51
|
Bennet BM, Pardo ID, Assaf BT, Buza E, Cramer SD, Crawford LK, Engelhardt JA, Galbreath EJ, Grubor B, Morrison JP, Osborne TS, Sharma AK, Bolon B. Scientific and Regulatory Policy Committee Technical Review: Biology and Pathology of Ganglia in Animal Species Used for Nonclinical Safety Testing. Toxicol Pathol 2023; 51:278-305. [PMID: 38047294 DOI: 10.1177/01926233231213851] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Dorsal root ganglia (DRG), trigeminal ganglia (TG), other sensory ganglia, and autonomic ganglia may be injured by some test article classes, including anti-neoplastic chemotherapeutics, adeno-associated virus-based gene therapies, antisense oligonucleotides, nerve growth factor inhibitors, and aminoglycoside antibiotics. This article reviews ganglion anatomy, cytology, and pathology (emphasizing sensory ganglia) among common nonclinical species used in assessing product safety for such test articles (TAs). Principal histopathologic findings associated with sensory ganglion injury include neuron degeneration, necrosis, and/or loss; increased satellite glial cell and/or Schwann cell numbers; and leukocyte infiltration and/or inflammation. Secondary nerve fiber degeneration and/or glial reactions may occur in nerves, dorsal spinal nerve roots, spinal cord (dorsal and occasionally lateral funiculi), and sometimes the brainstem. Ganglion findings related to TA administration may result from TA exposure and/or trauma related to direct TA delivery into the central nervous system or ganglia. In some cases, TA-related effects may need to be differentiated from a spectrum of artifactual and/or spontaneous background changes.
Collapse
Affiliation(s)
| | | | | | - Elizabeth Buza
- University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | | | | | | | | | - James P Morrison
- Charles River Laboratories, Inc., Shrewsbury, Massachusetts, USA
| | | | | | | |
Collapse
|
52
|
Ortega MA, Fraile-Martinez O, García-Montero C, Haro S, Álvarez-Mon MÁ, De Leon-Oliva D, Gomez-Lahoz AM, Monserrat J, Atienza-Pérez M, Díaz D, Lopez-Dolado E, Álvarez-Mon M. A comprehensive look at the psychoneuroimmunoendocrinology of spinal cord injury and its progression: mechanisms and clinical opportunities. Mil Med Res 2023; 10:26. [PMID: 37291666 PMCID: PMC10251601 DOI: 10.1186/s40779-023-00461-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 06/01/2023] [Indexed: 06/10/2023] Open
Abstract
Spinal cord injury (SCI) is a devastating and disabling medical condition generally caused by a traumatic event (primary injury). This initial trauma is accompanied by a set of biological mechanisms directed to ameliorate neural damage but also exacerbate initial damage (secondary injury). The alterations that occur in the spinal cord have not only local but also systemic consequences and virtually all organs and tissues of the body incur important changes after SCI, explaining the progression and detrimental consequences related to this condition. Psychoneuroimmunoendocrinology (PNIE) is a growing area of research aiming to integrate and explore the interactions among the different systems that compose the human organism, considering the mind and the body as a whole. The initial traumatic event and the consequent neurological disruption trigger immune, endocrine, and multisystem dysfunction, which in turn affect the patient's psyche and well-being. In the present review, we will explore the most important local and systemic consequences of SCI from a PNIE perspective, defining the changes occurring in each system and how all these mechanisms are interconnected. Finally, potential clinical approaches derived from this knowledge will also be collectively presented with the aim to develop integrative therapies to maximize the clinical management of these patients.
Collapse
Affiliation(s)
- Miguel A. Ortega
- Department of Medicine and Medical Specialities, University of Alcala, 28801 Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Oscar Fraile-Martinez
- Department of Medicine and Medical Specialities, University of Alcala, 28801 Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Cielo García-Montero
- Department of Medicine and Medical Specialities, University of Alcala, 28801 Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Sergio Haro
- Department of Medicine and Medical Specialities, University of Alcala, 28801 Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Miguel Ángel Álvarez-Mon
- Department of Medicine and Medical Specialities, University of Alcala, 28801 Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
- Department of Psychiatry and Mental Health, Hospital Universitario Infanta Leonor, 28031 Madrid, Spain
| | - Diego De Leon-Oliva
- Department of Medicine and Medical Specialities, University of Alcala, 28801 Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Ana M. Gomez-Lahoz
- Department of Medicine and Medical Specialities, University of Alcala, 28801 Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Jorge Monserrat
- Department of Medicine and Medical Specialities, University of Alcala, 28801 Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Mar Atienza-Pérez
- Service of Rehabilitation, National Hospital for Paraplegic Patients, Carr. de la Peraleda, S/N, 45004 Toledo, Spain
| | - David Díaz
- Department of Medicine and Medical Specialities, University of Alcala, 28801 Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Elisa Lopez-Dolado
- Department of Medicine and Medical Specialities, University of Alcala, 28801 Alcala de Henares, Spain
- Department of Psychiatry and Mental Health, Hospital Universitario Infanta Leonor, 28031 Madrid, Spain
| | - Melchor Álvarez-Mon
- Department of Medicine and Medical Specialities, University of Alcala, 28801 Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
- Immune System Diseases-Rheumatology Service and Internal Medicine, University Hospital Príncipe de Asturias (CIBEREHD), 28806 Alcala de Henares, Spain
| |
Collapse
|
53
|
Bennet BM, Pardo ID, Assaf BT, Buza E, Cramer S, Crawford LK, Engelhardt JA, Grubor B, Morrison JP, Osborne TS, Sharma AK, Bolon B. Scientific and Regulatory Policy Committee Points to Consider: Sampling, Processing, Evaluation, Interpretation, and Reporting of Test Article-Related Ganglion Pathology for Nonclinical Toxicity Studies. Toxicol Pathol 2023; 51:176-204. [PMID: 37489508 DOI: 10.1177/01926233231179707] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/26/2023]
Abstract
Certain biopharmaceutical products consistently affect dorsal root ganglia, trigeminal ganglia, and/or autonomic ganglia. Product classes targeting ganglia include antineoplastic chemotherapeutics, adeno-associated virus-based gene therapies, antisense oligonucleotides, and anti-nerve growth factor agents. This article outlines "points to consider" for sample collection, processing, evaluation, interpretation, and reporting of ganglion findings; these points are consistent with published best practices for peripheral nervous system evaluation in nonclinical toxicity studies. Ganglion findings often occur as a combination of neuronal injury (e.g., degeneration, necrosis, and/or loss) and/or glial effects (e.g., increased satellite glial cell cellularity) with leukocyte accumulation (e.g., mononuclear cell infiltration or inflammation). Nerve fiber degeneration and/or glial reactions may be seen in nerves, dorsal spinal nerve roots, spinal cord, and occasionally brainstem. Interpretation of test article (TA)-associated effects may be confounded by incidental background changes or experimental procedure-related changes and limited historical control data. Reports should describe findings at these sites, any TA relationship, and the criteria used for assigning severity grades. Contextualizing adversity of ganglia findings can require a weight-of-evidence approach because morphologic changes of variable severity occur in ganglia but often are not accompanied by observable overt in-life functional alterations detectable by conventional behavioral and neurological testing techniques.
Collapse
Affiliation(s)
| | | | | | - Elizabeth Buza
- University of Pennsylvania, Gene Therapy Program, Philadelphia, Pennsylvania, USA
| | | | - LaTasha K Crawford
- University of Wisconsin-Madison, School of Veterinary Medicine, Madison, Wisconsin, USA
| | | | | | - James P Morrison
- Charles River Laboratories, Inc., Shrewsbury, Massachusetts, USA
| | | | | | | |
Collapse
|
54
|
Tiwari MN, Hall BE, Terse A, Amin N, Chung MK, Kulkarni AB. ACTIVATION OF CYCLIN-DEPENDENT KINASE 5 BROADENS ACTION POTENTIALS IN HUMAN SENSORY NEURONS. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.31.543017. [PMID: 37398398 PMCID: PMC10312556 DOI: 10.1101/2023.05.31.543017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Chronic pain is one of the most devastating and unpleasant conditions, associated with many pathological conditions. Tissue or nerve injuries induce comprehensive neurobiological plasticity in nociceptive neurons, which leads to chronic pain. Recent studies suggest that cyclin-dependent kinase 5 (CDK5) in primary afferents is a key neuronal kinase that modulates nociception through phosphorylation-dependent manner under pathological conditions. However, the impact of the CDK5 on nociceptor activity especially in human sensory neurons are not known. To determine the CDK5-mediated regulation of human dorsal root ganglia (hDRG) neuronal properties, we have performed the whole-cell patch clamp recordings in neurons dissociated from hDRG. CDK5 activation induced by overexpression of p35 depolarized the resting membrane potential and reduced the rheobase currents as compared to the uninfected neurons. CDK5 activation evidently changed the shape of the action potential (AP) by increasing AP rise time, AP fall time, and AP half width. The application of a prostaglandin E2 (PG) and bradykinin (BK) cocktail in uninfected hDRG neurons induced the depolarization of RMP and the reduction of rheobase currents along with increased AP rise time. However, PG and BK applications failed to induce any further significant changes in addition to the aforementioned changes of the membrane properties and AP parameters in the p35-overexpressing group. We conclude that CDK5 activation through the overexpression of p35 in dissociated hDRG neurons broadens AP in hDRG neurons and that CDK5 may play important roles in the modulation of AP properties in human primary afferents under pathological conditions, contributing to chronic pain.
Collapse
Affiliation(s)
- Manindra Nath Tiwari
- Department of Neural and Pain Sciences, School of Dentistry, Program in Neuroscience, Center to Advance Chronic Pain Research, The University of Maryland, Baltimore, Maryland 21201
| | - Bradford E. Hall
- Functional Genomics Section, National Institute of Dental and Craniofacial Research
| | - Anita Terse
- Functional Genomics Section, National Institute of Dental and Craniofacial Research
| | - Niranjana Amin
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Man-Kyo Chung
- Department of Neural and Pain Sciences, School of Dentistry, Program in Neuroscience, Center to Advance Chronic Pain Research, The University of Maryland, Baltimore, Maryland 21201
| | - Ashok B. Kulkarni
- Functional Genomics Section, National Institute of Dental and Craniofacial Research
| |
Collapse
|
55
|
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
|
56
|
Wang K, Cai B, Song Y, Chen Y, Zhang X. Somatosensory neuron types and their neural networks as revealed via single-cell transcriptomics. Trends Neurosci 2023:S0166-2236(23)00130-3. [PMID: 37268541 DOI: 10.1016/j.tins.2023.05.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 04/24/2023] [Accepted: 05/06/2023] [Indexed: 06/04/2023]
Abstract
Single-cell RNA sequencing (scRNA-seq) has allowed profiling cell types of the dorsal root ganglia (DRG) and their transcriptional states in physiology and chronic pain. However, the evaluation criteria used in previous studies to classify DRG neurons varied, which presents difficulties in determining the various types of DRG neurons. In this review, we aim to integrate findings from previous transcriptomic studies of the DRG. We first briefly introduce the history of DRG-neuron cell-type profiling, and discuss the advantages and disadvantages of different scRNA-seq methods. We then examine the classification of DRG neurons based on single-cell profiling under physiological and pathological conditions. Finally, we propose further studies on the somatosensory system at the molecular, cellular, and neural network levels.
Collapse
Affiliation(s)
- Kaikai Wang
- Guangdong Institute of Intelligence Science and Technology, Hengqin 519031, Zhuhai, Guangdong, China; Research Unit of Pain Medicine, Chinese Academy of Medical Sciences, Hengqin, Zhuhai, China
| | - Bing Cai
- Guangdong Institute of Intelligence Science and Technology, Hengqin 519031, Zhuhai, Guangdong, China; Research Unit of Pain Medicine, Chinese Academy of Medical Sciences, Hengqin, Zhuhai, China
| | - Yurang Song
- Guangdong Institute of Intelligence Science and Technology, Hengqin 519031, Zhuhai, Guangdong, China; Research Unit of Pain Medicine, Chinese Academy of Medical Sciences, Hengqin, Zhuhai, China
| | - Yan Chen
- Guangdong Institute of Intelligence Science and Technology, Hengqin 519031, Zhuhai, Guangdong, China; Research Unit of Pain Medicine, Chinese Academy of Medical Sciences, Hengqin, Zhuhai, China; Xuhui Central Hospital, Shanghai, 200031, China
| | - Xu Zhang
- Guangdong Institute of Intelligence Science and Technology, Hengqin 519031, Zhuhai, Guangdong, China; SIMR Joint Lab of Drug Innovation, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, 201210, China; Research Unit of Pain Medicine, Chinese Academy of Medical Sciences, Hengqin, Zhuhai, China; Xuhui Central Hospital, Shanghai, 200031, China.
| |
Collapse
|
57
|
Belo TCA, Santos GX, da Silva BEG, Rocha BLG, Abdala DW, Freire LAM, Rocha FS, Galdino G. IL-10/β-Endorphin-Mediated Neuroimmune Modulation on Microglia during Antinociception. Brain Sci 2023; 13:brainsci13050789. [PMID: 37239261 DOI: 10.3390/brainsci13050789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/09/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
Microglia are glial cells centrally related to pathophysiology and neuroimmunological regulation of pain through microglia-neuron crosstalk mechanisms. In contrast, anti-inflammatory mechanisms guided by immunological effectors such as IL-10 trigger the secretion of analgesic substances, culminating in the differential expression of genes encoding endogenous opioid peptides, especially β-endorphin. Thus, when β-endorphin binds to the µ-opioid receptor, it generates neuronal hyperpolarization, inhibiting nociceptive stimuli. This review aimed to summarize the recent advances in understanding the mechanism by which IL-10/β-endorphin can reduce pain. For this, databases were searched for articles from their inception up until November 2022. Two independent reviewers extracted the data and assessed the methodological quality of the included studies, and seventeen studies were considered eligible for this review. Several studies have demonstrated the impact of IL-10/β-endorphin in reducing pain, where IL-10 can stimulate GLP-1R, GRP40, and α7nAChR receptors, as well as intracellular signaling pathways, such as STAT3, resulting in increased β-endorphin expression and secretion. In addition, molecules such as gabapentinoids, thalidomide, cynandione A, morroniside, lemairamin, and cinobufagin, as well as non-pharmacological treatments such as electroacupuncture, reduce pain through IL-10 mediated mechanisms, reflecting a microglia-dependent β-endorphin differential increase. This process represents a cornerstone in pain neuroimmunology knowledge, and the results obtained by different studies about the theme are presented in this review.
