granada, Granada, Spain.,Department of Biochemistry and Molecular Biology I, University of Granada, Granada, Spain
| | - Francisco J Esteban
- Systems Biology Unit, Department of Experimental Biology, University of Jaén, Jaén, Spain.
| | | | - Eduardo Ros
- Department of Computer Architecture and Technology - CITIC, University of Granada, Granada, Spain
| | - María José Sáez-Lara
- Department of Biochemistry and Molecular Biology I, University of Granada, Granada, Spain.
| |
Collapse
|
29
|
Coates BA, McKenzie JA, Buettmann EG, Liu X, Gontarz PM, Zhang B, Silva MJ. Transcriptional profiling of intramembranous and endochondral ossification after fracture in mice. Bone 2019; 127:577-591. [PMID: 31369916 PMCID: PMC6708791 DOI: 10.1016/j.bone.2019.07.022] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 06/27/2019] [Accepted: 07/18/2019] [Indexed: 12/21/2022]
Abstract
Bone fracture repair represents an important clinical challenge with nearly 1 million non-union fractures occurring annually in the U.S. Gene expression differs between non-union and healthy repair, suggesting there is a pattern of gene expression that is indicative of optimal repair. Despite this, the gene expression profile of fracture repair remains incompletely understood. In this work, we used RNA-seq of two well-established murine fracture models to describe gene expression of intramembranous and endochondral bone formation. We used top differentially expressed genes, enriched gene ontology terms and pathways, callus cellular phenotyping, and histology to describe and contrast these bone formation processes across time. Intramembranous repair, as modeled by ulnar stress fracture, and endochondral repair, as modeled by femur full fracture, exhibited vastly different transcriptional profiles throughout repair. Stress fracture healing had enriched differentially expressed genes associated with bone repair and osteoblasts, highlighting the strong osteogenic repair process of this model. Interestingly, the PI3K-Akt signaling pathway was one of only a few pathways uniquely enriched in stress fracture repair. Full fracture repair involved a higher level of inflammatory and immune cell related genes than did stress fracture repair. Full fracture repair also differed from stress fracture in a robust downregulation of ion channel genes following injury, the role of which in fracture repair is unclear. This study offers a broad description of gene expression in intramembranous and endochondral ossification across several time points throughout repair and suggests several potentially intriguing genes, pathways, and cells whose role in fracture repair requires further study.
Collapse
Affiliation(s)
- Brandon A Coates
- Department of Orthopaedic Surgery, Washington University in St. Louis, MO, United States of America; Department of Biomedical Engineering, Washington University in St. Louis, MO, United States of America.
| | - Jennifer A McKenzie
- Department of Orthopaedic Surgery, Washington University in St. Louis, MO, United States of America
| | - Evan G Buettmann
- Department of Orthopaedic Surgery, Washington University in St. Louis, MO, United States of America; Department of Biomedical Engineering, Washington University in St. Louis, MO, United States of America
| | - Xiaochen Liu
- Department of Orthopaedic Surgery, Washington University in St. Louis, MO, United States of America
| | - Paul M Gontarz
- Department of Developmental Biology, Washington University in St. Louis, MO, United States of America
| | - Bo Zhang
- Department of Developmental Biology, Washington University in St. Louis, MO, United States of America
| | - Matthew J Silva
- Department of Orthopaedic Surgery, Washington University in St. Louis, MO, United States of America; Department of Biomedical Engineering, Washington University in St. Louis, MO, United States of America
| |
Collapse
|
30
|
de Greef BTA, Hoeijmakers JGJ, Geerts M, Oakes M, Church TJE, Waxman SG, Dib-Hajj SD, Faber CG, Merkies ISJ. Lacosamide in patients with Nav1.7 mutations-related small fibre neuropathy: a randomized controlled trial. Brain 2019; 142:263-275. [PMID: 30649227 DOI: 10.1093/brain/awy329] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Accepted: 11/02/2018] [Indexed: 11/14/2022] Open
Abstract
Symptomatic treatment of neuropathic pain in small fibre neuropathy is often disappointing. The finding of voltage-gated sodium channel mutations in small fibre neuropathy (with mutations in SCN9A, encoding for Nav1.7) being most frequently reported suggest a specific target for therapy. The anticonvulsant lacosamide acts on Nav1.3, Nav1.7, and Nav1.8. The aim of this study was to evaluate the efficacy, safety, and tolerability of lacosamide as a potential treatment for pain in Nav1.7-related small fibre neuropathy. The Lacosamide-Efficacy-'N'-Safety in SFN (LENSS) was a randomized, placebo-controlled, double-blind, crossover-design study. Subjects were recruited in the Netherlands between November 2014 and July 2016. Patients with Nav1.7-related small fibre neuropathy were randomized to start with lacosamide followed by placebo or vice versa. In both 8-week treatment phases, patients received 200 mg two times a day (BID), preceded by a titration period, and ended by a tapering period. The primary outcome was efficacy, defined as the proportion of patients with 1-point average pain score reduction compared to baseline using the Pain Intensity Numerical Rating Scale. The trial is registered with ClinicalTrials.gov, number NCT01911975. Twenty-four subjects received lacosamide, and 23 received placebo. In 58.3% of patients receiving lacosamide, mean average pain decreased by at least 1 point, compared to 21.7% in the placebo group [sensitivity analyses, odds ratio 5.65 (95% confidence interval: 1.83-17.41); P = 0.0045]. In the lacosamide group, 33.3% reported that their general condition improved versus 4.3% in the placebo group (P-value = 0.0156). Additionally, a significant decrease in daily sleep interference, and in surface pain intensity was demonstrated. No significant changes in quality of life or autonomic symptoms were found. Lacosamide was well tolerated and safe in use. This study shows that lacosamide has a significant effect on pain, general wellbeing, and sleep quality. Lacosamide was well tolerated and safe, suggesting that it can be used for pain treatment in Nav1.7-related small fibre neuropathy.