Collapse
Affiliation(s)
| | - Gabriela Xavier Santos
- Laboratory of Neuroimmunobiology of Pain, Federal University of Alfenas, Alfenas 37130-001, Brazil
| | | | | | - Dennis William Abdala
- Laboratory of Movement Analysis, Federal University of Alfenas, Alfenas 37130-001, Brazil
| | - Larissa Alves Moreira Freire
- Laboratory of Neuroscience, Neuroimmunomodulation and Pain Study, Federal University of Alfenas, Alfenas 37130-001, Brazil
| | - Fernanda Santos Rocha
- Laboratory of Neuroimmunobiology of Pain, Federal University of Alfenas, Alfenas 37130-001, Brazil
| | - Giovane Galdino
- Laboratory of Neuroimmunobiology of Pain, Federal University of Alfenas, Alfenas 37130-001, Brazil
| |
Collapse
|
58
|
Haberberger RV, Kuramatilake J, Barry CM, Matusica D. Ultrastructure of dorsal root ganglia. Cell Tissue Res 2023:10.1007/s00441-023-03770-w. [PMID: 37079097 PMCID: PMC10115609 DOI: 10.1007/s00441-023-03770-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 03/28/2023] [Indexed: 04/21/2023]
Abstract
Dorsal root ganglia (DRG) contains thousands of sensory neurons that transmit information about our external and internal environment to the central nervous system. This includes signals related to proprioception, temperature, and nociception. Our understanding of DRG has increased tremendously over the last 50 years and has established the DRG as an active participant in peripheral processes. This includes interactions between neurons and non-neuronal cells such as satellite glia cells and macrophages that contribute to an increasingly complex cellular environment that modulates neuronal function. Early ultrastructural investigations of the DRG have described subtypes of sensory neurons based on differences in the arrangement of organelles such as the Golgi apparatus and the endoplasmic reticulum. The neuron-satellite cell complex and the composition of the axon hillock in DRG have also been investigated, but, apart from basic descriptions of Schwann cells, ultrastructural investigations of other cell types in DRG are limited. Furthermore, detailed descriptions of key components of DRG, such as blood vessels and the capsule that sits at the intersection of the meninges and the connective tissue covering the peripheral nervous system, are lacking to date. With rising interest in DRG as potential therapeutic targets for aberrant signalling associated with chronic pain conditions, gaining further insights into DRG ultrastructure will be fundamental to understanding cell-cell interactions that modulate DRG function. In this review, we aim to provide a synopsis of the current state of knowledge on the ultrastructure of the DRG and its components, as well as to identify areas of interest for future studies.
Collapse
Affiliation(s)
- Rainer Viktor Haberberger
- Division of Anatomy and Pathology, School of Biomedicine, The University of Adelaide, Adelaide, Australia.
| | - Jaliya Kuramatilake
- Division of Anatomy and Pathology, School of Biomedicine, The University of Adelaide, Adelaide, Australia
| | - Christine M Barry
- Anatomy, Histology & Pathology, College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Dusan Matusica
- Anatomy, Histology & Pathology, College of Medicine and Public Health, Flinders University, Adelaide, Australia
| |
Collapse
|
59
|
Schulte A, Lohner H, Degenbeck J, Segebarth D, Rittner HL, Blum R, Aue A. Unbiased analysis of the dorsal root ganglion after peripheral nerve injury: no neuronal loss, no gliosis, but satellite glial cell plasticity. Pain 2023; 164:728-740. [PMID: 35969236 PMCID: PMC10026836 DOI: 10.1097/j.pain.0000000000002758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 07/13/2022] [Accepted: 07/26/2022] [Indexed: 11/26/2022]
Abstract
ABSTRACT Pain syndromes are often accompanied by complex molecular and cellular changes in dorsal root ganglia (DRG). However, the evaluation of cellular plasticity in the DRG is often performed by heuristic manual analysis of a small number of representative microscopy image fields. In this study, we introduce a deep learning-based strategy for objective and unbiased analysis of neurons and satellite glial cells (SGCs) in the DRG. To validate the approach experimentally, we examined serial sections of the rat DRG after spared nerve injury (SNI) or sham surgery. Sections were stained for neurofilament, glial fibrillary acidic protein (GFAP), and glutamine synthetase (GS) and imaged using high-resolution large-field (tile) microscopy. After training of deep learning models on consensus information of different experts, thousands of image features in DRG sections were analyzed. We used known (GFAP upregulation), controversial (neuronal loss), and novel (SGC phenotype switch) changes to evaluate the method. In our data, the number of DRG neurons was similar 14 d after SNI vs sham. In GFAP-positive subareas, the percentage of neurons in proximity to GFAP-positive cells increased after SNI. In contrast, GS-positive signals, and the percentage of neurons in proximity to GS-positive SGCs decreased after SNI. Changes in GS and GFAP levels could be linked to specific DRG neuron subgroups of different size. Hence, we could not detect gliosis but plasticity changes in the SGC marker expression. Our objective analysis of DRG tissue after peripheral nerve injury shows cellular plasticity responses of SGCs in the whole DRG but neither injury-induced neuronal death nor gliosis.
Collapse
Affiliation(s)
- Annemarie Schulte
- Department of Neurology, University Hospital of Würzburg, Würzburg, Germany
| | - Hannah Lohner
- Department of Anesthesiology, Center for Interdisciplinary Pain Medicine, Intensive Care, Emergency Medicine and Pain Therapy, University Hospital of Würzburg, Würzburg, Germany
| | - Johannes Degenbeck
- Department of Anesthesiology, Center for Interdisciplinary Pain Medicine, Intensive Care, Emergency Medicine and Pain Therapy, University Hospital of Würzburg, Würzburg, Germany
| | - Dennis Segebarth
- Institute of Clinical Neurobiology, University Hospital of Würzburg, Würzburg, Germany
| | - Heike L. Rittner
- Department of Anesthesiology, Center for Interdisciplinary Pain Medicine, Intensive Care, Emergency Medicine and Pain Therapy, University Hospital of Würzburg, Würzburg, Germany
| | - Robert Blum
- Department of Neurology, University Hospital of Würzburg, Würzburg, Germany
| | - Annemarie Aue
- Department of Anesthesiology, Center for Interdisciplinary Pain Medicine, Intensive Care, Emergency Medicine and Pain Therapy, University Hospital of Würzburg, Würzburg, Germany
| |
Collapse
|
60
|
Greer S, Cramberg M, Young BA. Morphology of the distal tip of the spinal cord in Alligator mississippiensis. Anat Rec (Hoboken) 2023; 306:889-904. [PMID: 35684989 DOI: 10.1002/ar.25016] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/06/2022] [Accepted: 05/16/2022] [Indexed: 11/11/2022]
Abstract
Secondary neurulation is a common feature of vertebrate development, which in non-mammalian and non-anuran vertebrates, results in the formation of a caudal spinal cord. The present study was undertaken to describe the terminal end of the caudal spinal cord in a crocodylian, a group chosen for their unique status of a living-tailed archosaur. The caudal spinal cord of Alligator mississippiensis terminates near the intervertebral joint between the fourth and fifth terminal vertebrae. Prior to this termination, the dorsal root ganglia get proportionately larger, then stop before the termination of the spinal cord; and the gray matter of the spinal cord is lost producing an unusual morphology in which an ependymal-lined central canal is surrounded by only white matter which is not divided into a cauda equina. The inner layer of the meninges (the pia-arachnoid) courses over the distal end of the spinal cord and forms a ventral attachment, reminiscent of a very short Filum terminale; there is no caudal cistern. The dura extends beyond the termination of the spinal cord, continuing for at least the length of the fourth terminal vertebra, forming a structure herein termed the distal meningeal sheath. During its course, the distal meningeal sheath surrounds a mass of mesothelial cells, then terminates as an attachment on the dorsal surface of the vertebra.
Collapse
Affiliation(s)
- Skye Greer
- Department of Anatomy, Kirksville College of Osteopathic Medicine, Kirksville, Missouri
| | - Michael Cramberg
- Department of Anatomy, Kirksville College of Osteopathic Medicine, Kirksville, Missouri
| | - Bruce A Young
- Department of Anatomy, Kirksville College of Osteopathic Medicine, Kirksville, Missouri
| |
Collapse
|
61
|
Abdelhamid MS, El Bohi KM, Sherif MH, Abdelhamid MS, Abdel-Daim MM, Elewa YHA, Metwally MMM, Albadrani GM, Najda A, El Abdel-Hamid S, Abu-Zeid EH. Apitoxin alleviates methyl mercury-induced peripheral neurotoxicity in male rats by regulating dorsal root ganglia neuronal degeneration and oxidative stress. Biomed Pharmacother 2023; 161:114521. [PMID: 36921536 DOI: 10.1016/j.biopha.2023.114521] [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: 01/06/2023] [Revised: 03/02/2023] [Accepted: 03/09/2023] [Indexed: 03/16/2023] Open
Abstract
Methylmercury (MeHg) toxicity is associated with extensive neuronal degeneration of dorsal root ganglia (DRG). This study aimed to assess the ameliorative effect of bee venom (BV) on methyl mercury chloride (MeHgCl)-induced peripheral neurotoxicity using DRGs in rats. Forty-eight adult male Sprague Dawley rats were allocated into four equal groups: G I: control (gavaged MilliQ water 1 ml/rat), G II: subcutaneously injected with BV (0.5 mg/kg b.wt), G III: gavaged MeHgCl (6.7 mg/kg b.wt), and G IV: received MeHgCl+BV. Dosing was done five times/week for 2 weeks. Ataxic behavior and visual impairments were significantly increased, whereas the movement behavior and motility gait were suppressed in the MeHgCl group. MeHgCl significantly decreased total antioxidant capacity (TAC) in DRG and significantly decreased the serum levels of glutathione (GSH), catalase (CAT), and superoxide dismutase (SOD). Tumor necrosis factor-alpha (TNF-α) and interleukin 1β (IL-1β) levels were significantly elevated, whereas interleukin 10 (IL-10) levels were significantly decreased in the MeHgCl group compared with the control group. DRGs of the MeHgCl-exposed rats showed pyknotic shrunken neurons with perineural vacuolations, demyelination of nerve axons, and proliferation of the satellite cells. MeHgCl significantly induced a higher positive index ratio of Iba-1, SOX10, neurofilament, pan-neuron, and vimentin immunostaining in the DRG. BV administration significantly mitigated the MeHgCl-induced alterations in oxidative stress-related indices. BV modified the immunostaining of Iba-1, SOX10, neurofilament, pan-neuron, and vimentin-positive index ratio in the DRG of the MeHgCl group. Our findings acknowledged that BV could enhance in vivo neuroprotective effects against MeHgCl-induced DRGs damage in male rats.
Collapse
Affiliation(s)
- Moustafa S Abdelhamid
- Biochemistry division, Chemistry Department, Faculty of Science, Zagazig University, 44511, Egypt
| | - Khlood M El Bohi
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Zagazig University, 44511, Egypt
| | - Mohamed H Sherif
- Biochemistry division, Chemistry Department, Faculty of Science, Zagazig University, 44511, Egypt
| | - Manar S Abdelhamid
- Biochemistry division, Chemistry Department, Faculty of Science, Zagazig University, 44511, Egypt
| | - Mohamed M Abdel-Daim
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia; Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Yaser H A Elewa
- Department of Histology and Cytology, Faculty of Veterinary Medicine, Zagazig University, 44511, Egypt; Laboratory of Anatomy, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Mohamed M M Metwally
- Department of Pathology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
| | - Ghadeer M Albadrani
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, B.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Agnieszka Najda
- Department of Vegetable Crops and Medicinal Plants University of Life Sciences in Lublin, 50 A Doświadczalna Street, 20-280 Lublin, Poland.
| | - Shereen El Abdel-Hamid
- Department of Behavior and Management of Animal, Poultry and Aquatics, Faculty of Veterinary Medicine, Zagazig University, 44511, Egypt
| | - Ehsan H Abu-Zeid
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Zagazig University, 44511, Egypt.
| |
Collapse
|
62
|
Talagas M. Anatomical contacts between sensory neurons and epidermal cells: an unrecognized anatomical network for neuro-immuno-cutaneous crosstalk. Br J Dermatol 2023; 188:176-185. [PMID: 36763869 DOI: 10.1093/bjd/ljac066] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 10/11/2022] [Accepted: 10/22/2022] [Indexed: 01/09/2023]
Abstract
Sensory neurons innervating the skin are conventionally thought to be the sole transducers of touch, temperature, pain and itch. However, recent studies have shown that keratinocytes - like Merkel cells - act as sensory transducers, whether for innocuous or noxious mechanical, thermal or chemical stimuli, and communicate with intraepidermal free nerve endings via chemical synaptic contacts. This paradigm shift leads to consideration of the whole epidermis as a sensory epithelium. Sensory neurons additionally function as an efferent system. Through the release of neuropeptides in intimate neuroepidermal contact areas, they contribute to epidermal homeostasis and to the pathogenesis of inflammatory skin diseases. To counteract the dogma regarding neurocutaneous interactions, seen exclusively from the perspective of soluble and spreading mediators, this review highlights the essential contribution of the unrecognized anatomical contacts between sensory neurons and epidermal cells (keratinocytes, melanocytes, Langerhans cells and Merkel cells), which take part in the reciprocal dialogue between the skin, nervous system and immune system.
Collapse
Affiliation(s)
- Matthieu Talagas
- University of Brest, LIEN, F-29200 Brest, France.,Department of Dermatology, Brest University Hospital, Brest, France
| |
Collapse
|
63
|
Yadav A, Matson KJE, Li L, Hua I, Petrescu J, Kang K, Alkaslasi MR, Lee DI, Hasan S, Galuta A, Dedek A, Ameri S, Parnell J, Alshardan MM, Qumqumji FA, Alhamad SM, Wang AP, Poulen G, Lonjon N, Vachiery-Lahaye F, Gaur P, Nalls MA, Qi YA, Maric D, Ward ME, Hildebrand ME, Mery PF, Bourinet E, Bauchet L, Tsai EC, Phatnani H, Le Pichon CE, Menon V, Levine AJ. A cellular taxonomy of the adult human spinal cord. Neuron 2023; 111:328-344.e7. [PMID: 36731429 PMCID: PMC10044516 DOI: 10.1016/j.neuron.2023.01.007] [Citation(s) in RCA: 79] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 11/30/2022] [Accepted: 01/11/2023] [Indexed: 02/04/2023]
Abstract
The mammalian spinal cord functions as a community of cell types for sensory processing, autonomic control, and movement. While animal models have advanced our understanding of spinal cellular diversity, characterizing human biology directly is important to uncover specialized features of basic function and human pathology. Here, we present a cellular taxonomy of the adult human spinal cord using single-nucleus RNA sequencing with spatial transcriptomics and antibody validation. We identified 29 glial clusters and 35 neuronal clusters, organized principally by anatomical location. To demonstrate the relevance of this resource to human disease, we analyzed spinal motoneurons, which degenerate in amyotrophic lateral sclerosis (ALS) and other diseases. We found that compared with other spinal neurons, human motoneurons are defined by genes related to cell size, cytoskeletal structure, and ALS, suggesting a specialized molecular repertoire underlying their selective vulnerability. We include a web resource to facilitate further investigations into human spinal cord biology.