Collapse
Affiliation(s)
- Bianca T A de Greef
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Janneke G J Hoeijmakers
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Margot Geerts
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Mike Oakes
- ParamStat Limited, University of Sussex, Fairlight, East Sussex, UK
| | | | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA.,Center for Neuroscience and Regeneration Research, Veterans Affairs Medical Center, West Haven, Connecticut, USA
| | - Sulayman D Dib-Hajj
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA.,Center for Neuroscience and Regeneration Research, Veterans Affairs Medical Center, West Haven, Connecticut, USA
| | - Catharina G Faber
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Ingemar S J Merkies
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Center+, Maastricht, The Netherlands.,Department of Neurology, St. Elisabeth Hospital, Willemstad, Curaçao
| |
Collapse
|
31
|
Dusch M, Schmelz M. [Erythromelalgia: skin redness and pain]. Schmerz 2019; 33:475-490. [PMID: 31485751 DOI: 10.1007/s00482-019-00401-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Erythromelalgia is a rare disease that is associated with hemato-oncological diseases or after taking certain drugs and toxins, but it can also occur as an independent clinical picture, for example, due to mutations in the sodium channel NaV1.7. Clinically, there is a characteristic triad of attack-like burning pain and skin redness in the area of the distal extremities, which can be alleviated by excessive cooling. The attacks are triggered by heat, exertion, and stress. The diagnosis is primarily made clinically and can be confirmed by genetic testing if a sodium channel NaV1.7 mutation is present. Important differential diagnoses are complex regional pain syndrome, the non-freezing cold injury, and small fiber neuropathies. Therapy is multidisciplinary and has to be planned individually and include physical therapy and psychotherapy as well as drug therapy as integral components.
Collapse
Affiliation(s)
- M Dusch
- Klinik für Anästhesiologie und Intensivmedizin, Fachbereich Schmerzmedizin, Medizinische Hochschule Hannover, Carl-Neuberg-Straße 1, 30625, Hannover, Deutschland.
| | - M Schmelz
- Abteilung Experimentelle Schmerzforschung, CBTM, Medizinische Fakultät Mannheim, Universität Heidelberg, Mannheim, Deutschland
| |
Collapse
|
32
|
Blitshteyn S, Brinth L, Hendrickson JE, Martinez-Lavin M. Autonomic dysfunction and HPV immunization: an overview. Immunol Res 2019; 66:744-754. [PMID: 30478703 DOI: 10.1007/s12026-018-9036-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
This article reviews the case series reported from several countries describing patients with suspected severe side effects to the HPV vaccines. The described symptom clusters are remarkably similar and include disabling fatigue, headache, widespread pain, fainting, gastrointestinal dysmotility, limb weakness, memory impairment episodes of altered awareness, and abnormal movements. This constellation of symptoms and signs has been labeled with different diagnoses such as complex regional pain syndrome (CRPS), postural orthostatic tachycardia syndrome (POTS), small fiber neuropathy (SFN), myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS), or fibromyalgia. It is known that autoimmunity and autoantibodies are present in a subset of patients with CRPS, POTS, SFN, ME/CFS, and fibromyalgia. This article proposes that vaccine-triggered, immune-mediated autonomic dysfunction could lead to the development of de novo post-HPV vaccination syndrome possibly in genetically susceptible individuals. Being cognizant that a temporal relationship between vaccination and symptom onset does not necessarily equate to causality, mounting evidence of case series calls for well-designed case-control studies to determine the prevalence and possible causation between these symptom clusters and HPV vaccines. Since personalized medicine is gaining momentum, the use of adversomics and pharmacogenetics may eventually help identify individuals who are predisposed to HPV vaccine adverse events.
Collapse
Affiliation(s)
- Svetlana Blitshteyn
- Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Louise Brinth
- Syncope Unit, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
| | - Jeanne E Hendrickson
- Laboratory Medicine and Pediatrics, Yale University School of Medicine, New Haven, CT, USA
| | | |
Collapse
|
33
|
Abstract
Acute pain is adaptive, but chronic pain is a global challenge. Many chronic pain syndromes are peripheral in origin and reflect hyperactivity of peripheral pain-signaling neurons. Current treatments are ineffective or only partially effective and in some cases can be addictive, underscoring the need for better therapies. Molecular genetic studies have now linked multiple human pain disorders to voltage-gated sodium channels, including disorders characterized by insensitivity or reduced sensitivity to pain and others characterized by exaggerated pain in response to normally innocuous stimuli. Here, we review recent developments that have enhanced our understanding of pathophysiological mechanisms in human pain and advances in targeting sodium channels in peripheral neurons for the treatment of pain using novel and existing sodium channel blockers.
Collapse
Affiliation(s)
- Sulayman D Dib-Hajj
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut 06510, USA; .,Rehabilitation Research Center, Veterans Affairs, Connecticut Healthcare System, West Haven, Connecticut 06516, USA
| | - Stephen G Waxman
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut 06510, USA; .,Rehabilitation Research Center, Veterans Affairs, Connecticut Healthcare System, West Haven, Connecticut 06516, USA
| |
Collapse
|
34
|
Belinskaia DA, Belinskaia MA, Barygin OI, Vanchakova NP, Shestakova NN. Psychotropic Drugs for the Management of Chronic Pain and Itch. Pharmaceuticals (Basel) 2019; 12:ph12020099. [PMID: 31238561 PMCID: PMC6631469 DOI: 10.3390/ph12020099] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Revised: 06/19/2019] [Accepted: 06/21/2019] [Indexed: 12/11/2022] Open
Abstract
Clinical observations have shown that patients with chronic neuropathic pain or itch exhibit symptoms of increased anxiety, depression and cognitive impairment. Such patients need corrective therapy with antidepressants, antipsychotics or anticonvulsants. It is known that some psychotropic drugs are also effective for the treatment of neuropathic pain and pruritus syndromes due to interaction with the secondary molecular targets. Our own clinical studies have identified antipruritic and/or analgesic efficacy of the following compounds: tianeptine (atypical tricyclic antidepressant), citalopram (selective serotonin reuptake inhibitor), mianserin (tetracyclic antidepressant), carbamazepine (anticonvulsant), trazodone (serotonin antagonist and reuptake inhibitor), and chlorprothixene (antipsychotic). Venlafaxine (serotonin-norepinephrine reuptake inhibitor) is known to have an analgesic effect too. The mechanism of such effect of these drugs is not fully understood. Herein we review and correlate the literature data on analgesic/antipruritic activity with pharmacological profile of these compounds.
Collapse
Affiliation(s)
- Daria A Belinskaia
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, pr. Torez 44, St. Petersburg 194223, Russia.
| | - Mariia A Belinskaia
- International Centre for Neurotherapeutics, Dublin City University, Glasnevin, Dublin 9, Ireland.
| | - Oleg I Barygin
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, pr. Torez 44, St. Petersburg 194223, Russia.
| | - Nina P Vanchakova
- Department of Pedagogy and Psychology, Faculty of Postgraduate Education, First Pavlov State Medical University, L'va Tolstogo str. 6-8, St. Petersburg 197022, Russia.
| | - Natalia N Shestakova
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, pr. Torez 44, St. Petersburg 194223, Russia.
| |
Collapse
|
35
|
Nociceptor Signalling through ion Channel Regulation via GPCRs. Int J Mol Sci 2019; 20:ijms20102488. [PMID: 31137507 PMCID: PMC6566991 DOI: 10.3390/ijms20102488] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 05/08/2019] [Accepted: 05/13/2019] [Indexed: 12/23/2022] Open
Abstract
The prime task of nociceptors is the transformation of noxious stimuli into action potentials that are propagated along the neurites of nociceptive neurons from the periphery to the spinal cord. This function of nociceptors relies on the coordinated operation of a variety of ion channels. In this review, we summarize how members of nine different families of ion channels expressed in sensory neurons contribute to nociception. Furthermore, data on 35 different types of G protein coupled receptors are presented, activation of which controls the gating of the aforementioned ion channels. These receptors are not only targeted by more than 20 separate endogenous modulators, but can also be affected by pharmacotherapeutic agents. Thereby, this review provides information on how ion channel modulation via G protein coupled receptors in nociceptors can be exploited to provide improved analgesic therapy.
Collapse
|
36
|
Eijkenboom I, Sopacua M, Hoeijmakers JGJ, de Greef BTA, Lindsey P, Almomani R, Marchi M, Vanoevelen J, Smeets HJM, Waxman SG, Lauria G, Merkies ISJ, Faber CG, Gerrits MM. Yield of peripheral sodium channels gene screening in pure small fibre neuropathy. J Neurol Neurosurg Psychiatry 2019; 90:342-352. [PMID: 30554136 DOI: 10.1136/jnnp-2018-319042] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 11/01/2018] [Accepted: 11/18/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND Neuropathic pain is common in peripheral neuropathy. Recent genetic studies have linked pathogenic voltage-gated sodium channel (VGSC) variants to human pain disorders. Our aims are to determine the frequency of SCN9A, SCN10A and SCN11A variants in patients with pure small fibre neuropathy (SFN), analyse their clinical features and provide a rationale for genetic screening. METHODS Between September 2009 and January 2017, 1139 patients diagnosed with pure SFN at our reference centre were screened for SCN9A, SCN10A and SCN11A variants. Pathogenicity of variants was classified according to established guidelines of the Association for Clinical Genetic Science and frequencies were determined. Patients with SFN were grouped according to the VGSC variants detected, and clinical features were compared. RESULTS Among 1139 patients with SFN, 132 (11.6%) patients harboured 73 different (potentially) pathogenic VGSC variants, of which 50 were novel and 22 were found in ≥ 1 patient. The frequency of (potentially) pathogenic variants was 5.1% (n=58/1139) for SCN9A, 3.7% (n=42/1139) for SCN10A and 2.9% (n=33/1139) for SCN11A. Only erythromelalgia-like symptoms and warmth-induced pain were significantly more common in patients harbouring VGSC variants. CONCLUSION (Potentially) pathogenic VGSC variants are present in 11.6% of patients with pure SFN. Therefore, genetic screening of SCN9A, SCN10A and SCN11A should be considered in patients with pure SFN, independently of clinical features or underlying conditions.
Collapse
Affiliation(s)
- Ivo Eijkenboom
- Department of Genetics and Cell Biology, Clinical Genomics Unit, Maastricht University, Maastricht, The Netherlands.,MHeNs School of Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Maurice Sopacua
- MHeNs School of Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands.,Department of Neurology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Janneke G J Hoeijmakers
- MHeNs School of Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands.,Department of Neurology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Bianca T A de Greef
- MHeNs School of Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands.,Department of Neurology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Patrick Lindsey
- Department of Genetics and Cell Biology, Clinical Genomics Unit, Maastricht University, Maastricht, The Netherlands
| | - Rowida Almomani
- Department of Genetics and Cell Biology, Clinical Genomics Unit, Maastricht University, Maastricht, The Netherlands.,MHeNs School of Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands.,Department of Medical Laboratory Sciences, Jordan University of Science and Technology, Irbid, Jordan
| | - Margherita Marchi
- Neuroalgology Unit, IRCCS Fondazione Istituto Neurologico "Carlo Besta", Milan, Italy
| | - Jo Vanoevelen
- Department of Genetics and Cell Biology, Clinical Genomics Unit, Maastricht University, Maastricht, The Netherlands
| | - Hubertus J M Smeets
- Department of Genetics and Cell Biology, Clinical Genomics Unit, Maastricht University, Maastricht, The Netherlands.,MHeNs School of Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Stephen G Waxman
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut, USA.,Centre for Neuroscience and Regeneration Research, Veterans Affairs Medical Center, West Haven, Connecticut, USA
| | - Giuseppe Lauria
- Neuroalgology Unit, IRCCS Fondazione Istituto Neurologico "Carlo Besta", Milan, Italy.,Department of Biomedical and Clinical Sciences "Luigi Sacco", University of Milan, Milan, Italy
| | - Ingemar S J Merkies
- MHeNs School of Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands.,Department of Neurology, St. Elisabeth Hospital, Willemstad, Curaçao
| | - Catharina G Faber
- MHeNs School of Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands.,Department of Neurology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Monique M Gerrits
- Department of Clinical Genetics, Maastricht University Medical Centre, Maastricht, The Netherlands
| |
Collapse
|
37
|
Abstract
Persistent, in particular neuropathic pain affects millions of people worldwide. However, the response rate of patients to existing analgesic drugs is less than 50%. There are several possibilities to increase this response rate, such as optimization of the pharmacokinetic and pharmacodynamic properties of analgesics. Another promising approach is to use prognostic biomarkers in patients to determine the optimal pharmacological therapy for each individual. Here, we discuss recent efforts to identify plasma and CSF biomarkers, as well as genetic biomarkers and sensory testing, and how these readouts could be exploited for the prediction of a suitable pharmacological treatment. Collectively, the information on single biomarkers may be stored in knowledge bases and processed by machine-learning and related artificial intelligence techniques, resulting in the optimal pharmacological treatment for individual pain patients. We highlight the potential for biomarker-based individualized pain therapies and discuss biomarker reliability and their utility in clinical practice, as well as limitations of this approach.