Collapse
Affiliation(s)
- Archana Yadav
- Department of Neurology, Center for Translational and Computational Neuroimmunology, Columbia University, New York, NY, USA
| | - Kaya J E Matson
- Spinal Circuits and Plasticity Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA; Johns Hopkins University Department of Biology, Baltimore, MD 21218, USA
| | - Li Li
- Spinal Circuits and Plasticity Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Isabelle Hua
- Spinal Circuits and Plasticity Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Joana Petrescu
- Department of Neurology, Center for Translational and Computational Neuroimmunology, Columbia University, New York, NY, USA; Center for Genomics of Neurodegenerative Disease, New York Genome Center, New York, NY, USA
| | - Kristy Kang
- Department of Neurology, Center for Translational and Computational Neuroimmunology, Columbia University, New York, NY, USA; Center for Genomics of Neurodegenerative Disease, New York Genome Center, New York, NY, USA
| | - Mor R Alkaslasi
- Unit on the Development of Neurodegeneration, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA; Department of Neuroscience, Brown University, Providence, RI, USA
| | - Dylan I Lee
- Department of Neurology, Center for Translational and Computational Neuroimmunology, Columbia University, New York, NY, USA
| | - Saadia Hasan
- Inherited Neurodegenerative Diseases Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Ahmad Galuta
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Annemarie Dedek
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada; Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | - Sara Ameri
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Jessica Parnell
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada; Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | | | | | - Saud M Alhamad
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Alick Pingbei Wang
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Gaetan Poulen
- Department of Neurosurgery, Gui de Chauliac Hospital, and Donation and Transplantation Coordination Unit, Montpellier University Medical Center, Montpellier, France
| | - Nicolas Lonjon
- Department of Neurosurgery, Gui de Chauliac Hospital, and Donation and Transplantation Coordination Unit, Montpellier University Medical Center, Montpellier, France
| | - Florence Vachiery-Lahaye
- Department of Neurosurgery, Gui de Chauliac Hospital, and Donation and Transplantation Coordination Unit, Montpellier University Medical Center, Montpellier, France
| | - Pallavi Gaur
- Department of Neurology, Center for Translational and Computational Neuroimmunology, Columbia University, New York, NY, USA
| | - Mike A Nalls
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA; Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD, USA; Data Tecnica International LLC, Glen Echo, MD, USA
| | - Yue A Qi
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD, USA
| | - Dragan Maric
- Flow and Imaging Cytometry Core Facility, National Institute of Neurological Disorders and Stroke; Bethesda, MD, USA
| | - Michael E Ward
- Inherited Neurodegenerative Diseases Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Michael E Hildebrand
- Inherited Neurodegenerative Diseases Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA; Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Pierre-Francois Mery
- Institute of Functional Genomics, Montpellier University, CNRS, INSERM, Montpellier, France
| | - Emmanuel Bourinet
- Institute of Functional Genomics, Montpellier University, CNRS, INSERM, Montpellier, France
| | - Luc Bauchet
- Department of Neurosurgery, Gui de Chauliac Hospital, and Donation and Transplantation Coordination Unit, Montpellier University Medical Center, Montpellier, France; Institute of Functional Genomics, Montpellier University, CNRS, INSERM, Montpellier, France
| | - Eve C Tsai
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Hemali Phatnani
- Department of Neurology, Center for Translational and Computational Neuroimmunology, Columbia University, New York, NY, USA; Center for Genomics of Neurodegenerative Disease, New York Genome Center, New York, NY, USA
| | - Claire E Le Pichon
- Unit on the Development of Neurodegeneration, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA
| | - Vilas Menon
- Department of Neurology, Center for Translational and Computational Neuroimmunology, Columbia University, New York, NY, USA.
| | - Ariel J Levine
- Spinal Circuits and Plasticity Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA.
| |
Collapse
|
64
|
Röderer P, Belu A, Heidrich L, Siobal M, Isensee J, Prolingheuer J, Janocha E, Valdor M, Hagendorf S, Bahrenberg G, Opitz T, Segschneider M, Haupt S, Nitzsche A, Brüstle O, Hucho T. Emergence of nociceptive functionality and opioid signaling in human induced pluripotent stem cell-derived sensory neurons. Pain 2023:00006396-990000000-00249. [PMID: 36727909 DOI: 10.1097/j.pain.0000000000002860] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 11/15/2022] [Indexed: 02/03/2023]
Abstract
ABSTRACT Induced pluripotent stem cells (iPSCs) have enabled the generation of various difficult-to-access cell types such as human nociceptors. A key challenge associated with human iPSC-derived nociceptors (hiPSCdNs) is their prolonged functional maturation. While numerous studies have addressed the expression of classic neuronal markers and ion channels in hiPSCdNs, the temporal development of key signaling cascades regulating nociceptor activity has remained largely unexplored. In this study, we used an immunocytochemical high-content imaging approach alongside electrophysiological staging to assess metabotropic and ionotropic signaling of large scale-generated hiPSCdNs across 70 days of in vitro differentiation. During this period, the resting membrane potential became more hyperpolarized, while rheobase, action potential peak amplitude, and membrane capacitance increased. After 70 days, hiPSCdNs exhibited robust physiological responses induced by GABA, pH shift, ATP, and capsaicin. Direct activation of protein kinase A type II (PKA-II) through adenylyl cyclase stimulation with forskolin resulted in PKA-II activation at all time points. Depolarization-induced activation of PKA-II emerged after 35 days of differentiation. However, effective inhibition of forskolin-induced PKA-II activation by opioid receptor agonists required 70 days of in vitro differentiation. Our results identify a pronounced time difference between early expression of functionally important ion channels and emergence of regulatory metabotropic sensitizing and desensitizing signaling only at advanced stages of in vitro cultivation, suggesting an independent regulation of ionotropic and metabotropic signaling. These data are relevant for devising future studies into the development and regulation of human nociceptor function and for defining time windows suitable for hiPSCdN-based drug discovery.
Collapse
Affiliation(s)
- Pascal Röderer
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty and University Hospital Bonn, Bonn
- LIFE & BRAIN GmbH, Cellomics Unit, Bonn, Germany, Germany
| | - Andreea Belu
- Translational Pain Research, Department of Anaesthesiology and Intensive Care Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Luzia Heidrich
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty and University Hospital Bonn, Bonn
- LIFE & BRAIN GmbH, Cellomics Unit, Bonn, Germany, Germany
| | - Maike Siobal
- Translational Pain Research, Department of Anaesthesiology and Intensive Care Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Jörg Isensee
- Translational Pain Research, Department of Anaesthesiology and Intensive Care Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Jonathan Prolingheuer
- Translational Pain Research, Department of Anaesthesiology and Intensive Care Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | | | | | | | | | - Thoralf Opitz
- Institute of Experimental Epileptology and Cognition Research, University of Bonn, Bonn, Germany
| | - Michaela Segschneider
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty and University Hospital Bonn, Bonn
| | - Simone Haupt
- LIFE & BRAIN GmbH, Cellomics Unit, Bonn, Germany, Germany
| | - Anja Nitzsche
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty and University Hospital Bonn, Bonn
- LIFE & BRAIN GmbH, Cellomics Unit, Bonn, Germany, Germany
| | - Oliver Brüstle
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty and University Hospital Bonn, Bonn
- LIFE & BRAIN GmbH, Cellomics Unit, Bonn, Germany, Germany
| | - Tim Hucho
- Translational Pain Research, Department of Anaesthesiology and Intensive Care Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
65
|
Jin Y, Connors T, Bouyer J, Fischer I. Regulation of Tau Expression in Superior Cervical Ganglion (SCG) Neurons In Vivo and In Vitro. Cells 2023; 12:cells12020226. [PMID: 36672160 PMCID: PMC9856632 DOI: 10.3390/cells12020226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 12/23/2022] [Accepted: 12/29/2022] [Indexed: 01/06/2023] Open
Abstract
The superior cervical ganglion (SCG) is part of the autonomic nervous system providing sympathetic innervation to the head and neck, and has been regularly used to prepare postnatal neuronal cultures for cell biological studies. We found that during development these neurons change tau expression from the low molecular weight (LMW) isoforms to Big tau, with the potential to affect functions associated with tau such as microtubule dynamic and axonal transport. Big tau contains the large 4a exon that transforms tau from LMW isoforms of 45-60 kDa to 110 kDa. We describe tau expression during postnatal development reporting that the transition from LMW tau to Big tau which started at late embryonic stages is completed by about 4-5 weeks postnatally. We confirmed the presence of Big tau in dissociated postnatal SCG neurons making them an ideal system to study the function of Big tau in neurons. We used SCG explants to examine the response of SCG neurons to lesion and found that Big tau expression returned gradually along the regrowing neurites suggesting that it does not drives regeneration, but facilitates the structure/function of mature SCG neurons. The structural/functional roles of Big tau remain unknown, but it is intriguing that neurons that express Big tau appear less vulnerable to tauopathies.
Collapse
|
66
|
Giorgi S, Lamberti A, Butrón L, Gross-Amat O, Alarcón-Alarcón D, Rodríguez-Cañas E, Fernández-Carvajal A, Ferrer-Montiel A. Compartmentalized primary cultures of dorsal root ganglion neurons to model peripheral pathophysiological conditions. Mol Pain 2023; 19:17448069231197102. [PMID: 37578145 PMCID: PMC10521292 DOI: 10.1177/17448069231197102] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 07/24/2023] [Accepted: 08/08/2023] [Indexed: 08/15/2023] Open
Abstract
Neurosensory disorders such as pain and pruritus remain a major health problem greatly impacting the quality of life, and often increasing the risk of mortality. Current pre-clinical models to investigate dysfunction of sensory neurons have shown a limited clinical translation, in part, by failing to mimic the compartmentalized nociceptor anatomy that exhibits a central compartment containing the soma and a peripheral one harboring the axon endings with distinct molecular and cellular environmental composition. Thus, there is a need to validate compartmentalized preclinical neurosensory models for investigating the pathophysiology of peripheral sensory disorders and to test drug candidates. Here, we have addressed this issue and developed a microfluidic-based preclinical nociceptor model and validated it for investigating inflammatory and neuropathic peripheral disorders. We show that this model reproduces the peripheral sensitization and resolution produced by an inflammatory soup and by the chemotherapeutic drug paclitaxel. Furthermore, compartmentalized nociceptor primary cultures were amenable to co-culture with keratinocytes in the axonal compartment. Interaction of axonal endings with keratinocytes modulated neuronal responses, consistent with a crosstalk between both cell types. These findings pave the way towards translational pre-clinical sensory models for skin pathophysiological research and drug development.
Collapse
Affiliation(s)
- Simona Giorgi
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universitas Miguel Hernández, Elche, Spain
| | - Angela Lamberti
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universitas Miguel Hernández, Elche, Spain
| | - Laura Butrón
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universitas Miguel Hernández, Elche, Spain
| | - Olivia Gross-Amat
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universitas Miguel Hernández, Elche, Spain
| | - David Alarcón-Alarcón
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universitas Miguel Hernández, Elche, Spain
| | - Enrique Rodríguez-Cañas
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universitas Miguel Hernández, Elche, Spain
| | - Asia Fernández-Carvajal
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universitas Miguel Hernández, Elche, Spain
| | - Antonio Ferrer-Montiel
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universitas Miguel Hernández, Elche, Spain
| |
Collapse
|
67
|
Zhai J, Li X, Zhou Y, Fan L, Xia W, Wang X, Li Y, Hou M, Wang J, Wu L. Correlation and predictive ability of sensory characteristics and social interaction in children with autism spectrum disorder. Front Psychiatry 2023; 14:1056051. [PMID: 37091701 PMCID: PMC10117963 DOI: 10.3389/fpsyt.2023.1056051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 03/13/2023] [Indexed: 04/25/2023] Open
Abstract
Background Individuals with autism spectrum disorder (ASD) often have different social characteristics and particular sensory processing patterns, and these sensory behaviors may affect their social functioning. The objective of our study is to investigate the sensory profiles of children with ASD and their association with social behavior. Specifically, we aim to identify the predictive role of sensory processing in social functioning. Methods The Short Sensory Profile (SSP) was utilized to analyze sensory differences between ASD children and their peers. The Social Responsiveness Scale (SRS) and other clinical scales were employed to assess the social functioning of children with ASD. Additionally, the predictive ability of sensory perception on social performance was discussed using random forest and support vector machine (SVM) models. Results The SSP scores of ASD children were lower than those of the control group, and there was a significant negative correlation between SSP scores and clinical scale scores (P < 0.05). The random forest and SVM models, using all the features, showed higher sensitivity, while the random forest model with 7-feature factors had the highest specificity. The area under the receiver operating characteristic (ROC) curve (AUC) for all the models was higher than 0.8. Conclusion Autistic children in our study have different patterns of sensory processing than their peers, which are significantly related to their patterns of social functioning. Sensory features can serve as a good predictor of social functioning in individuals with ASD.
Collapse
Affiliation(s)
- Jinhe Zhai
- School of Public Health, Harbin Medical University, Harbin, China
| | - Xiaoxue Li
- School of Public Health, Harbin Medical University, Harbin, China
| | - Yong Zhou
- Heilongjiang Provincial Center for Disease Control and Prevention, Harbin, China
| | - Lili Fan
- School of Public Health, Harbin Medical University, Harbin, China
| | - Wei Xia
- School of Public Health, Harbin Medical University, Harbin, China
| | - Xiaomin Wang
- School of Public Health, Harbin Medical University, Harbin, China
| | - Yutong Li
- School of Public Health, Harbin Medical University, Harbin, China
| | - Meiru Hou
- School of Public Health, Harbin Medical University, Harbin, China
| | - Jia Wang
- School of Public Health, Harbin Medical University, Harbin, China
- *Correspondence: Jia Wang,
| | - Lijie Wu
- School of Public Health, Harbin Medical University, Harbin, China
- Lijie Wu,
| |
Collapse
|
68
|
Towards a mechanistic understanding of axon transport and endocytic changes underlying paclitaxel-induced peripheral neuropathy. Exp Neurol 2023; 359:114258. [PMID: 36279934 DOI: 10.1016/j.expneurol.2022.114258] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 10/11/2022] [Accepted: 10/17/2022] [Indexed: 11/11/2022]
Abstract
Paclitaxel is a common chemotherapeutic agent widely used to treat solid cancer. However, it frequently causes peripheral sensory neuropathy, resulting in sensory abnormalities and pain in patients receiving treatment for cancer. As one of the most widely used chemotherapeutics, many preclinical studies on paclitaxel-induced peripheral neuropathy (PIPN) have been performed. Yet, there remain no effective options for treatment or prevention. Due to paclitaxel's ability to bind to and stabilize microtubules, a change in microtubule dynamics and subsequent disruptions in axonal transport has been predicted as a major underlying cause of paclitaxel-induced toxicity. However, the systemic understanding of PIPN mechanisms is largely incomplete, and various phenotypes have not been directly attributed to microtubule-related effects. This review aims to provide an overview of the literature involving paclitaxel-induced alteration in microtubule dynamics, axonal transport, and endocytic changes. It also aims to provide insights into how the microtubule-mediated hypothesis may relate to various phenotypes reported in PIPN studies.
Collapse
|
69
|
Tiwari MN, Hall BE, Ton AT, Ghetti R, Terse A, Amin N, Chung MK, Kulkarni AB. Activation of cyclin-dependent kinase 5 broadens action potentials in human sensory neurons. Mol Pain 2023; 19:17448069231218353. [PMID: 37982142 PMCID: PMC10687939 DOI: 10.1177/17448069231218353] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 11/06/2023] [Accepted: 11/10/2023] [Indexed: 11/21/2023] Open
Abstract
Chronic pain is one of the most devastating and unpleasant conditions, associated with many pathological states. Tissue or nerve injuries induce extensive neurobiological plasticity in nociceptive neurons, which leads to chronic pain. Recent studies suggest that cyclin-dependent kinase 5 (CDK5) in primary afferents is a key neuronal kinase that modulates nociception through phosphorylation under pathological conditions. However, the impact of the CDK5 on nociceptor activity especially in human sensory neurons is not known. To determine the CDK5-mediated regulation of human dorsal root ganglia (hDRG) neuronal properties, we have performed the whole-cell patch clamp recordings in neurons dissociated from hDRG. CDK5 activation induced by overexpression of p35 depolarized the resting membrane potential (RMP) and reduced the rheobase currents as compared to the control neurons. CDK5 activation changed the shape of the action potential (AP) by increasing AP -rise time, -fall time, and -half width. The application of a prostaglandin E2 (PG) and bradykinin (BK) cocktail in control hDRG neurons induced the depolarization of RMP and the reduction of rheobase currents along with increased AP rise time. However, PG and BK applications failed to induce any significant changes in the p35-overexpressing group. We conclude that, in dissociated hDRGs neurons, CDK5 activation through the overexpression of p35 broadens the AP and that CDK5 may play important roles in the modulation of AP properties in human primary afferents under the condition in which CDK5 is upregulated, contributing to chronic pain.