Collapse
|
38
|
Sopacua M, Hoeijmakers JGJ, Merkies ISJ, Lauria G, Waxman SG, Faber CG. Small‐fiber neuropathy: Expanding the clinical pain universe. J Peripher Nerv Syst 2019; 24:19-33. [DOI: 10.1111/jns.12298] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 11/27/2018] [Accepted: 12/14/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Maurice Sopacua
- Department of Neurology, School of Mental Health and NeuroscienceMaastricht University Medical Centre+ Maastricht The Netherlands
| | - Janneke G. J. Hoeijmakers
- Department of Neurology, School of Mental Health and NeuroscienceMaastricht University Medical Centre+ Maastricht The Netherlands
| | - Ingemar S. J. Merkies
- Department of Neurology, School of Mental Health and NeuroscienceMaastricht University Medical Centre+ Maastricht The Netherlands
- Department of NeurologySt. Elisabeth Hospital Willemstad Curaçao
| | - Giuseppe Lauria
- Neuroalgology UnitIRCCS Foundation, “Carlo Besta” Neurological Institute Milan Italy
- Department of Biomedical and Clinical Sciences “Luigi Sacco”University of Milan Milan Italy
| | - Stephen G. Waxman
- Department of NeurologyYale University School of Medicine New Haven Connecticut
- Center for Neuroscience and Regeneration ResearchVA Connecticut Healthcare System West Haven Connecticut
| | - Catharina G. Faber
- Department of Neurology, School of Mental Health and NeuroscienceMaastricht University Medical Centre+ Maastricht The Netherlands
| |
Collapse
|
39
|
Brouwer BA, van Kuijk SMJ, Bouwhuis A, Faber CG, van Kleef M, Merkies ISJ, Hoeijmakers JGJ. The Pain Dynamics of Small Fiber Neuropathy. THE JOURNAL OF PAIN 2018; 20:655-663. [PMID: 30529697 DOI: 10.1016/j.jpain.2018.11.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Revised: 10/25/2018] [Accepted: 11/13/2018] [Indexed: 01/19/2023]
Abstract
Pain is a central feature in small fiber neuropathy (SFN), with only moderate effects of pharmacologic treatment. The evaluation of the efficacy of therapies on pain has been driven by static measures, and a circadian cycle has been suggested. The aim of this study is to evaluate the pain dynamics in SFN. A total of 165 patients completed a standardized pain diary 4 times per week over a 4-week period. Patients used the 11-point numeric rating scale for average diurnal, nocturnal, and maximum pain, taking into account the circumstances in which pain emerged most. Medication used, SFN-related complaints, sleep quality, and anatomic location of pain were also assessed. Neuropathic pain showed a length-dependent pattern. For pain intensity, marginal higher pain scores at night than during the day were shown, likely not clinically meaningful, with stable lower pain intensities in weekends compared with weekdays. The average pain intensity was stable during the 4-week period. Maximum pain was experienced mainly at rest and during sleep. Besides, pain intensity at night showed an inverse relationship with quality of sleep. In conclusion, for trials in SFN it is justified to use pain scores from any time of the day during short measurement periods. PERSPECTIVE: This article presents for the first time the pain dynamics of SFN. The effect of pharmacologic treatment in SFN is often disappointing, partly owing to insufficient trial designs. The results of this study have added value in the development of new proper clinical trials in SFN.
Collapse
Affiliation(s)
| | - Sander M J van Kuijk
- Department of Clinical Epidemiology and Medical Technology Assessment, Maastricht University Medical Center+, 6202 AZ Maastricht, the Netherlands
| | - Anne Bouwhuis
- Department of Anesthesiology, VU University Medical Center, 1081 HV Amsterdam, the Netherlands
| | - Catharina G Faber
- Department of Neurology, Maastricht University Medical Center+, 6202 AZ Maastricht, the Netherlands
| | | | - Ingemar S J Merkies
- Department of Neurology, Maastricht University Medical Center+, 6202 AZ Maastricht, the Netherlands; Department of Neurology, St. Elisabeth Hospital, Willemstad, Curaçao
| | - Janneke G J Hoeijmakers
- Department of Neurology, Maastricht University Medical Center+, 6202 AZ Maastricht, the Netherlands.
| |
Collapse
|
40
|
|
41
|
Chang KV, Mezian K, Naňka O, Wu WT, Lou YM, Wang JC, Martinoli C, Özçakar L. Ultrasound Imaging for the Cutaneous Nerves of the Extremities and Relevant Entrapment Syndromes: From Anatomy to Clinical Implications. J Clin Med 2018; 7:E457. [PMID: 30469370 PMCID: PMC6262579 DOI: 10.3390/jcm7110457] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 11/18/2018] [Accepted: 11/19/2018] [Indexed: 12/31/2022] Open
Abstract
Cutaneous nerve entrapment plays an important role in neuropathic pain syndrome. Due to the advancement of ultrasound technology, the cutaneous nerves can be visualized by high-resolution ultrasound. As the cutaneous nerves course superficially in the subcutaneous layer, they are vulnerable to entrapment or collateral damage in traumatic insults. Scanning of the cutaneous nerves is challenging due to fewer anatomic landmarks for referencing. Therefore, the aim of the present article is to summarize the anatomy of the limb cutaneous nerves, to elaborate the scanning techniques, and also to discuss the clinical implications of pertinent entrapment syndromes of the medial brachial cutaneous nerve, intercostobrachial cutaneous nerve, medial antebrachial cutaneous nerve, lateral antebrachial cutaneous nerve, posterior antebrachial cutaneous nerve, superficial branch of the radial nerve, dorsal cutaneous branch of the ulnar nerve, palmar cutaneous branch of the median nerve, anterior femoral cutaneous nerve, posterior femoral cutaneous nerve, lateral femoral cutaneous nerve, sural nerve, and saphenous nerve.
Collapse
Affiliation(s)
- Ke-Vin Chang
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital, Bei-Hu Branch, Taipei 10845, Taiwan.
- Department of Physical Medicine and Rehabilitation, National Taiwan University College of Medicine, Taipei 10048, Taiwan.
| | - Kamal Mezian
- Department of Rehabilitation Medicine, Charles University, First Faculty of Medicine, 12800 Prague, Czech Republic.
| | - Ondřej Naňka
- Institute of Anatomy, Charles University, First Faculty of Medicine, 12800 Prague, Czech Republic.
| | - Wei-Ting Wu
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital, Bei-Hu Branch, Taipei 10845, Taiwan.
| | - Yueh-Ming Lou
- Department of Physical Medicine and Rehabilitation, National Taiwan University College of Medicine, Taipei 10048, Taiwan.
| | - Jia-Chi Wang
- Department of Physical Medicine and Rehabilitation, Taipei Veterans General Hospital, Taipei 11217, Taiwan.
| | - Carlo Martinoli
- Department of Health Sciences (DISSAL), University of Genoa, 16132 Genoa, Italy.
| | - Levent Özçakar
- Department of Physical and Rehabilitation Medicine, Hacettepe University Medical School, 06100 Ankara, Turkey.
| |
Collapse
|
42
|
Abstract
PURPOSE OF REVIEW This article describes the methods of diagnosis and management of the sensory-predominant polyneuropathies. To simplify the approach to this category of patients, sensory-predominant polyneuropathies are divided broadly into either small fiber (or pain-predominant) neuropathies and large fiber (or ataxia-predominant) neuropathies, of which the sensory neuronopathies (dorsal root ganglionopathies) are highlighted. RECENT FINDINGS Physicians can now easily perform skin biopsies in their offices, allowing access to the gold standard pathologic diagnostic tool for small fiber neuropathies. Additional diagnostic techniques, such as corneal confocal microscopy, are emerging. Recently, small fiber neuropathies have been associated with a broader spectrum of diseases, including fibromyalgia, sodium channel mutations, and voltage-gated potassium channel antibody autoimmune disease. SUMMARY Despite advances in diagnosing small fiber neuropathies and sensory neuronopathies, many of these neuropathies remain refractory to treatment. In select cases, early identification and treatment may result in better outcomes. "Idiopathic" should be a diagnosis of exclusion and a thorough investigation for treatable causes pursued.