Collapse
Affiliation(s)
- Manindra Nath Tiwari
- Department of Neural and Pain Sciences, School of Dentistry, Program in Neuroscience, Center to Advance Chronic Pain Research, The University of Maryland, Baltimore, MD, United States
| | - Bradford E Hall
- Functional Genomics Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, United States
| | | | - Re Ghetti
- AnaBios, San Diego, CA, United States
| | - Anita Terse
- Functional Genomics Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, United States
| | - Niranjana Amin
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Man-Kyo Chung
- Department of Neural and Pain Sciences, School of Dentistry, Program in Neuroscience, Center to Advance Chronic Pain Research, The University of Maryland, Baltimore, MD, United States
| | - Ashok B Kulkarni
- Functional Genomics Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
70
|
Murakami N, Kurogi A, Suzuki SO, Akitake N, Shimogawa T, Mukae N, Yoshimoto K, Morioka T. Ectopic dorsal root ganglion in cauda equina mimicking schwannoma in a child. Surg Neurol Int 2023; 14:33. [PMID: 36895208 PMCID: PMC9990762 DOI: 10.25259/sni_1089_2022] [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: 12/05/2022] [Accepted: 01/13/2023] [Indexed: 01/28/2023] Open
Abstract
Background A heterotopic dorsal root ganglion (DRG) is sometimes observed in the vicinity of dysplastic neural structures during surgery for open spinal dysraphism; however, it is rarely associated with closed spinal dysraphism. Distinguish from neoplasms by preoperative imaging study is difficult. Although the embryopathogenesis of a heterotopic DRG has been speculated to be migration disorder of neural crest cells from primary neural tube, its details remain unelucidated. Case Description We report a pediatric case with an ectopic DRG in cauda equina associated with a fatty terminal filum and bifid sacrum. The DRG mimicked a schwannoma in the cauda equina on preoperative magnetic resonance imaging. Laminotomy at L3 revealed that the tumor was entangled in the nerve roots, and small parts of the tumor were resected for biopsy. Histopathologically, the tumor consisted of ganglion cells and peripheral nerve fibers. Ki-67 immunopositive cells were observed at the periphery of the ganglion cells. These findings indicate the tumor comprised DRG tissue. Conclusion We report detailed neuroradiological, intraoperative and histological findings and discuss the embryopathogenesis of the ectopic DRG. One should be aware of the possibility of ectopic or heterotopic DRGs when cauda equina tumors are observed in pediatric patients with neurulation disorders.
Collapse
Affiliation(s)
- Nobuya Murakami
- Department of Neurosurgery, Fukuoka Children's Hospital, Fukuoka, Japan
| | - Ai Kurogi
- Department of Neurosurgery, Fukuoka Children's Hospital, Fukuoka, Japan
| | | | - Naoko Akitake
- Department of Urology, Fukuoka Children's Hospital, Fukuoka, Japan
| | - Takafumi Shimogawa
- Department of Neurosurgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Nobutaka Mukae
- Department of Neurosurgery, Iizuka Hospital, Iizuka, Japan
| | - Koji Yoshimoto
- Department of Neurosurgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takato Morioka
- Department of Neurosurgery, Hachisuga Hospital, Munakata, Japan
| |
Collapse
|
71
|
Chen L, Li Y, Zhu L, Jin H, Kang X, Feng Z. Single-cell RNA sequencing in the context of neuropathic pain: progress, challenges, and prospects. Transl Res 2023; 251:96-103. [PMID: 35902034 DOI: 10.1016/j.trsl.2022.07.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 07/17/2022] [Accepted: 07/18/2022] [Indexed: 02/09/2023]
Abstract
Neuropathic pain, characterized by persistent or intermittent spontaneous pain as well as some unpleasant abnormal sensations, is one of the most prevalent health problems in the world. Ectopic nerve activity, central and peripheral nociceptive sensitization and many other potential mechanisms may participate in neuropathic pain. The complexity and ambiguity of neuropathic pain mechanisms result in difficulties in pain management, and existing treatment plans provide less-than-satisfactory relief. In recent years, single-cell RNA sequencing (scRNA-seq) has been increasingly applied and has become a powerful means for biological researchers to explore the complexity of neurobiology. This technique can be used to perform unbiased, high-throughput and high-resolution transcriptional analyses of neuropathic pain-associated cells, improving the understanding of neuropathic pain mechanisms and enabling individualized pain management. To date, scRNA-seq has been preliminarily used in neuropathic pain research for applications such as compiling a dorsal root ganglion atlas, identifying new cell types and discovering gene regulatory networks associated with neuropathic pain. Although scRNA-seq is a relatively new technique in the neuropathic pain field, there have been several studies based on animal models. However, because of the various differences between animals and humans, more attention should be given to translational medicine research. With the aid of scRNA-seq, researchers can further explore the mechanism of neuropathic pain to improve the clinical understanding of the diagnosis, treatment and management of neuropathic pain.
Collapse
Affiliation(s)
- Lei Chen
- Department of Pain Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yunze Li
- Department of Pain Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Lina Zhu
- Department of Pain Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Department of Anesthesiology, Rongjun Hospital of Zhejiang Province, Jiaxing, Zhejiang, China
| | - Haifei Jin
- Department of Pain Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Department of Anesthesiology, Rongjun Hospital of Zhejiang Province, Jiaxing, Zhejiang, China
| | - Xianhui Kang
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - Zhiying Feng
- Department of Pain Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
72
|
Johnson EW, Sutherland JJ, Meseck E, McElroy C, Chand DH, Tukov FF, Hudry E, Penraat K. Neurofilament light chain and dorsal root ganglia injury after adeno-associated virus 9 gene therapy in nonhuman primates. Mol Ther Methods Clin Dev 2022; 28:208-219. [PMID: 36700120 PMCID: PMC9852542 DOI: 10.1016/j.omtm.2022.12.012] [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: 09/19/2022] [Accepted: 12/22/2022] [Indexed: 12/27/2022]
Abstract
In nonhuman primates (NHPs), adeno-associated virus serotype 9 (AAV9) vectorized gene therapy can cause asymptomatic microscopic injury to dorsal root ganglia (DRG) and trigeminal ganglia (TG) somatosensory neurons, causing neurofilament light chain (NfL) to diffuse into cerebrospinal fluid (CSF) and blood. Data from 260 cynomolgus macaques administered vehicle or AAV9 vectors (intrathecally or intravenously) were analyzed to investigate NfL as a soluble biomarker for monitoring DRG/TG microscopic findings. The incidence of key DRG/TG findings with AAV9 vectors was 78% (maximum histopathology severity, moderate) at 2-12 weeks after the dose. When examined up to 52 weeks after the dose, the incidence was 42% (maximum histopathology severity, minimal). Terminal NfL concentrations in plasma, serum, and CSF correlated with microscopic severity. After 52 weeks, NfL returned to pre-dose baseline concentrations, correlating with microscopic findings of lesser incidence and/or severity compared with interim time points. Blood and CSF NfL concentrations correlated with asymptomatic DRG/TG injury, suggesting that monitoring serum and plasma concentrations is as useful for assessment as more invasive CSF sampling. Longitudinal assessment of NfL concentrations related to microscopic findings associated with AAV9 administration in NHPs indicates NfL could be a useful biomarker in nonclinical toxicity testing. Caution should be applied for any translation to humans.
Collapse
Affiliation(s)
- Eric W. Johnson
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | | | - Emily Meseck
- Novartis Pharmaceuticals Corporation, East Hanover, NJ 07936, USA
| | - Cameron McElroy
- Novartis Pharmaceuticals Corporation, East Hanover, NJ 07936, USA
| | - Deepa H. Chand
- Novartis Pharmaceuticals Corporation, East Hanover, NJ 07936, USA,University of Illinois College of Medicine-Peoria, Children’s Hospital of Illinois, Peoria IL 61605, USA
| | | | - Eloise Hudry
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Kelley Penraat
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA,Corresponding author: Kelley Penraat, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA 02139, USA.
| |
Collapse
|
73
|
Tsai NW, Lin CC, Yeh TY, Chiu YA, Chiu HH, Huang HP, Hsieh ST. An induced pluripotent stem cell-based model identifies molecular targets of vincristine neurotoxicity. Dis Model Mech 2022; 15:dmm049471. [PMID: 36518084 PMCID: PMC10655812 DOI: 10.1242/dmm.049471] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 09/29/2022] [Indexed: 11/19/2023] Open
Abstract
To model peripheral nerve degeneration and investigate molecular mechanisms of neurodegeneration, we established a cell system of induced pluripotent stem cell (iPSC)-derived sensory neurons exposed to vincristine, a drug that frequently causes chemotherapy-induced peripheral neuropathy. Sensory neurons differentiated from iPSCs exhibit distinct neurochemical patterns according to the immunocytochemical phenotypes, and gene expression of peripherin (PRPH, hereafter referred to as Peri) and neurofilament heavy chain (NEFH, hereafter referred to as NF). The majority of iPSC-derived sensory neurons were PRPH positive/NEFH negative, i.e. Peri(+)/NF(-) neurons, whose somata were smaller than those of Peri(+)/NF(+) neurons. On exposure to vincristine, projections from the cell body of a neuron, i.e. neurites, were degenerated quicker than somata, the lethal concentration to kill 50% (LC50) of neurites being below the LC50 for somata, consistent with the clinical pattern of length-dependent neuropathy. We then examined the molecular expression in the MAP kinase signaling pathways of, extracellular signal-regulated kinases 1/2 (MAPK1/3, hereafter referred to as ERK), p38 mitogen-activated protein kinases (MAPK11/12/13/14, hereafter referred to as p38) and c-Jun N-terminal kinases (MAPK8/9/10, hereafter referred to as JNK). Regarding these three cascades, only phosphorylation of JNK was upregulated but not that of p38 or ERK1/2. Furthermore, vincristine-treatment resulted in impaired autophagy and reduced autophagic flux. Rapamycin-treatment reversed the effect of impaired autophagy and JNK activation. These results not only established a platform to study peripheral degeneration of human neurons but also provide molecular mechanisms for neurodegeneration with the potential for therapeutic targets.
Collapse
Affiliation(s)
- Neng-Wei Tsai
- Department of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei 100, Taiwan
| | - Cheng-Chen Lin
- Department of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei 100, Taiwan
| | - Ti-Yen Yeh
- Department of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei 100, Taiwan
| | - Yu-An Chiu
- Department of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei 100, Taiwan
| | - Hsin-Hui Chiu
- Department of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei 100, Taiwan
| | - Hsiang-Po Huang
- Department of Medical Genomics and Proteomics, National Taiwan University College of Medicine, Taipei 100, Taiwan
- Department of Pediatrics, National Taiwan University Children's Hospital, Taipei 100, Taiwan
| | - Sung-Tsang Hsieh
- Department of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei 100, Taiwan
- Department of Brain and Mind Sciences, National Taiwan University College of Medicine, Taipei 100, Taiwan
- Department of Neurology, National Taiwan University Hospital, Taipei 100, Taiwan
| |
Collapse
|
74
|
Avraham O, Chamessian A, Feng R, Yang L, Halevi AE, Moore AM, Gereau RW, Cavalli V. Profiling the molecular signature of satellite glial cells at the single cell level reveals high similarities between rodents and humans. Pain 2022; 163:2348-2364. [PMID: 35503034 PMCID: PMC9522926 DOI: 10.1097/j.pain.0000000000002628] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 02/23/2022] [Indexed: 11/25/2022]
Abstract
ABSTRACT Peripheral sensory neurons located in dorsal root ganglia relay sensory information from the peripheral tissue to the brain. Satellite glial cells (SGCs) are unique glial cells that form an envelope completely surrounding each sensory neuron soma. This organization allows for close bidirectional communication between the neuron and its surrounding glial coat. Morphological and molecular changes in SGC have been observed in multiple pathological conditions such as inflammation, chemotherapy-induced neuropathy, viral infection, and nerve injuries. There is evidence that changes in SGC contribute to chronic pain by augmenting the neuronal activity in various rodent pain models. Satellite glial cells also play a critical role in axon regeneration. Whether findings made in rodent model systems are relevant to human physiology have not been investigated. Here, we present a detailed characterization of the transcriptional profile of SGC in mice, rats, and humans at the single cell level. Our findings suggest that key features of SGC in rodent models are conserved in humans. Our study provides the potential to leverage rodent SGC properties and identify potential targets in humans for the treatment of nerve injuries and alleviation of painful conditions.
Collapse
Affiliation(s)
- Oshri Avraham
- Department of Neuroscience, Washington University School of Medicine, St Louis 63110, Missouri, USA
| | - Alexander Chamessian
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis 63110, Missouri, USA
- Department of Neurology, Washington University School of Medicine, St Louis 63110, Missouri, USA
| | - Rui Feng
- Department of Neuroscience, Washington University School of Medicine, St Louis 63110, Missouri, USA
| | - Lite Yang
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis 63110, Missouri, USA
- Neuroscience Program, Washington University School of Medicine, St Louis 63110, Missouri, USA
| | - Alexandra E. Halevi
- Department of Plastic and Reconstructive Surgery, Washington University School of Medicine, St Louis 63110, Missouri, USA
| | - Amy M. Moore
- Department of Plastic and Reconstructive Surgery, The Ohio State University, Columbus Ohio, USA
| | - Robert W. Gereau
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis 63110, Missouri, USA
| | - Valeria Cavalli
- Department of Neuroscience, Washington University School of Medicine, St Louis 63110, Missouri, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri 63110, USA
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| |
Collapse
|
75
|
Kang YR, Hong SM, Choi JH, Lee SJ, Kim JM, Wook Kang K, Nam TS. Sensory neuronopathy in a patient with neurofibromatosis type 1: A case report. Medicine (Baltimore) 2022; 101:e31718. [PMID: 36397418 PMCID: PMC9666172 DOI: 10.1097/md.0000000000031718] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
RATIONALE Neurofibromatosis type 1 (NF-1) can manifest with various neurological symptoms. However, sensory ataxia has not been reported. PATIENT CONCERNS A 44-year-old man with NF-1 presented with several weeks of unsteady gait. He was diagnosed with gastric neuroendocrine tumor with multiple hepatic metastases 6 years ago and received palliative chemotherapy. Neurological examination revealed ataxia veering to the right side with no motor weakness. DIAGNOSES Clinical manifestations and electrodiagnostic studies suggested the dysfunction of the thoracic dorsal column (DC). Initial magnetic resonance imaging showed a lateral thoracic meningocele (LTM) located in the right paravertebral area at the T3-T4 vertebral level, but the spinal cord was unremarkable. Gait disturbance worsened after 9 months, and follow-up magnetic resonance imaging showed high signal intensity involving the right DC at the level adjacent to the LTM and spinal cord atrophy distal to the DC lesion. Tests for well-characterized paraneoplastic antibodies were negative. Ultimately, the patient was assumed to have sensory neuronopathy due to compressive damage to the dorsal root ganglia within the intervertebral foramina by LTM. INTERVENTIONS Empirical treatment with vitamin B12 supplementation and corticosteroids failed to improve his condition. The patient underwent decompressive laminectomy and excision of the meningocele with dura repair. OUTCOMES The patient temporarily improved to walk with assistance postoperatively. However, he developed dyspnea and hypotension 5 weeks later. Carcinoid heart disease confined the patient to the bed. The patient died of pneumonia 3 months after the operation. LESSONS This case with NF-1 shows asymmetric sensory ataxia of subacute progression. LTM may contribute to the development of sensory neuronopathy by damaging sensory neurons of the dorsal root ganglia. The comorbidities of the patient, including gastric neuroendocrine tumor and LTM, made it challenging to investigate the pathomechanism.