Collapse
|
43
|
Lurati AR. How a Simple Ankle Sprain Turned Into Neuropathic Pain: Complex Reflex Sympathetic Dystrophy Versus Erythromelalgia. Workplace Health Saf 2018; 66:169-172. [DOI: 10.1177/2165079917736786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
A 36-year-old woman sustained a Grade 2 ankle sprain at work. Two days after the injury, the ankle and foot became red and she complained of “intense burning pain.” First diagnosed with complex reflex sympathetic dystrophy, the employee was prescribed medications that provided some pain relief; a subsequent temporary nerve block provided additional relief. However, the symptoms returned and she was treated unsuccessfully with surgical sympathectomy. The employee was referred to a neurologist and diagnosed with primary erythromelalgia, a rare pain disorder that can be mistaken as complex reflex sympathetic dystrophy.
Collapse
|
44
|
de Greef BTA, Hoeijmakers JGJ, Gorissen‐Brouwers CML, Geerts M, Faber CG, Merkies ISJ. Associated conditions in small fiber neuropathy - a large cohort study and review of the literature. Eur J Neurol 2018; 25:348-355. [PMID: 29112785 PMCID: PMC5814938 DOI: 10.1111/ene.13508] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 11/02/2017] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND PURPOSE Small fiber neuropathy (SFN) is a common disorder leading to neuropathic pain and autonomic symptoms. The objective of this study was to investigate associated conditions in a large cohort of SFN patients and compare the prevalence to healthy individuals. METHODS A total of 921 patients with pure SFN were screened according to a standardized comprehensive diagnostic algorithm and compared with literature findings. RESULTS No associated condition could be found in 53% of the patients. Autoimmune diseases, sodium channel gene mutations, diabetes mellitus including glucose intolerance, and vitamin B12 deficiencies were more prevalent than reported literature findings, followed by alcohol abuse, chemotherapy, monoclonal gammopathy of undetermined significance, and haemochromatosis. In patients who were already known with a possible underlying condition at screening, additional underlying conditions were still found in another 26.7% of patients. CONCLUSIONS Based on these results, it is recommended that patients with pure SFN are screened at least for autoimmune diseases, sodium channel gene mutations, diabetes mellitus including glucose intolerance, and vitamin B12 deficiency, even when they already have a potential underlying condition at referral.
Collapse
Affiliation(s)
- B. T. A. de Greef
- Department of NeurologySchool of Mental Health and NeuroscienceMaastricht University Medical Center+MaastrichtThe Netherlands
| | - J. G. J. Hoeijmakers
- Department of NeurologySchool of Mental Health and NeuroscienceMaastricht University Medical Center+MaastrichtThe Netherlands
| | - C. M. L. Gorissen‐Brouwers
- Department of NeurologySchool of Mental Health and NeuroscienceMaastricht University Medical Center+MaastrichtThe Netherlands
| | - M. Geerts
- Department of NeurologySchool of Mental Health and NeuroscienceMaastricht University Medical Center+MaastrichtThe Netherlands
| | - C. G. Faber
- Department of NeurologySchool of Mental Health and NeuroscienceMaastricht University Medical Center+MaastrichtThe Netherlands
| | - I. S. J. Merkies
- Department of NeurologySchool of Mental Health and NeuroscienceMaastricht University Medical Center+MaastrichtThe Netherlands
- Department of NeurologySt Elisabeth HospitalWillemstadCuraçao
| |
Collapse
|
45
|
Abstract
Erythromelalgia is a rare syndrome characterized by the intermittent or, less commonly, by the permanent occurrence of extremely painful hyperperfused skin areas mainly located in the distal extremities. Primary erythromelalgia is nowadays considered to be a genetically determined neuropathic disorder affecting SCN9A, SCN10A, and SCN11A coding for NaV1.7, NaV1.8, and NaV1.9 neuronal sodium channels. Secondary forms might be associated with myeloproliferative disorders, connective tissue disease, cancer, infections, and poisoning. Between the pain episodes, the affected skin areas are usually asymptomatic, but there are patients with typical features of acrocyanosis and/or Raynaud's phenomenon preceding or occurring in between the episodes of erythromelalgia. Diagnosis is made by ascertaining the typical clinical features. Thereafter, the differentiation between primary and secondary forms should be made. Genetic testing is recommended, especially in premature cases and in cases of family clustering in specialized genetic institutions after genetic counselling. Multimodal therapeutic intervention aims toward attenuation of pain and improvement of the patient's quality of life. For this purpose, a wide variety of nonpharmacological approaches and pharmacological substances for topical and systemic use have been proposed, which are usually applied individually in a step-by-step approach. Prognosis mainly depends on the underlying condition and the ability of the patients and their relatives to cope with the disease.
Collapse
Affiliation(s)
| | | | - Jutta Gisela Richter
- 2 Poliklinik, Funktionsbereich und Hiller Forschungszentrum für Rheumatologie, Medizinische Fakultät, Universitätsklinikum Düsseldorf, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
46
|
Abstract
The low prevalence of erythromelalgia, classified as an orphan disease, poses diagnostic and therapeutic difficulties. The aim of this review is to be an update of the specialized bibliography. Erythromelalgia is an infrequent episodic acrosyndrome affecting mainly both lower limbs symmetrically with the classic triad of erythema, warmth and burning pain. Primary erythromelalgia is an autosomal dominant inherited disorder, while secondary is associated with myeloproliferative diseases, among others. In its etiopathogenesis, there are neural and vascular abnormalities that can be combined. The diagnosis is based on exhaustive clinical history and physical examination. Complications are due to changes in the skin barrier function, ischemia and compromise of cutaneous nerves. Because of the complexity of its pathogenesis, erythromelalgia should always be included in the differential diagnosis of conditions that cause chronic pain and/or peripheral edema. The prevention of crisis is based on a strict control of triggers and promotion of preventive measures. Since there is no specific and effective treatment, control should focus on the underlying disease. However, there are numerous topical and systemic therapies that patients can benefit from.
Collapse
|
47
|
Translational Model Systems for Complex Sodium Channel Pathophysiology in Pain. Handb Exp Pharmacol 2018; 246:355-369. [PMID: 29374838 DOI: 10.1007/164_2017_91] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Chronic pain patients are often left with insufficient treatment as the pathophysiology especially of neuropathic pain remains enigmatic. Recently, genetic variations in the genes of the voltage-gated sodium channels (Navs) were linked to inherited neuropathic pain syndromes, opening a research pathway to foster our understanding of the pathophysiology of neuropathic pain. More than 10 years ago, the rare, inherited pain syndrome erythromelalgia was linked to mutations in the subtype Nav1.7, and since then a plethora of mutations and genetic variations in this and other Nav genes were identified. Often the biophysical changes induced by the genetic alteration offer a straightforward explanation for the clinical symptoms, but mutations in some channels, especially Nav1.9, paint a more complex picture. Although efforts were undertaken to significantly advance our knowledge, translation from heterologous or animal model systems to humans remains a challenge. Here we present recent advances in translation using stem cell-derived human sensory neurons and their potential application for identification of better, effective, and more precise treatment for the individual pain patient.
Collapse
|
48
|
Wadhawan S, Pant S, Golhar R, Kirov S, Thompson J, Jacobsen L, Qureshi I, Ajroud-Driss S, Freeman R, Simpson DM, Smith AG, Hoke A, Bristow LJ. Na V channel variants in patients with painful and nonpainful peripheral neuropathy. NEUROLOGY-GENETICS 2017; 3:e207. [PMID: 29264398 PMCID: PMC5732007 DOI: 10.1212/nxg.0000000000000207] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 10/02/2017] [Indexed: 12/21/2022]
Abstract
Objective: To examine the incidence of nonsynonymous missense variants in SCN9A (NaV1.7), SCN10A (NaV1.8), and SCN11A (NaV1.9) in patients with painful and nonpainful peripheral neuropathy. Methods: Next-generation sequencing was performed on 457 patient DNA samples provided by the Peripheral Neuropathy Research Registry (PNRR). The patient diagnosis was as follows: 278 idiopathic peripheral neuropathy (67% painful and 33% nonpainful) and 179 diabetic distal polyneuropathy (77% painful and 23% nonpainful). Results: We identified 36 (SCN9A), 31 (SCN10A), and 15 (SCN11A) nonsynonymous missense variants, with 47.7% of patients carrying a low-frequency (minor allele frequency <5%) missense variant in at least 1 gene. The incidence of previously reported gain-of-function missense variants was low (≤3%), and these were detected in patients with and without pain. There were no significant differences in missense variant allele frequencies of any gene, or SCN9A haplotype frequencies, between PNRR patients with painful or nonpainful peripheral neuropathy. PNRR patient SCN9A and SCN11A missense variant allele frequencies were not significantly different from the Exome Variant Server, European American (EVS-EA) reference population. For SCN10A, there was a significant increase in the alternate allele frequency of the common variant p.V1073A and low-frequency variant pS509P in PNRR patients compared with EVS-EA and the 1000 Genomes European reference populations. Conclusions: These results suggest that identification of a genetically defined subpopulation for testing of NaV1.7 inhibitors in patients with peripheral neuropathy is unlikely and that additional factors, beyond expression of previously reported disease “mutations,” are more important for the development of painful neuropathy than previously discussed.
Collapse
Affiliation(s)
- Samir Wadhawan
- Department of Translational Biomarkers and Computational Genomics (S.W., S.P., R.G., S.K., J.T.), Bristol-Myers Squibb, Hopewell Site, Pennington, NJ; Department of Genetically Defined Diseases Discovery Biology (L.J.B.), Bristol-Myers Squibb, Wallingford, CT; Department of Innovative Clinical Development (I.Q., L.J.), Bristol-Myers Squibb, Lawrenceville, Princeton, NJ; Department of Neurology (S.A.-D.), Northwestern Medical Faculty Foundation, Chicago, IL; Department of Neurology (R.F.), Beth Israel Medical Center, Harvard School of Medicine, Boston, MA; Department of Neurology (D.M.S.), Icahn School of Medicine at Mount Sinai Medical Center, New York, NY; Department of Neurology (A.G.S.), University of Utah School of Medicine, Salt Lake City, UT; Department of Neurology (A.H.), Johns Hopkins University, Baltimore, MD. S.P. is currently affiliated with Biocon Bristol-Myers Squibb Research Center, Bangalore, India
| | - Saumya Pant
- Department of Translational Biomarkers and Computational Genomics (S.W., S.P., R.G., S.K., J.T.), Bristol-Myers Squibb, Hopewell Site, Pennington, NJ; Department of Genetically Defined Diseases Discovery Biology (L.J.B.), Bristol-Myers Squibb, Wallingford, CT; Department of Innovative Clinical Development (I.Q., L.J.), Bristol-Myers Squibb, Lawrenceville, Princeton, NJ; Department of Neurology (S.A.-D.), Northwestern Medical Faculty Foundation, Chicago, IL; Department of Neurology (R.F.), Beth Israel Medical Center, Harvard School of Medicine, Boston, MA; Department of Neurology (D.M.S.), Icahn School of Medicine at Mount Sinai Medical Center, New York, NY; Department of Neurology (A.G.S.), University of Utah School of Medicine, Salt Lake City, UT; Department of Neurology (A.H.), Johns Hopkins University, Baltimore, MD. S.P. is currently affiliated with Biocon Bristol-Myers Squibb Research Center, Bangalore, India
| | - Ryan Golhar
- Department of Translational Biomarkers and Computational Genomics (S.W., S.P., R.G., S.K., J.T.), Bristol-Myers Squibb, Hopewell Site, Pennington, NJ; Department of Genetically Defined Diseases Discovery Biology (L.J.B.), Bristol-Myers Squibb, Wallingford, CT; Department of Innovative Clinical Development (I.Q., L.J.), Bristol-Myers Squibb, Lawrenceville, Princeton, NJ; Department of Neurology (S.A.-D.), Northwestern Medical Faculty Foundation, Chicago, IL; Department of Neurology (R.F.), Beth Israel Medical Center, Harvard School of Medicine, Boston, MA; Department of Neurology (D.M.S.), Icahn School of Medicine at Mount Sinai Medical Center, New York, NY; Department of Neurology (A.G.S.), University of Utah School of Medicine, Salt Lake City, UT; Department of Neurology (A.H.), Johns Hopkins University, Baltimore, MD. S.P. is currently affiliated with Biocon Bristol-Myers Squibb Research Center, Bangalore, India