Collapse
Affiliation(s)
- You-Ri Kang
- Department of Neurology, Chonnam National University Hospital, Gwangju, Korea
| | - Seong-Min Hong
- Department of Neurology, Chonnam National University Hospital, Gwangju, Korea
| | - Jong-Hee Choi
- Department of Neurology, Chonnam National University Hospital, Gwangju, Korea
| | - Seung-Jin Lee
- Department of Radiology, Chonnam National University Bitgoeul Hospital, Gwangju, Korea
| | - Jae-Myung Kim
- Department of Neurology, Chonnam National University Hospital, Gwangju, Korea
| | - Kyung Wook Kang
- Department of Neurology, Chonnam National University Hospital, Gwangju, Korea
- Department of Neurology, Chonnam National University Medical School, Gwangju, Korea
| | - Tai-Seung Nam
- Department of Neurology, Chonnam National University Hospital, Gwangju, Korea
- Department of Neurology, Chonnam National University Medical School, Gwangju, Korea
- *Correspondence: Tai-Seung Nam, Department of Neurology, Chonnam National University Medical School, Chonnam National University Hospital, 42 Jebong-ro, Dong-gu, Gwangju 61469, Korea (e-mail: )
| |
Collapse
|
76
|
Papalampropoulou-Tsiridou M, Shiers S, Wang F, Godin AG, Price TJ, De Koninck Y. Distribution of acid-sensing ion channel subunits in human sensory neurons contrasts with that in rodents. Brain Commun 2022; 4:fcac256. [PMID: 36337346 PMCID: PMC9629378 DOI: 10.1093/braincomms/fcac256] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 07/27/2022] [Accepted: 10/26/2022] [Indexed: 11/28/2022] Open
Abstract
Acid-sensing ion channels (ASICs) play a critical role in nociception in human sensory neurons. Four genes (ASIC1, ASIC2, ASIC3, and ASIC4) encoding multiple subunits through alternative splicing have been identified in humans. Real time-PCR experiments showed strong expression of three subunits ASIC1, ASIC2, and ASIC3 in human dorsal root ganglia; however, their detailed expression pattern in different neuronal populations has not been investigated yet. In the current study, using an in situ hybridization approach (RNAscope), we examined the presence of ASIC1, ASIC2, and ASIC3 mRNA in three subpopulations of human dorsal root ganglia neurons. Our results revealed that ASIC1 and ASIC3 were present in the vast majority of dorsal root ganglia neurons, while ASIC2 was only expressed in less than half of dorsal root ganglia neurons. The distribution pattern of the three ASIC subunits was the same across the three populations of dorsal root ganglia neurons examined, including neurons expressing the REarranged during Transfection (RET) receptor tyrosine kinase, calcitonin gene-related peptide, and a subpopulation of nociceptors expressing Transient Receptor Potential Cation Channel Subfamily V Member 1. These results strongly contrast the expression pattern of Asics in mice since our previous study demonstrated differential distribution of Asics among the various subpopulation of dorsal root ganglia neurons. Given the distinct acid-sensitivity and activity dynamics among different ASIC channels, the expression differences between human and rodents should be taken under consideration when evaluating the translational potential and efficiency of drugs targeting ASICs in rodent studies.
Collapse
Affiliation(s)
- Melina Papalampropoulou-Tsiridou
- CERVO Brain Research Centre, Québec Mental Health Institute, Québec, QC G1J 2G3, Canada,Graduate Program in Neuroscience, Université Laval, Québec, QC G1V 0A6, Canada
| | - Stephanie Shiers
- Center for Advanced Pain Studies and Department of Neuroscience, University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX 75080, USA
| | - Feng Wang
- CERVO Brain Research Centre, Québec Mental Health Institute, Québec, QC G1J 2G3, Canada
| | - Antoine G Godin
- CERVO Brain Research Centre, Québec Mental Health Institute, Québec, QC G1J 2G3, Canada,Graduate Program in Neuroscience, Université Laval, Québec, QC G1V 0A6, Canada,Department of Psychiatry and Neuroscience, Université Laval, Québec, QC G1V 0A6, Canada
| | - Theodore J Price
- Center for Advanced Pain Studies and Department of Neuroscience, University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX 75080, USA
| | - Yves De Koninck
- Correspondence to: Yves De Koninck 2601 Chemin de la Canardière Québec G1J 2G3 Canada. E-mail:
| |
Collapse
|
77
|
Pricope CV, Tamba BI, Stanciu GD, Cuciureanu M, Neagu AN, Creanga-Murariu I, Dobrovat BI, Uritu CM, Filipiuc SI, Pricope BM, Alexa-Stratulat T. The Roles of Imaging Biomarkers in the Management of Chronic Neuropathic Pain. Int J Mol Sci 2022; 23:13038. [PMID: 36361821 PMCID: PMC9657736 DOI: 10.3390/ijms232113038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/22/2022] [Accepted: 10/24/2022] [Indexed: 08/04/2023] Open
Abstract
Chronic neuropathic pain (CNP) affects around 10% of the general population and has a significant social, emotional, and economic impact. Current diagnosis techniques rely mainly on patient-reported outcomes and symptoms, which leads to significant diagnostic heterogeneity and subsequent challenges in management and assessment of outcomes. As such, it is necessary to review the approach to a pathology that occurs so frequently, with such burdensome and complex implications. Recent research has shown that imaging methods can detect subtle neuroplastic changes in the central and peripheral nervous system, which can be correlated with neuropathic symptoms and may serve as potential markers. The aim of this paper is to review available imaging methods used for diagnosing and assessing therapeutic efficacy in CNP for both the preclinical and clinical setting. Of course, further research is required to standardize and improve detection accuracy, but available data indicate that imaging is a valuable tool that can impact the management of CNP.
Collapse
Affiliation(s)
- Cosmin Vasilica Pricope
- Advanced Research and Development Center for Experimental Medicine (CEMEX), Grigore T. Popa University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania
- Department of Pharmacology, Clinical Pharmacology and Algesiology, Grigore T. Popa University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania
| | - Bogdan Ionel Tamba
- Advanced Research and Development Center for Experimental Medicine (CEMEX), Grigore T. Popa University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania
- Department of Pharmacology, Clinical Pharmacology and Algesiology, Grigore T. Popa University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania
| | - Gabriela Dumitrita Stanciu
- Advanced Research and Development Center for Experimental Medicine (CEMEX), Grigore T. Popa University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania
| | - Magdalena Cuciureanu
- Department of Pharmacology, Clinical Pharmacology and Algesiology, Grigore T. Popa University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania
| | - Anca Narcisa Neagu
- Laboratory of Animal Histology, Faculty of Biology, Alexandru Ioan Cuza University of Iasi, Carol I bvd. No. 22, 700505 Iasi, Romania
| | - Ioana Creanga-Murariu
- Advanced Research and Development Center for Experimental Medicine (CEMEX), Grigore T. Popa University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania
| | - Bogdan-Ionut Dobrovat
- Department of Radiology, Grigore T. Popa University of Medicine and Pharmacy of Iasi, 16 University Street, 700115 Iasi, Romania
| | - Cristina Mariana Uritu
- Advanced Research and Development Center for Experimental Medicine (CEMEX), Grigore T. Popa University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania
| | - Silviu Iulian Filipiuc
- Advanced Research and Development Center for Experimental Medicine (CEMEX), Grigore T. Popa University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania
| | - Bianca-Mariana Pricope
- Department of Preventive Medicine and Interdisciplinarity, Grigore T. Popa University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania
| | - Teodora Alexa-Stratulat
- Advanced Research and Development Center for Experimental Medicine (CEMEX), Grigore T. Popa University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania
- Medical Oncology-Radiotherapy Department, Grigore T. Popa University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania
| |
Collapse
|
78
|
Zdora I, Jubran L, Allnoch L, Hansmann F, Baumgärtner W, Leitzen E. Morphological and phenotypical characteristics of porcine satellite glial cells of the dorsal root ganglia. Front Neuroanat 2022; 16:1015281. [PMID: 36337140 PMCID: PMC9626980 DOI: 10.3389/fnana.2022.1015281] [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] [Received: 08/09/2022] [Accepted: 10/04/2022] [Indexed: 10/23/2023] Open
Abstract
Satellite glial cells (SGCs) of the dorsal root ganglia (DRG) ensure homeostasis and proportional excitability of sensory neurons and gained interest in the field of development and maintenance of neuropathic pain. Pigs represent a suitable species for translational medicine with a more similar anatomy and physiology to humans compared to rodents, and are used in research regarding treatment of neuropathic pain. Knowledge of anatomical and physiological features of porcine SGCs is prerequisite for interpreting potential alterations. However, state of knowledge is still limited. In the present study, light microscopy, ultrastructural analysis and immunofluorescence staining was performed. SGCs tightly surround DRG neurons with little vascularized connective tissue between SGC-neuron units, containing, among others, axons and Schwann cells. DRG were mainly composed of large sized neurons (∼59%), accompanied by fewer medium sized (∼36%) and small sized sensory neurons (∼6%). An increase of neuronal body size was concomitant with an increased number of surrounding SGCs. The majority of porcine SGCs expressed glutamine synthetase and inwardly rectifying potassium channel Kir 4.1, known as SGC-specific markers in other species. Similar to canine SGCs, marked numbers of porcine SGCs were immunopositive for glial fibrillary acidic protein, 2',3'-cyclic-nucleotide 3'-phosphodiesterase and the transcription factor Sox2. Low to moderate numbers of SGCs showed aquaporin 4-immunoreactivity (AQP4) as described for murine SGCs. AQP4-immunoreactivity was primarily found in SGCs ensheathing small and medium sized neuronal somata. Low numbers of SGCs were immunopositive for ionized calcium-binding adapter molecule 1, indicating a potential immune cell character. No immunoreactivity for common leukocyte antigen CD45 nor neural/glial antigen 2 was detected. The present study provides essential insights into the characteristic features of non-activated porcine SGCs, contributing to a better understanding of this cell population and its functional aspects. This will help to interpret possible changes that might occur under activating conditions such as pain.
Collapse
Affiliation(s)
- Isabel Zdora
- Department of Pathology, University of Veterinary Medicine, Hanover, Germany
- Center of Systems Neuroscience, Hanover Graduate School for Neurosciences, Infection Medicine, and Veterinary Sciences (HGNI), Hanover, Germany
| | - Lorna Jubran
- Department of Pathology, University of Veterinary Medicine, Hanover, Germany
- Center of Systems Neuroscience, Hanover Graduate School for Neurosciences, Infection Medicine, and Veterinary Sciences (HGNI), Hanover, Germany
| | - Lisa Allnoch
- Department of Pathology, University of Veterinary Medicine, Hanover, Germany
| | - Florian Hansmann
- Department of Pathology, University of Veterinary Medicine, Hanover, Germany
| | - Wolfgang Baumgärtner
- Department of Pathology, University of Veterinary Medicine, Hanover, Germany
- Center of Systems Neuroscience, Hanover Graduate School for Neurosciences, Infection Medicine, and Veterinary Sciences (HGNI), Hanover, Germany
| | - Eva Leitzen
- Department of Pathology, University of Veterinary Medicine, Hanover, Germany
| |
Collapse
|
79
|
Oleszycka E, Kwiecien K, Kwiecinska P, Morytko A, Pocalun N, Camacho M, Brzoza P, Zabel BA, Cichy J. Soluble mediators in the function of the epidermal-immune-neuro unit in the skin. Front Immunol 2022; 13:1003970. [PMID: 36330530 PMCID: PMC9623011 DOI: 10.3389/fimmu.2022.1003970] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 10/05/2022] [Indexed: 09/19/2023] Open
Abstract
Skin is the largest, environmentally exposed (barrier) organ, capable of integrating various signals into effective defensive responses. The functional significance of interactions among the epidermis and the immune and nervous systems in regulating and maintaining skin barrier function is only now becoming recognized in relation to skin pathophysiology. This review focuses on newly described pathways that involve soluble mediator-mediated crosstalk between these compartments. Dysregulation of these connections can lead to chronic inflammatory diseases and/or pathologic conditions associated with chronic pain or itch.
Collapse
Affiliation(s)
- Ewa Oleszycka
- Department of Immunology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Kamila Kwiecien
- Department of Immunology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Patrycja Kwiecinska
- Department of Immunology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Agnieszka Morytko
- Department of Immunology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Natalia Pocalun
- Department of Immunology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Michelle Camacho
- Department of Immunology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Piotr Brzoza
- Department of Immunology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Brian A. Zabel
- Palo Alto Veterans Institute for Research, VA Palo Alto Health Care System, Palo Alto, CA, United States
| | - Joanna Cichy
- Department of Immunology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| |
Collapse
|
80
|
Andreeva D, Murashova L, Burzak N, Dyachuk V. Satellite Glial Cells: Morphology, functional heterogeneity, and role in pain. Front Cell Neurosci 2022; 16:1019449. [PMID: 36274990 PMCID: PMC9583829 DOI: 10.3389/fncel.2022.1019449] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Neurons in the somatic, sympathetic, and parasympathetic ganglia are surrounded by envelopes consisting of satellite glial cells (SGCs). Recently, it has become clear that SGCs are highly altered after nerve injury, which influences neuronal excitability and, consequently, the development and maintenance of pain in different animal models of chronic pain. However, the exact mechanism underlying chronic pain is not fully understood yet because it is assumed that SGCs in different ganglia share many common peculiarities, making the process complex. Here, we review recent data on morphological and functional heterogeneity and changes in SGCs in various pain conditions and their role in response to injury. More research is required to decipher the role of SGCs in diseases, such as chronic pain, neuropathology, and neurodegenerative diseases.
Collapse
|
81
|
Antoine JC. Sensory neuronopathies, diagnostic criteria and causes. Curr Opin Neurol 2022; 35:553-561. [PMID: 35950727 DOI: 10.1097/wco.0000000000001105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW To stress on the diagnostic strategy of sensory neuronopathies (SNN), including new genes and antibodies. RECENT FINDING SNN involve paraneoplastic, dysimmune, toxic, viral and genetic mechanisms. About one-third remains idiopathic. Recently, new antibodies and genes have reduced this proportion. Anti-FGFR3 and anti-AGO antibodies are not specific of SNN, although SNN is predominant and may occur with systemic autoimmune diseases. These antibodies are the only marker of an underlying dysimmune context in two-thirds (anti-FGFR3 antibodies) and one-third of the cases (anti-AGO antibodies), respectively. Patients with anti-AGO antibodies may improve with treatment, which is less clear with anti-FGFR3 antibodies. A biallelic expansion in the RFC1 gene is responsible for the cerebellar ataxia, neuropathy, vestibular areflexia syndrome (CANVAS) in which SNN is a predominant manifestation. Most of the patients have an adult onset and are sporadic. The RFC1 mutation may represent one-third of idiopathic sensory neuropathies. Finally, the criteria for the diagnosis of paraneoplastic SNN have recently been updated. SUMMARY The diagnostic of SNN relies on criteria distinguishing SNN from other neuropathies. The strategy in search of their cause now needs to include these recent findings.