| | - Stefan Kirov
- Department of Translational Biomarkers and Computational Genomics (S.W., S.P., R.G., S.K., J.T.), Bristol-Myers Squibb, Hopewell Site, Pennington, NJ; Department of Genetically Defined Diseases Discovery Biology (L.J.B.), Bristol-Myers Squibb, Wallingford, CT; Department of Innovative Clinical Development (I.Q., L.J.), Bristol-Myers Squibb, Lawrenceville, Princeton, NJ; Department of Neurology (S.A.-D.), Northwestern Medical Faculty Foundation, Chicago, IL; Department of Neurology (R.F.), Beth Israel Medical Center, Harvard School of Medicine, Boston, MA; Department of Neurology (D.M.S.), Icahn School of Medicine at Mount Sinai Medical Center, New York, NY; Department of Neurology (A.G.S.), University of Utah School of Medicine, Salt Lake City, UT; Department of Neurology (A.H.), Johns Hopkins University, Baltimore, MD. S.P. is currently affiliated with Biocon Bristol-Myers Squibb Research Center, Bangalore, India
| | - John Thompson
- Department of Translational Biomarkers and Computational Genomics (S.W., S.P., R.G., S.K., J.T.), Bristol-Myers Squibb, Hopewell Site, Pennington, NJ; Department of Genetically Defined Diseases Discovery Biology (L.J.B.), Bristol-Myers Squibb, Wallingford, CT; Department of Innovative Clinical Development (I.Q., L.J.), Bristol-Myers Squibb, Lawrenceville, Princeton, NJ; Department of Neurology (S.A.-D.), Northwestern Medical Faculty Foundation, Chicago, IL; Department of Neurology (R.F.), Beth Israel Medical Center, Harvard School of Medicine, Boston, MA; Department of Neurology (D.M.S.), Icahn School of Medicine at Mount Sinai Medical Center, New York, NY; Department of Neurology (A.G.S.), University of Utah School of Medicine, Salt Lake City, UT; Department of Neurology (A.H.), Johns Hopkins University, Baltimore, MD. S.P. is currently affiliated with Biocon Bristol-Myers Squibb Research Center, Bangalore, India
| | - Leslie Jacobsen
- Department of Translational Biomarkers and Computational Genomics (S.W., S.P., R.G., S.K., J.T.), Bristol-Myers Squibb, Hopewell Site, Pennington, NJ; Department of Genetically Defined Diseases Discovery Biology (L.J.B.), Bristol-Myers Squibb, Wallingford, CT; Department of Innovative Clinical Development (I.Q., L.J.), Bristol-Myers Squibb, Lawrenceville, Princeton, NJ; Department of Neurology (S.A.-D.), Northwestern Medical Faculty Foundation, Chicago, IL; Department of Neurology (R.F.), Beth Israel Medical Center, Harvard School of Medicine, Boston, MA; Department of Neurology (D.M.S.), Icahn School of Medicine at Mount Sinai Medical Center, New York, NY; Department of Neurology (A.G.S.), University of Utah School of Medicine, Salt Lake City, UT; Department of Neurology (A.H.), Johns Hopkins University, Baltimore, MD. S.P. is currently affiliated with Biocon Bristol-Myers Squibb Research Center, Bangalore, India
| | - Irfan Qureshi
- Department of Translational Biomarkers and Computational Genomics (S.W., S.P., R.G., S.K., J.T.), Bristol-Myers Squibb, Hopewell Site, Pennington, NJ; Department of Genetically Defined Diseases Discovery Biology (L.J.B.), Bristol-Myers Squibb, Wallingford, CT; Department of Innovative Clinical Development (I.Q., L.J.), Bristol-Myers Squibb, Lawrenceville, Princeton, NJ; Department of Neurology (S.A.-D.), Northwestern Medical Faculty Foundation, Chicago, IL; Department of Neurology (R.F.), Beth Israel Medical Center, Harvard School of Medicine, Boston, MA; Department of Neurology (D.M.S.), Icahn School of Medicine at Mount Sinai Medical Center, New York, NY; Department of Neurology (A.G.S.), University of Utah School of Medicine, Salt Lake City, UT; Department of Neurology (A.H.), Johns Hopkins University, Baltimore, MD. S.P. is currently affiliated with Biocon Bristol-Myers Squibb Research Center, Bangalore, India
| | - Senda Ajroud-Driss
- Department of Translational Biomarkers and Computational Genomics (S.W., S.P., R.G., S.K., J.T.), Bristol-Myers Squibb, Hopewell Site, Pennington, NJ; Department of Genetically Defined Diseases Discovery Biology (L.J.B.), Bristol-Myers Squibb, Wallingford, CT; Department of Innovative Clinical Development (I.Q., L.J.), Bristol-Myers Squibb, Lawrenceville, Princeton, NJ; Department of Neurology (S.A.-D.), Northwestern Medical Faculty Foundation, Chicago, IL; Department of Neurology (R.F.), Beth Israel Medical Center, Harvard School of Medicine, Boston, MA; Department of Neurology (D.M.S.), Icahn School of Medicine at Mount Sinai Medical Center, New York, NY; Department of Neurology (A.G.S.), University of Utah School of Medicine, Salt Lake City, UT; Department of Neurology (A.H.), Johns Hopkins University, Baltimore, MD. S.P. is currently affiliated with Biocon Bristol-Myers Squibb Research Center, Bangalore, India
| | - Roy Freeman
- Department of Translational Biomarkers and Computational Genomics (S.W., S.P., R.G., S.K., J.T.), Bristol-Myers Squibb, Hopewell Site, Pennington, NJ; Department of Genetically Defined Diseases Discovery Biology (L.J.B.), Bristol-Myers Squibb, Wallingford, CT; Department of Innovative Clinical Development (I.Q., L.J.), Bristol-Myers Squibb, Lawrenceville, Princeton, NJ; Department of Neurology (S.A.-D.), Northwestern Medical Faculty Foundation, Chicago, IL; Department of Neurology (R.F.), Beth Israel Medical Center, Harvard School of Medicine, Boston, MA; Department of Neurology (D.M.S.), Icahn School of Medicine at Mount Sinai Medical Center, New York, NY; Department of Neurology (A.G.S.), University of Utah School of Medicine, Salt Lake City, UT; Department of Neurology (A.H.), Johns Hopkins University, Baltimore, MD. S.P. is currently affiliated with Biocon Bristol-Myers Squibb Research Center, Bangalore, India
| | - David M Simpson
- Department of Translational Biomarkers and Computational Genomics (S.W., S.P., R.G., S.K., J.T.), Bristol-Myers Squibb, Hopewell Site, Pennington, NJ; Department of Genetically Defined Diseases Discovery Biology (L.J.B.), Bristol-Myers Squibb, Wallingford, CT; Department of Innovative Clinical Development (I.Q., L.J.), Bristol-Myers Squibb, Lawrenceville, Princeton, NJ; Department of Neurology (S.A.-D.), Northwestern Medical Faculty Foundation, Chicago, IL; Department of Neurology (R.F.), Beth Israel Medical Center, Harvard School of Medicine, Boston, MA; Department of Neurology (D.M.S.), Icahn School of Medicine at Mount Sinai Medical Center, New York, NY; Department of Neurology (A.G.S.), University of Utah School of Medicine, Salt Lake City, UT; Department of Neurology (A.H.), Johns Hopkins University, Baltimore, MD. S.P. is currently affiliated with Biocon Bristol-Myers Squibb Research Center, Bangalore, India