Collapse
Affiliation(s)
- Jean-Christophe Antoine
- University Hospital of Saint-Etienne, European Reference Network for Rare Diseases- Euro-NMD, INSERM U1314/CNRS UMR 5284, Université Claude Bernard Lyon 1, Lyon, France
| |
Collapse
|
82
|
Use of a human induced pluripotent stem cell-derived dorsal root ganglion neurone model to study analgesics in vitro: proof of principle using lidocaine. Br J Anaesth 2022; 129:e172-e175. [DOI: 10.1016/j.bja.2022.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/04/2022] [Accepted: 09/17/2022] [Indexed: 11/17/2022] Open
|
83
|
Hargreaves R, Akinsanya K, Ajit SK, Dhruv NT, Driscoll J, Farina P, Gavva N, Gill M, Houghton A, Iyengar S, Jones C, Kavelaars A, Kaykas A, Koroshetz WJ, Laeng P, Laird JM, Lo DC, Luthman J, Munro G, Oshinsky ML, Sittampalam GS, Woller SA, Tamiz AP. Preclinical target validation for non-addictive therapeutics development for pain. Expert Opin Ther Targets 2022; 26:811-822. [DOI: 10.1080/14728222.2022.2147063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
| | | | - Seena K. Ajit
- Department of Pharmacology, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States
| | - Neel T. Dhruv
- National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, United States
| | - Jamie Driscoll
- National Institute of Mental Health, Bethesda, Maryland, United States
| | - Peter Farina
- Canaan Partners, Westport, Connecticut, United States
| | - Narender Gavva
- Drug Discovery Sciences, Takeda Pharmaceuticals, San Diego, California, United States
| | - Marie Gill
- National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, United States
| | | | - Smriti Iyengar
- National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, United States
| | - Carrie Jones
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, United States
| | - Annemieke Kavelaars
- The University of Texas MD Anderson Cancer Center, Houston, Texas, United States
| | | | - Walter J. Koroshetz
- National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, United States
| | - Pascal Laeng
- National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, United States
| | - Jennifer M. Laird
- Eli Lilly and Company, Windlesham, United Kingdom of Great Britain and Northern Ireland
| | - Donald C. Lo
- National Center for Advancing Translational Sciences, Bethesda, Maryland, United States
| | | | | | - Michael L. Oshinsky
- National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, United States
| | - G. Sitta Sittampalam
- National Center for Advancing Translational Sciences, Bethesda, Maryland, United States
| | - Sarah A. Woller
- National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, United States
| | - Amir P. Tamiz
- National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, United States
| |
Collapse
|
84
|
McIlvried LA, Atherton MA, Horan NL, Goch TN, Scheff NN. Sensory Neurotransmitter Calcitonin Gene-Related Peptide Modulates Tumor Growth and Lymphocyte Infiltration in Oral Squamous Cell Carcinoma. Adv Biol (Weinh) 2022; 6:e2200019. [PMID: 35388989 PMCID: PMC9474661 DOI: 10.1002/adbi.202200019] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 03/15/2022] [Indexed: 01/28/2023]
Abstract
Head and neck squamous cell carcinoma are highly innervated by peripheral sensory neurons. Local neurotransmitter release (e.g., calcitonin gene-related peptide (CGRP)) from sensory neurons innervating cancer is linked to tumorigenesis. CGRP-immunoreactive nerve presence comprised 9.53±1.9% of total nerve area across 11 HNSCC patients. A syngeneic tongue tumor transplant mouse model of oral cancer and a global Calca knockout mouse (CGRPKO ) are used to investigate the impact of CGRP signaling on tumor growth and the associated immune response in vivo. In tumor-bearing CGRPKO mice, there is a significant reduction in tumor size over time compared to wildtype mice using two different mouse oral cancer cell lines. Furthermore, tumor tissue from CGRPKO mice had a significant increase in tumor-infiltrating CD4+ T cells, cytotoxic CD8+ T cells, and NK1.1+ NK cells compared to wildtype. Fluorescent-activated cell sorting and real-time qPCR are used to confirm that CD4+ T cells are isolated from tumor-bearing wildtype mice containing a high expression of Ramp1 compared to sham mice. These data suggest that sensory neurotransmitter CGRP may modulate oral cancer progression via tumor immunosurveillance. Understanding the relationship between sensory neurons and cancer will aid in repurposing clinically available nervous system drugs for the treatment of cancer.
Collapse
Affiliation(s)
- Lisa A McIlvried
- Department of Neurobiology, University of Pittsburgh, 4200 Fifth Ave, Pittsburgh, PA, 15260, USA
- Hillman Cancer Center, University of Pittsburgh Medical Center, 5117 Centre Ave, Pittsburgh, PA, 15213, USA
| | - Megan A Atherton
- Department of Neurobiology, University of Pittsburgh, 4200 Fifth Ave, Pittsburgh, PA, 15260, USA
- Hillman Cancer Center, University of Pittsburgh Medical Center, 5117 Centre Ave, Pittsburgh, PA, 15213, USA
| | - Nicole L Horan
- Department of Neurobiology, University of Pittsburgh, 4200 Fifth Ave, Pittsburgh, PA, 15260, USA
| | - Tori N Goch
- Hillman Cancer Center, University of Pittsburgh Medical Center, 5117 Centre Ave, Pittsburgh, PA, 15213, USA
| | - Nicole N Scheff
- Department of Neurobiology, University of Pittsburgh, 4200 Fifth Ave, Pittsburgh, PA, 15260, USA
- Hillman Cancer Center, University of Pittsburgh Medical Center, 5117 Centre Ave, Pittsburgh, PA, 15213, USA
| |
Collapse
|
85
|
T-Type Calcium Channels: A Mixed Blessing. Int J Mol Sci 2022; 23:ijms23179894. [PMID: 36077291 PMCID: PMC9456242 DOI: 10.3390/ijms23179894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 08/26/2022] [Accepted: 08/29/2022] [Indexed: 11/17/2022] Open
Abstract
The role of T-type calcium channels is well established in excitable cells, where they preside over action potential generation, automaticity, and firing. They also contribute to intracellular calcium signaling, cell cycle progression, and cell fate; and, in this sense, they emerge as key regulators also in non-excitable cells. In particular, their expression may be considered a prognostic factor in cancer. Almost all cancer cells express T-type calcium channels to the point that it has been considered a pharmacological target; but, as the drugs used to reduce their expression are not completely selective, several complications develop, especially within the heart. T-type calcium channels are also involved in a specific side effect of several anticancer agents, that act on microtubule transport, increase the expression of the channel, and, thus, the excitability of sensory neurons, and make the patient more sensitive to pain. This review puts into context the relevance of T-type calcium channels in cancer and in chemotherapy side effects, considering also the cardiotoxicity induced by new classes of antineoplastic molecules.
Collapse
|
86
|
Su PYP, Zhang L, He L, Zhao N, Guan Z. The Role of Neuro-Immune Interactions in Chronic Pain: Implications for Clinical Practice. J Pain Res 2022; 15:2223-2248. [PMID: 35957964 PMCID: PMC9359791 DOI: 10.2147/jpr.s246883] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 07/19/2022] [Indexed: 11/23/2022] Open
Abstract
Chronic pain remains a public health problem and contributes to the ongoing opioid epidemic. Current pain management therapies still leave many patients with poorly controlled pain, thus new or improved treatments are desperately needed. One major challenge in pain research is the translation of preclinical findings into effective clinical practice. The local neuroimmune interface plays an important role in the initiation and maintenance of chronic pain and is therefore a promising target for novel therapeutic development. Neurons interface with immune and immunocompetent cells in many distinct microenvironments along the nociceptive circuitry. The local neuroimmune interface can modulate the activity and property of the neurons to affect peripheral and central sensitization. In this review, we highlight a specific subset of many neuroimmune interfaces. In the central nervous system, we examine the interface between neurons and microglia, astrocytes, and T lymphocytes. In the periphery, we profile the interface between neurons in the dorsal root ganglion with T lymphocytes, satellite glial cells, and macrophages. To bridge the gap between preclinical research and clinical practice, we review the preclinical studies of each neuroimmune interface, discuss current clinical treatments in pain medicine that may exert its action at the neuroimmune interface, and highlight opportunities for future clinical research efforts.
Collapse
Affiliation(s)
- Po-Yi Paul Su
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, USA
| | - Lingyi Zhang
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, USA
- Department of Anesthesiology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, People’s Republic of China
| | - Liangliang He
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, USA
- Department of Pain Management, Xuanwu Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Na Zhao
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, USA
| | - Zhonghui Guan
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
87
|
North RY, Odem MA, Li Y, Tatsui CE, Cassidy RM, Dougherty PM, Walters ET. Electrophysiological Alterations Driving Pain-Associated Spontaneous Activity in Human Sensory Neuron Somata Parallel Alterations Described in Spontaneously Active Rodent Nociceptors. THE JOURNAL OF PAIN 2022; 23:1343-1357. [PMID: 35292377 PMCID: PMC9357108 DOI: 10.1016/j.jpain.2022.02.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 02/21/2022] [Accepted: 02/24/2022] [Indexed: 06/10/2023]
Abstract
Neuropathic pain in rodents can be driven by ectopic spontaneous activity (SA) generated by sensory neurons in dorsal root ganglia (DRG). The recent demonstration that SA in dissociated human DRG neurons is associated with reported neuropathic pain in patients enables a detailed comparison of pain-linked electrophysiological alterations driving SA in human DRG neurons to alterations that distinguish SA in nociceptors from SA in low-threshold mechanoreceptors (LTMRs) in rodent neuropathy models. Analysis of recordings from dissociated somata of patient-derived DRG neurons showed that SA and corresponding pain in both sexes were significantly associated with the three functional electrophysiological alterations sufficient to generate SA in the absence of extrinsic depolarizing inputs. These include enhancement of depolarizing spontaneous fluctuations of membrane potential (DSFs), which were analyzed quantitatively for the first time in human DRG neurons. The functional alterations were indistinguishable from SA-driving alterations reported for nociceptors in rodent chronic pain models. Irregular, low-frequency DSFs in human DRG neurons closely resemble DSFs described in rodent nociceptors while differing substantially from the high-frequency sinusoidal oscillations described in rodent LTMRs. These findings suggest that conserved physiological mechanisms of SA in human nociceptor somata can drive neuropathic pain despite documented cellular differences between human and rodent DRG neurons. PERSPECTIVE: Electrophysiological alterations in human sensory neurons associated with patient-reported neuropathic pain include all three of the functional alterations that logically can promote spontaneous activity. The similarity of distinctively altered spontaneous depolarizations in human DRG neurons and rodent nociceptors suggests that spontaneously active human nociceptors can persistently promote neuropathic pain in patients.
Collapse
Affiliation(s)
- Robert Y North
- Department of Neurosurgery, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Max A Odem
- Department of Microbiology and Molecular Genetics, McGovern Medical School at UTHealth, Houston, Texas
| | - Yan Li
- Department of Anesthesia and Pain Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Claudio Esteves Tatsui
- Department of Neurosurgery, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Ryan M Cassidy
- M.D. Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas
| | - Patrick M Dougherty
- Department of Anesthesia and Pain Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Edgar T Walters
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UTHealth, Houston, Texas..
| |
Collapse
|
88
|
Anapindi KDB, Romanova EV, Checco JW, Sweedler JV. Mass Spectrometry Approaches Empowering Neuropeptide Discovery and Therapeutics. Pharmacol Rev 2022; 74:662-679. [PMID: 35710134 PMCID: PMC9553102 DOI: 10.1124/pharmrev.121.000423] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The discovery of insulin in the early 1900s ushered in the era of research related to peptides acting as hormones and neuromodulators, among other regulatory roles. These essential gene products are found in all organisms, from the most primitive to the most evolved, and carry important biologic information that coordinates complex physiology and behavior; their misregulation has been implicated in a variety of diseases. The evolutionary origins of at least 30 neuropeptide signaling systems have been traced to the common ancestor of protostomes and deuterostomes. With the use of relevant animal models and modern technologies, we can gain mechanistic insight into orthologous and paralogous endogenous peptides and translate that knowledge into medically relevant insights and new treatments. Groundbreaking advances in medicine and basic science influence how signaling peptides are defined today. The precise mechanistic pathways for over 100 endogenous peptides in mammals are now known and have laid the foundation for multiple drug development pipelines. Peptide biologics have become valuable drugs due to their unique specificity and biologic activity, lack of toxic metabolites, and minimal undesirable interactions. This review outlines modern technologies that enable neuropeptide discovery and characterization, and highlights lessons from nature made possible by neuropeptide research in relevant animal models that is being adopted by the pharmaceutical industry. We conclude with a brief overview of approaches/strategies for effective development of peptides as drugs. SIGNIFICANCE STATEMENT: Neuropeptides, an important class of cell-cell signaling molecules, are involved in maintaining a range of physiological functions. Since the discovery of insulin's activity, over 100 bioactive peptides and peptide analogs have been used as therapeutics. Because these are complex molecules not easily predicted from a genome and their activity can change with subtle chemical modifications, mass spectrometry (MS) has significantly empowered peptide discovery and characterization. This review highlights contributions of MS-based research towards the development of therapeutic peptides.
Collapse
Affiliation(s)
- Krishna D B Anapindi
- Department of Chemistry and the Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, Illinois (K.D.B.A., E.V.R., J.V.S.) and Department of Chemistry, University of Nebraska-Lincoln, Lincoln, Nebraska (J.W.C.)
| | - Elena V Romanova
- Department of Chemistry and the Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, Illinois (K.D.B.A., E.V.R., J.V.S.) and Department of Chemistry, University of Nebraska-Lincoln, Lincoln, Nebraska (J.W.C.)
| | - James W Checco
- Department of Chemistry and the Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, Illinois (K.D.B.A., E.V.R., J.V.S.) and Department of Chemistry, University of Nebraska-Lincoln, Lincoln, Nebraska (J.W.C.)
| | - Jonathan V Sweedler
- Department of Chemistry and the Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, Illinois (K.D.B.A., E.V.R., J.V.S.) and Department of Chemistry, University of Nebraska-Lincoln, Lincoln, Nebraska (J.W.C.)
| |
Collapse
|
89
|
Dorsal Root Ganglia Volume—Normative Values, Correlation with Demographic Determinants and Reliability of Three Different Methods of Volumetry. Diagnostics (Basel) 2022; 12:diagnostics12071570. [PMID: 35885475 PMCID: PMC9323629 DOI: 10.3390/diagnostics12071570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/22/2022] [Accepted: 06/27/2022] [Indexed: 11/17/2022] Open
Abstract
Background: Dorsal root ganglia (DRG) volume assessment by MR-Neurography (MRN) has evolved to an important imaging marker in the diagnostic workup of various peripheral neuropathies and pain syndromes. The aim of this study was (1) to assess normal values of DRG volume and correlations with demographic determinants and (2) to quantify the inter-reader and inter-method reliability of three different methods of DRG volumetry. Methods: Sixty healthy subjects (mean age: 59.1, range 23–79) were examined using a 3D T2-weighted MRN of the lumbosacral plexus at 3 Tesla. Normal values of DRG L3 to S2 were obtained after exact volumetry based on manual 3D segmentation and correlations with demographic variables were assessed. For the assessment of inter-reader and inter-method reliability, DRG volumes in a subset of 25 participants were measured by two independent readers, each applying (1) exact volumetry based on 3D segmentation, (2) axis-corrected, and (3) non-axis-corrected volume estimation. Intraclass correlation coefficients were reported and the Bland–Altman analysis was conducted. Results: Mean DRG volumes ranged from 124.8 mm3 for L3 to 323.3 mm3 for S1 and did not differ between right and left DRG. DRG volume (mean of L3 to S1) correlated with body height (r = 0.42; p = 0.0008) and weight (r = 0.34; p = 0.0087). DRG of men were larger than of women (p = 0.0002); however, no difference remained after correction for body height. Inter-reader reliability was high for all three methods but best for exact volumetry (ICC = 0.99). While axis-corrected estimation was not associated with a relevant bias, non-axis-corrected estimation systematically overestimated DRG volume by on average of 15.55 mm3 (reader 1) or 18.00 mm3 (reader 2) when compared with exact volumetry. Conclusion: The here presented normal values of lumbosacral DRG volume and the correlations with height and weight may be considered in future disease specific studies and possible clinical applications. Exact volumetry was most reliable and should be considered the gold standard. However, the reliability of axis-corrected and non-axis-corrected volume estimation was also high and might still be sufficient, depending on the degree of the required measurement accuracy.
Collapse
|
90
|
Valentine T, Hardowar L, Elphick-Ross J, Hulse RP, Paul-Clark M. The Role of Vascular-Immune Interactions in Modulating Chemotherapy Induced Neuropathic Pain. Front Pharmacol 2022; 13:887608. [PMID: 35814225 PMCID: PMC9257211 DOI: 10.3389/fphar.2022.887608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/23/2022] [Indexed: 12/05/2022] Open
Abstract
Chemotherapy causes sensory disturbances in cancer patients that results in neuropathies and pain. As cancer survivorships has dramatically increased over the past 10 years, pain management of these patients is becoming clinically more important. Current analgesic strategies are mainly ineffective and long-term use is associated with severe side effects. The issue being that common analgesic strategies are based on ubiquitous pain mediator pathways, so when applied to clinically diverse neuropathic pain and neurological conditions, are unsuccessful. This is principally due to the lack of understanding of the driving forces that lead to chemotherapy induced neuropathies. It is well documented that chemotherapy causes sensory neurodegeneration through axonal atrophy and intraepidermal fibre degeneration causing alterations in pain perception. Despite the neuropathological alterations associated with chemotherapy-induced neuropathic pain being extensively researched, underlying causes remain elusive. Resent evidence from patient and rodent studies have indicated a prominent inflammatory cell component in the peripheral sensory nervous system in effected areas post chemotherapeutic treatment. This is accompanied by modulation of auxiliary cells of the dorsal root ganglia sensory neurons such as activation of satellite glia and capillary dysfunction. The presence of a neuroinflammatory component was supported by transcriptomic analysis of dorsal root ganglia taken from mice treated with common chemotherapy agents. With key inflammatory mediators identified, having potent immunoregulatory effects that directly influences nociception. We aim to evaluate the current understanding of these immune-neuronal interactions across different cancer therapy drug classes. In the belief this may lead to better pain management approaches for cancer survivors.
Collapse
|
91
|
Fader KA, Pardo ID, Kovi RC, Somps CJ, Wang HH, Vaidya VS, Ramaiah SK, Sirivelu MP. Circulating neurofilament light chain as a promising biomarker of AAV-induced dorsal root ganglia toxicity in nonclinical toxicology species. Mol Ther Methods Clin Dev 2022; 25:264-277. [PMID: 35505662 PMCID: PMC9024379 DOI: 10.1016/j.omtm.2022.03.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 03/27/2022] [Indexed: 12/14/2022]
Abstract
Adeno-associated virus (AAV)-induced dorsal root ganglia (DRG) toxicity has been observed in several nonclinical species, where lesions are characterized by neuronal degeneration/necrosis, nerve fiber degeneration, and mononuclear cell infiltration. As AAV vectors become an increasingly common platform for novel therapeutics, non-invasive biomarkers are needed to better characterize and manage the risk of DRG neurotoxicity in both nonclinical and clinical studies. Based on biological relevance, reagent availability, antibody cross-reactivity, DRG protein expression, and assay performance, neurofilament light chain (NF-L) emerged as a promising biomarker candidate. Dose- and time-dependent changes in NF-L were evaluated in male Wistar Han rats and cynomolgus monkeys following intravenous or intrathecal AAV injection, respectively. NF-L profiles were then compared against microscopic DRG lesions on day 29 post-dosing. In animals exhibiting DRG toxicity, plasma/serum NF-L was strongly associated with the severity of neuronal degeneration/necrosis and nerve fiber degeneration, with elevations beginning as early as day 8 in rats (≥5 × 1013 vg/kg) and day 14 in monkeys (≥3.3 × 1013 vg/dose). Consistent with the unique positioning of DRGs outside the blood-brain barrier, NF-L in cerebrospinal fluid was only weakly associated with DRG findings. In summary, circulating NF-L is a promising biomarker of AAV-induced DRG toxicity in nonclinical species.
Collapse
Affiliation(s)
- Kelly A Fader
- Pfizer Worldwide Research, Development and Medical, Drug Safety Research and Development, Groton, CT 06340, USA
| | | | - Ramesh C Kovi
- Pfizer Worldwide Research, Development and Medical, Drug Safety Research and Development, Pfizer Inc., 300 Technology Square, Cambridge, MA 02139, USA
| | - Christopher J Somps
- Pfizer Worldwide Research, Development and Medical, Drug Safety Research and Development, Groton, CT 06340, USA
| | - Helen Hong Wang
- Pfizer Worldwide Research, Development and Medical, Drug Safety Research and Development, Pfizer Inc., 300 Technology Square, Cambridge, MA 02139, USA
| | - Vishal S Vaidya
- Pfizer Worldwide Research, Development and Medical, Drug Safety Research and Development, Pfizer Inc., 300 Technology Square, Cambridge, MA 02139, USA
| | - Shashi K Ramaiah
- Pfizer Worldwide Research, Development and Medical, Drug Safety Research and Development, Pfizer Inc., 300 Technology Square, Cambridge, MA 02139, USA
| | - Madhu P Sirivelu
- Pfizer Worldwide Research, Development and Medical, Drug Safety Research and Development, Pfizer Inc., 300 Technology Square, Cambridge, MA 02139, USA
| |
Collapse
|
92
|
Metea M, Palmero-Soler E, Crum L. Preclinical nerve conduction: Nerve battery options for primate studies. J Pharmacol Toxicol Methods 2022; 116:107187. [PMID: 35636693 DOI: 10.1016/j.vascn.2022.107187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 04/23/2022] [Accepted: 05/24/2022] [Indexed: 10/18/2022]
Abstract
Electrophysiological neurodiagnostic tests of nerve conduction (NC) are key assays included in preclinical safety and toxicology programs to assess the peripheral neuropathy (PN) liability of a new drug. Despite their increased use, standardization of nerve conduction studies (NCS) is lacking in the preclinical space, with limited regulatory guidelines stipulating type and number of nerves or minimum combinations appropriate for each stage of drug development or indication. Detection of subtle peripheral toxicities depends on choosing appropriate nerve targets for testing, especially when functional changes remain above the lower limit of normal values. To support robust preclinical toxicology study designs, the current short communication provides options and recommendations for selecting peripheral nerves for clinically translatable nerve conduction batteries applicable to toxicology and gene therapy, with a focus on clinically translatable primate models. A comprehensive compilation of accessible nerve locations is offered including lower and upper extremity motor nerves, and sensory nerves with origin at multiple DRG levels. Rankings of technique difficulty and repeatability across serial collections are presented for each assay informed by serial nerve conduction from 500 adult primates. The goal of this communication is to support the standardization and preclinical implementation of this important assay.
Collapse
Affiliation(s)
- Monica Metea
- Preclinical Electrophysiology Consulting, LLC, Mattapoisett, MA, USA.
| | | | - Lucas Crum
- Preclinical Electrophysiology Consulting, LLC, Mattapoisett, MA, USA
| |
Collapse
|
93
|
Weiner S, Strinitz M, Herfurth J, Hessenauer F, Nauroth-Kreß C, Kampf T, Homola GA, Üçeyler N, Sommer C, Pham M, Schindehütte M. Dorsal Root Ganglion Volumetry by MR Gangliography. AJNR Am J Neuroradiol 2022; 43:769-775. [PMID: 35450855 PMCID: PMC9089252 DOI: 10.3174/ajnr.a7487] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 02/12/2022] [Indexed: 12/29/2022]
Abstract
BACKGROUND AND PURPOSE Dorsal root ganglion MR imaging (MR gangliography) is increasingly gaining clinical-scientific relevance. However, dorsal root ganglion morphometry by MR imaging is typically performed under the assumption of ellipsoid geometry, which remains to be validated. MATERIALS AND METHODS Sixty-four healthy volunteers (37 [57.8%] men; mean age, 31.5 [SD, 8.3] years) underwent MR gangliography of the bilateral L4-S2 levels (3D-T2WI TSE spectral attenuated inversion recovery-sampling perfection with application-optimized contrasts by using different flip angle evolution, isotropic voxels = 1.1 mm³, TE = 301 ms). Ground truth dorsal root ganglion volumes were bilaterally determined for 96 dorsal root ganglia (derivation cohort) by expert manual 3D segmentation by 3 independent raters. These ground truth dorsal root ganglion volumes were then compared with geometric ellipsoid dorsal root ganglion approximations as commonly practiced for dorsal root ganglion morphometry. On the basis of the deviations from ellipsoid geometry, improved volume estimation could be derived and was finally applied to a large human validation cohort (510 dorsal root ganglia). RESULTS Commonly used equations of ellipsoid geometry underestimate true dorsal root ganglion volume by large degrees (factor = 0.42-0.63). Ground truth segmentation enabled substantially optimizing dorsal root ganglion geometric approximation using its principal axes lengths by deriving the dorsal root ganglion volume term of [Formula: see text]. Using this optimization, the mean volumes of 510 lumbosacral healthy dorsal root ganglia were as follows: L4: 211.3 (SD, 52.5) mm³, L5: 290.7 (SD, 90.9) mm³, S1: 384.2 (SD, 145.0) mm³, and S2: 192.4 (SD, 52.6) mm³. Dorsal root ganglion volume increased from L4 to S1 and decreased from S1 to S2 (P < .001). Dorsal root ganglion volume correlated with subject height (r = . 22, P < .001) and was higher in men (P < .001). CONCLUSIONS Dorsal root ganglion volumetry by measuring its principal geometric axes on MR gangliography can be substantially optimized. By means of this optimization, dorsal root ganglion volume distribution was estimated in a large healthy cohort for the clinically most relevant lumbosacral levels, L4-S2.
Collapse
Affiliation(s)
- S Weiner
- From the Department of Neuroradiology (S.W., M.S., J.H., F.H., C.N.-K., T.K., G.A.H., M.P., M.S.)
| | - M Strinitz
- From the Department of Neuroradiology (S.W., M.S., J.H., F.H., C.N.-K., T.K., G.A.H., M.P., M.S.)
| | - J Herfurth
- From the Department of Neuroradiology (S.W., M.S., J.H., F.H., C.N.-K., T.K., G.A.H., M.P., M.S.)
| | - F Hessenauer
- From the Department of Neuroradiology (S.W., M.S., J.H., F.H., C.N.-K., T.K., G.A.H., M.P., M.S.)
| | - C Nauroth-Kreß
- From the Department of Neuroradiology (S.W., M.S., J.H., F.H., C.N.-K., T.K., G.A.H., M.P., M.S.)
| | - T Kampf
- From the Department of Neuroradiology (S.W., M.S., J.H., F.H., C.N.-K., T.K., G.A.H., M.P., M.S.)
| | - G A Homola
- From the Department of Neuroradiology (S.W., M.S., J.H., F.H., C.N.-K., T.K., G.A.H., M.P., M.S.)
| | - N Üçeyler
- Neurology (N.U., C.S.), University Hospital Würzburg, Würzburg, Germany
| | - C Sommer
- Neurology (N.U., C.S.), University Hospital Würzburg, Würzburg, Germany
| | - M Pham
- From the Department of Neuroradiology (S.W., M.S., J.H., F.H., C.N.-K., T.K., G.A.H., M.P., M.S.)
| | - M Schindehütte
- From the Department of Neuroradiology (S.W., M.S., J.H., F.H., C.N.-K., T.K., G.A.H., M.P., M.S.)
| |
Collapse
|
94
|
Tukov FF, Mansfield K, Milton M, Meseck E, Penraat K, Chand D, Hartmann A. Single-Dose Intrathecal Dorsal Root Ganglia Toxicity of Onasemnogene Abeparvovec in Cynomolgus Monkeys. Hum Gene Ther 2022; 33:740-756. [PMID: 35331006 PMCID: PMC9347375 DOI: 10.1089/hum.2021.255] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Intravenous onasemnogene abeparvovec is approved for the treatment of spinal muscular atrophy in children < 2 years. For later-onset patients, intrathecal onasemnogene abeparvovec may be advantageous over intravenous administration. Recently, microscopic dorsal root ganglion (DRG) changes were observed in nonhuman primates (NHPs) following intrathecal onasemnogene abeparvovec administration. To characterize these DRG findings, two NHP studies evaluating intrathecal onasemnogene abeparvovec administration were conducted: a 12-month study with a 6-week interim cohort and a 13-week study with a 2-week interim cohort. The latter investigated the potential impact of prednisolone or rituximab plus everolimus on DRG toxicity. An additional 6-month, single-dose, intravenous NHP study conducted in parallel evaluated onasemnogene abeparvovec safety (including DRG toxicity) with or without prednisolone coadministration. Intrathecal onasemnogene abeparvovec administration was well tolerated and not associated with clinical observations. Microscopic onasemnogene abeparvovec-related changes were observed in the DRG and trigeminal ganglion (TG) and included mononuclear cell inflammation and/or neuronal degeneration, which was colocalized with high vector transcript expression at 6 weeks postdose. Incidence and severity of DRG changes were generally decreased after 52 weeks compared with 6 weeks postdose. Other onasemnogene abeparvovec-related microscopic findings of axonal degeneration, mononuclear cell infiltrates and/or gliosis in the spinal cord, dorsal spinal nerve root/spinal nerves, and/or peripheral nerves were absent or found at decreased incidences and/or severities after 52 weeks. DRG and/or TG microscopic findings following intravenous onasemnogene abeparvovec dosing included minimal to slight neuronal degeneration and mononuclear cell inflammation at 6 weeks and 6 months postdose. Nervous system microscopic findings following intrathecal onasemnogene abeparvovec (≥1.2 × 1013 vg/animal) trended toward resolution after 52 weeks, supporting nonprogression of changes, including in the DRG. Onasemnogene abeparvovec-related DRG findings were not associated with electrophysiology changes and were not ameliorated by prednisolone or rituximab plus everolimus coadministration. The pathogenesis is possibly a consequence of increased vector genome transduction and/or transgene expression.