| | - A Gordon Smith
- Department of Translational Biomarkers and Computational Genomics (S.W., S.P., R.G., S.K., J.T.), Bristol-Myers Squibb, Hopewell Site, Pennington, NJ; Department of Genetically Defined Diseases Discovery Biology (L.J.B.), Bristol-Myers Squibb, Wallingford, CT; Department of Innovative Clinical Development (I.Q., L.J.), Bristol-Myers Squibb, Lawrenceville, Princeton, NJ; Department of Neurology (S.A.-D.), Northwestern Medical Faculty Foundation, Chicago, IL; Department of Neurology (R.F.), Beth Israel Medical Center, Harvard School of Medicine, Boston, MA; Department of Neurology (D.M.S.), Icahn School of Medicine at Mount Sinai Medical Center, New York, NY; Department of Neurology (A.G.S.), University of Utah School of Medicine, Salt Lake City, UT; Department of Neurology (A.H.), Johns Hopkins University, Baltimore, MD. S.P. is currently affiliated with Biocon Bristol-Myers Squibb Research Center, Bangalore, India
| | - Ahmet Hoke
- Department of Translational Biomarkers and Computational Genomics (S.W., S.P., R.G., S.K., J.T.), Bristol-Myers Squibb, Hopewell Site, Pennington, NJ; Department of Genetically Defined Diseases Discovery Biology (L.J.B.), Bristol-Myers Squibb, Wallingford, CT; Department of Innovative Clinical Development (I.Q., L.J.), Bristol-Myers Squibb, Lawrenceville, Princeton, NJ; Department of Neurology (S.A.-D.), Northwestern Medical Faculty Foundation, Chicago, IL; Department of Neurology (R.F.), Beth Israel Medical Center, Harvard School of Medicine, Boston, MA; Department of Neurology (D.M.S.), Icahn School of Medicine at Mount Sinai Medical Center, New York, NY; Department of Neurology (A.G.S.), University of Utah School of Medicine, Salt Lake City, UT; Department of Neurology (A.H.), Johns Hopkins University, Baltimore, MD. S.P. is currently affiliated with Biocon Bristol-Myers Squibb Research Center, Bangalore, India
| | - Linda J Bristow
- Department of Translational Biomarkers and Computational Genomics (S.W., S.P., R.G., S.K., J.T.), Bristol-Myers Squibb, Hopewell Site, Pennington, NJ; Department of Genetically Defined Diseases Discovery Biology (L.J.B.), Bristol-Myers Squibb, Wallingford, CT; Department of Innovative Clinical Development (I.Q., L.J.), Bristol-Myers Squibb, Lawrenceville, Princeton, NJ; Department of Neurology (S.A.-D.), Northwestern Medical Faculty Foundation, Chicago, IL; Department of Neurology (R.F.), Beth Israel Medical Center, Harvard School of Medicine, Boston, MA; Department of Neurology (D.M.S.), Icahn School of Medicine at Mount Sinai Medical Center, New York, NY; Department of Neurology (A.G.S.), University of Utah School of Medicine, Salt Lake City, UT; Department of Neurology (A.H.), Johns Hopkins University, Baltimore, MD. S.P. is currently affiliated with Biocon Bristol-Myers Squibb Research Center, Bangalore, India
| |
Collapse
|
49
|
Terkelsen AJ, Karlsson P, Lauria G, Freeman R, Finnerup NB, Jensen TS. The diagnostic challenge of small fibre neuropathy: clinical presentations, evaluations, and causes. Lancet Neurol 2017; 16:934-944. [DOI: 10.1016/s1474-4422(17)30329-0] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 07/31/2017] [Accepted: 08/09/2017] [Indexed: 12/15/2022]
|
50
|
Knezevic NN, Yekkirala A, Yaksh TL. Basic/Translational Development of Forthcoming Opioid- and Nonopioid-Targeted Pain Therapeutics. Anesth Analg 2017; 125:1714-1732. [PMID: 29049116 PMCID: PMC5679134 DOI: 10.1213/ane.0000000000002442] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Opioids represent an efficacious therapeutic modality for some, but not all pain states. Singular reliance on opioid therapy for pain management has limitations, and abuse potential has deleterious consequences for patient and society. Our understanding of pain biology has yielded insights and opportunities for alternatives to conventional opioid agonists. The aim is to have efficacious therapies, with acceptable side effect profiles and minimal abuse potential, which is to say an absence of reinforcing activity in the absence of a pain state. The present work provides a nonexclusive overview of current drug targets and potential future directions of research and development. We discuss channel activators and blockers, including sodium channel blockers, potassium channel activators, and calcium channel blockers; glutamate receptor-targeted agents, including N-methyl-D-aspartate, α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid, and metabotropic receptors. Furthermore, we discuss therapeutics targeted at γ-aminobutyric acid, α2-adrenergic, and opioid receptors. We also considered antagonists of angiotensin 2 and Toll receptors and agonists/antagonists of adenosine, purine receptors, and cannabinoids. Novel targets considered are those focusing on lipid mediators and anti-inflammatory cytokines. Of interest is development of novel targeting strategies, which produce long-term alterations in pain signaling, including viral transfection and toxins. We consider issues in the development of druggable molecules, including preclinical screening. While there are examples of successful translation, mechanistically promising preclinical candidates may unexpectedly fail during clinical trials because the preclinical models may not recapitulate the particular human pain condition being addressed. Molecular target characterization can diminish the disconnect between preclinical and humans' targets, which should assist in developing nonaddictive analgesics.
Collapse
Affiliation(s)
- Nebojsa Nick Knezevic
- From the *Department of Anesthesiology, Advocate Illinois Masonic Medical Center Chicago, Illinois; Departments of †Anesthesiology and ‡Surgery, University of Illinois, Chicago, Illinois; §Department of Neurobiology, Harvard Medical School, and Boston Children's Hospital, Boston, Massachusetts; ‖Blue Therapeutics, Harvard Innovation Launch Lab, Allston, Massachusetts; and Departments of ¶Anesthesiology and #Pharmacology, University of California, San Diego, La Jolla, California
| | | | | |
Collapse
|