Collapse
Affiliation(s)
| | - Keith Mansfield
- Novartis Institutes for BioMedical Research Inc, 33413, Cambridge, Massachusetts, United States;
| | - Mark Milton
- Novartis Institutes for BioMedical Research Inc, 33413, Cambridge, Massachusetts, United States;
| | - Emily Meseck
- Novartis Pharmaceuticals Corp, 33412, East Hanover, New Jersey, United States;
| | - Kelley Penraat
- Novartis Institutes for BioMedical Research Inc, 33413, Cambridge, Massachusetts, United States;
| | - Deepa Chand
- Novartis Gene Therapies, Inc., Bannockburn, United States.,Washington University School of Medicine in Saint Louis, 12275, St Louis, Missouri, United States;
| | | |
Collapse
|
95
|
Hall BE, Macdonald E, Cassidy M, Yun S, Sapio MR, Ray P, Doty M, Nara P, Burton MD, Shiers S, Ray-Chaudhury A, Mannes AJ, Price TJ, Iadarola MJ, Kulkarni AB. Transcriptomic analysis of human sensory neurons in painful diabetic neuropathy reveals inflammation and neuronal loss. Sci Rep 2022; 12:4729. [PMID: 35304484 PMCID: PMC8933403 DOI: 10.1038/s41598-022-08100-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 02/25/2022] [Indexed: 01/02/2023] Open
Abstract
Pathological sensations caused by peripheral painful neuropathy occurring in Type 2 diabetes mellitus (T2DM) are often described as 'sharp' and 'burning' and are commonly spontaneous in origin. Proposed etiologies implicate dysfunction of nociceptive sensory neurons in dorsal root ganglia (DRG) induced by generation of reactive oxygen species, microvascular defects, and ongoing axonal degeneration and regeneration. To investigate the molecular mechanisms contributing to diabetic pain, DRGs were acquired postmortem from patients who had been experiencing painful diabetic peripheral neuropathy (DPN) and subjected to transcriptome analyses to identify genes contributing to pathological processes and neuropathic pain. DPN occurs in distal extremities resulting in the characteristic "glove and stocking" pattern. Accordingly, the L4 and L5 DRGs, which contain the perikarya of primary afferent neurons innervating the foot, were analyzed from five DPN patients and compared with seven controls. Transcriptome analyses identified 844 differentially expressed genes. We observed increases in levels of inflammation-associated transcripts from macrophages in DPN patients that may contribute to pain hypersensitivity and, conversely, there were frequent decreases in neuronally-related genes. The elevated inflammatory gene profile and the accompanying downregulation of multiple neuronal genes provide new insights into intraganglionic pathology and mechanisms causing neuropathic pain in DPN patients with T2DM.
Collapse
Affiliation(s)
- Bradford E Hall
- Functional Genomics Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Room 130, Bethesda, MD, 20892, USA
| | - Emma Macdonald
- Functional Genomics Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Room 130, Bethesda, MD, 20892, USA
- Present Affiliation: NIH Graduate Partnerships Program, Brown University, Providence, RI, 02912, USA
| | - Margaret Cassidy
- Functional Genomics Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Room 130, Bethesda, MD, 20892, USA
| | - Sijung Yun
- Yotta Biomed, LLC, Bethesda, MD, 20814, USA
| | - Matthew R Sapio
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Pradipta Ray
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Megan Doty
- Functional Genomics Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Room 130, Bethesda, MD, 20892, USA
| | - Pranavi Nara
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Michael D Burton
- Neuroimmunology and Behavior Group, School of Behavior and Brain Sciences, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Stephanie Shiers
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Abhik Ray-Chaudhury
- Surgical Neurology Branch, Disorders and Stroke, National Institute of Neurological, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Andrew J Mannes
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Theodore J Price
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Michael J Iadarola
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Ashok B Kulkarni
- Functional Genomics Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, 30 Convent Drive, Room 130, Bethesda, MD, 20892, USA.
| |
Collapse
|
96
|
Tavares-Ferreira D, Shiers S, Ray PR, Wangzhou A, Jeevakumar V, Sankaranarayanan I, Cervantes AM, Reese JC, Chamessian A, Copits BA, Dougherty PM, Gereau RW, Burton MD, Dussor G, Price TJ. Spatial transcriptomics of dorsal root ganglia identifies molecular signatures of human nociceptors. Sci Transl Med 2022; 14:eabj8186. [PMID: 35171654 PMCID: PMC9272153 DOI: 10.1126/scitranslmed.abj8186] [Citation(s) in RCA: 225] [Impact Index Per Article: 75.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Nociceptors are specialized sensory neurons that detect damaging or potentially damaging stimuli and are found in the dorsal root ganglia (DRG) and trigeminal ganglia. These neurons are critical for the generation of neuronal signals that ultimately create the perception of pain. Nociceptors are also primary targets for treating acute and chronic pain. Single-cell transcriptomics on mouse nociceptors has transformed our understanding of pain mechanisms. We sought to generate equivalent information for human nociceptors with the goal of identifying transcriptomic signatures of nociceptors, identifying species differences and potential drug targets. We used spatial transcriptomics to molecularly characterize transcriptomes of single DRG neurons from eight organ donors. We identified 12 clusters of human sensory neurons, 5 of which are C nociceptors, as well as 1 C low-threshold mechanoreceptors (LTMRs), 1 Aβ nociceptor, 2 Aδ, 2 Aβ, and 1 proprioceptor subtypes. By focusing on expression profiles for ion channels, G protein-coupled receptors (GPCRs), and other pharmacological targets, we provided a rich map of potential drug targets in the human DRG with direct comparison to mouse sensory neuron transcriptomes. We also compared human DRG neuronal subtypes to nonhuman primates showing conserved patterns of gene expression among many cell types but divergence among specific nociceptor subsets. Last, we identified sex differences in human DRG subpopulation transcriptomes, including a marked increase in calcitonin-related polypeptide alpha (CALCA) expression in female pruritogen receptor-enriched nociceptors. This comprehensive spatial characterization of human nociceptors might open the door to development of better treatments for acute and chronic pain disorders.
Collapse
Affiliation(s)
- Diana Tavares-Ferreira
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson TX 75080, USA.,Corresponding author: (T.J.P.); (D.T.-F.)
| | - Stephanie Shiers
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson TX 75080, USA
| | - Pradipta R. Ray
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson TX 75080, USA
| | - Andi Wangzhou
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson TX 75080, USA
| | - Vivekanand Jeevakumar
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson TX 75080, USA
| | - Ishwarya Sankaranarayanan
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson TX 75080, USA
| | | | | | - Alexander Chamessian
- Department of Anesthesiology, Washington University Pain Center, St. Louis, MO 63110, USA
| | - Bryan A. Copits
- Department of Anesthesiology, Washington University Pain Center, St. Louis, MO 63110, USA
| | - Patrick M. Dougherty
- Department of Pain Medicine, Division of Anesthesiology and Critical Care, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Robert W. Gereau
- Department of Anesthesiology, Washington University Pain Center, St. Louis, MO 63110, USA
| | - Michael D. Burton
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson TX 75080, USA
| | - Gregory Dussor
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson TX 75080, USA
| | - Theodore J. Price
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson TX 75080, USA.,Corresponding author: (T.J.P.); (D.T.-F.)
| |
Collapse
|
97
|
Tavares-Ferreira D, Shiers S, Ray PR, Wangzhou A, Jeevakumar V, Sankaranarayanan I, Cervantes AM, Reese JC, Chamessian A, Copits BA, Dougherty PM, Gereau RW, Burton MD, Dussor G, Price TJ. Spatial transcriptomics of dorsal root ganglia identifies molecular signatures of human nociceptors. Sci Transl Med 2022. [DOI: 10.1126/scitranslmed.abj8186\] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Nociceptors are specialized sensory neurons that detect damaging or potentially damaging stimuli and are found in the dorsal root ganglia (DRG) and trigeminal ganglia. These neurons are critical for the generation of neuronal signals that ultimately create the perception of pain. Nociceptors are also primary targets for treating acute and chronic pain. Single-cell transcriptomics on mouse nociceptors has transformed our understanding of pain mechanisms. We sought to generate equivalent information for human nociceptors with the goal of identifying transcriptomic signatures of nociceptors, identifying species differences and potential drug targets. We used spatial transcriptomics to molecularly characterize transcriptomes of single DRG neurons from eight organ donors. We identified 12 clusters of human sensory neurons, 5 of which are C nociceptors, as well as 1 C low-threshold mechanoreceptors (LTMRs), 1 Aβ nociceptor, 2 Aδ, 2 Aβ, and 1 proprioceptor subtypes. By focusing on expression profiles for ion channels, G protein–coupled receptors (GPCRs), and other pharmacological targets, we provided a rich map of potential drug targets in the human DRG with direct comparison to mouse sensory neuron transcriptomes. We also compared human DRG neuronal subtypes to nonhuman primates showing conserved patterns of gene expression among many cell types but divergence among specific nociceptor subsets. Last, we identified sex differences in human DRG subpopulation transcriptomes, including a marked increase in calcitonin-related polypeptide alpha (
CALCA
) expression in female pruritogen receptor–enriched nociceptors. This comprehensive spatial characterization of human nociceptors might open the door to development of better treatments for acute and chronic pain disorders.
Collapse
Affiliation(s)
- Diana Tavares-Ferreira
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson TX 75080, USA
| | - Stephanie Shiers
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson TX 75080, USA
| | - Pradipta R. Ray
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson TX 75080, USA
| | - Andi Wangzhou
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson TX 75080, USA
| | - Vivekanand Jeevakumar
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson TX 75080, USA
| | - Ishwarya Sankaranarayanan
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson TX 75080, USA
| | | | | | - Alexander Chamessian
- Department of Anesthesiology , Washington University Pain Center, St. Louis, MO 63110, USA
| | - Bryan A. Copits
- Department of Anesthesiology , Washington University Pain Center, St. Louis, MO 63110, USA
| | - Patrick M. Dougherty
- Department of Pain Medicine, Division of Anesthesiology and Critical Care, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Robert W. Gereau
- Department of Anesthesiology , Washington University Pain Center, St. Louis, MO 63110, USA
| | - Michael D. Burton
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson TX 75080, USA
| | - Gregory Dussor
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson TX 75080, USA
| | - Theodore J. Price
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson TX 75080, USA
| |
Collapse
|
98
|
Graham RD, Sankarasubramanian V, Lempka SF. Dorsal Root Ganglion Stimulation for Chronic Pain: Hypothesized Mechanisms of Action. THE JOURNAL OF PAIN 2022; 23:196-211. [PMID: 34425252 PMCID: PMC8943693 DOI: 10.1016/j.jpain.2021.07.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 06/28/2021] [Accepted: 07/20/2021] [Indexed: 02/03/2023]
Abstract
Dorsal root ganglion stimulation (DRGS) is a neuromodulation therapy for chronic pain that is refractory to conventional medical management. Currently, the mechanisms of action of DRGS-induced pain relief are unknown, precluding both our understanding of why DRGS fails to provide pain relief to some patients and the design of neurostimulation technologies that directly target these mechanisms to maximize pain relief in all patients. Due to the heterogeneity of sensory neurons in the dorsal root ganglion (DRG), the analgesic mechanisms could be attributed to the modulation of one or many cell types within the DRG and the numerous brain regions that process sensory information. Here, we summarize the leading hypotheses of the mechanisms of DRGS-induced analgesia, and propose areas of future study that will be vital to improving the clinical implementation of DRGS. PERSPECTIVE: This article synthesizes the evidence supporting the current hypotheses of the mechanisms of action of DRGS for chronic pain and suggests avenues for future interdisciplinary research which will be critical to fully elucidate the analgesic mechanisms of the therapy.
Collapse
Affiliation(s)
- Robert D. Graham
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States,Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, United States
| | - Vishwanath Sankarasubramanian
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States,Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, United States
| | - Scott F. Lempka
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States,Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, United States,Department of Anesthesiology, University of Michigan, Ann Arbor, MI 48109, United States,Corresponding author: Scott F. Lempka, PhD, Department of Biomedical Engineering, University of Michigan, 2800 Plymouth Road, NCRC 14-184, Ann Arbor, MI 48109-2800,
| |
Collapse
|
99
|
Körner J, Lampert A. Functional subgroups of rat and human sensory neurons: a systematic review of electrophysiological properties. Pflugers Arch 2022; 474:367-385. [PMID: 35031856 PMCID: PMC8924089 DOI: 10.1007/s00424-021-02656-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 11/23/2021] [Accepted: 12/14/2021] [Indexed: 11/15/2022]
Abstract
Sensory neurons are responsible for the generation and transmission of nociceptive signals from the periphery to the central nervous system. They encompass a broadly heterogeneous population of highly specialized neurons. The understanding of the molecular choreography of individual subpopulations is essential to understand physiological and pathological pain states. Recently, it became evident that species differences limit transferability of research findings between human and rodents in pain research. Thus, it is necessary to systematically compare and categorize the electrophysiological data gained from human and rodent dorsal root ganglia neurons (DRGs). In this systematic review, we condense the available electrophysiological data defining subidentities in human and rat DRGs. A systematic search on PUBMED yielded 30 studies on rat and 3 studies on human sensory neurons. Defined outcome parameters included current clamp, voltage clamp, cell morphology, pharmacological readouts, and immune reactivity parameters. We compare evidence gathered for outcome markers to define subgroups, offer electrophysiological parameters for the definition of neuronal subtypes, and give a framework for the transferability of electrophysiological findings between species. A semiquantitative analysis revealed that for rat DRGs, there is an overarching consensus between studies that C-fiber linked sensory neurons display a lower action potential threshold, higher input resistance, a larger action potential overshoot, and a longer afterhyperpolarization duration compared to other sensory neurons. They are also more likely to display an infliction point in the falling phase of the action potential. This systematic review points out the need of more electrophysiological studies on human sensory neurons.
Collapse
Affiliation(s)
- Jannis Körner
- Institute of Physiology, Uniklinik RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany.,Clinic of Anesthesiology, Uniklinik RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Angelika Lampert
- Institute of Physiology, Uniklinik RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany.
| |
Collapse
|
100
|
Emily MF, Agrawal L, Barzaghi P, Otsuki M, Terenzio M. Use of Microfluidics Chambers to Image Axonal transport in Adult Sensory Neurons. Methods Mol Biol 2022; 2431:271-288. [PMID: 35412282 DOI: 10.1007/978-1-0716-1990-2_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Transport of cargoes along axons is crucial for ensuring effective neuronal function and survival. Lysosomes, which are membrane-bound organelles responsible for the degradation of macromolecules, are among the many cargoes being transported. Compartmentalized systems that allow for the separation of the somatic compartment from the axonal network, are widely used in the field of neurobiology and in the study of axonal transport in particular. Among the various solutions available, microfluidics chambers that take advantage of fluidic separation between different compartments, have seen widespread adoption. Said chambers are made of polydimethylsiloxane (PDMS), a transparent, gas permeable compound, which is compatible with fluorescence microscopy, and have significantly positively impacted cellular neuroscience, drastically increasing our understanding of axonal peripheral signaling. Here we describe a two-layered microfluidics chamber, engineered to allow for the culture of adult sensory neurons. This device was designed to promote the proper placement of adult sensory neurons in the somatic chamber in proximity of the microgrooves. We detail the production of the master mold, how to fabricate and assemble the device and how to disaggregate and load the cells in it. In addition, we provide details on how to conduct and analyze an axonal transport experiment using a custom made script in MATLAB designed by our laboratory.
Collapse
Affiliation(s)
- Maria Fransiska Emily
- Molecular Neuroscience Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Lokesh Agrawal
- Molecular Neuroscience Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Paolo Barzaghi
- Imaging Section, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Miki Otsuki
- Molecular Neuroscience Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Marco Terenzio
- Molecular Neuroscience Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan.
| |
Collapse
|