1
|
Hartmannsberger B, Ben-Kraiem A, Kramer S, Guidolin C, Kazerani I, Doppler K, Thomas D, Gurke R, Sisignano M, Kalelkar PP, García AJ, Monje PV, Sammeth M, Nusrat A, Brack A, Krug SM, Sommer C, Rittner HL. TAM receptors mediate the Fpr2-driven pain resolution and fibrinolysis after nerve injury. Acta Neuropathol 2024; 149:1. [PMID: 39680199 DOI: 10.1007/s00401-024-02840-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 11/29/2024] [Accepted: 12/05/2024] [Indexed: 12/17/2024]
Abstract
Nerve injury causes neuropathic pain and multilevel nerve barrier disruption. Nerve barriers consist of perineurial, endothelial and myelin barriers. So far, it is unclear whether resealing nerve barriers fosters pain resolution and recovery. To this end, we analysed the nerve barrier property portfolio, pain behaviour battery and lipidomics for precursors of specialized pro-resolving meditators (SPMs) and their receptors in chronic constriction injury of the rat sciatic nerve to identify targets for pain resolution by resealing the selected nerve barriers. Of the three nerve barriers-perineurium, capillaries and myelin-only capillary tightness specifically against larger molecules, such as fibrinogen, recuperated with pain resolution. Fibrinogen immunoreactivity was elevated in rats not only at the time of neuropathic pain but also in nerve biopsies from patients with (but not without) painful polyneuropathy, indicating that sealing of the vascular barrier might be a novel approach in pain treatment. Hydroxyeicosatetraenoic acid (15R-HETE), a precursor of aspirin-triggered lipoxin A4, was specifically upregulated at the beginning of pain resolution. Repeated local application of resolvin D1-laden nanoparticles or Fpr2 agonists sex-independently resulted in accelerated pain resolution and fibrinogen removal. Clearing macrophages (Cd206) were boosted and fibrinolytic pathways (Plat) were induced, while inflammation (Tnfα) and inflammasomes (Nlrp3) were unaffected by this treatment. Blocking TAM receptors (Tyro3, Axl and Mer) and tyrosine kinase receptors linking haemostasis and inflammation completely inhibited all the effects. In summary, nanoparticles can be used as transporters for fleeting lipids, such as SPMs, and therefore expand the array of possible therapeutic agents. Thus, the Fpr2-Cd206-TAM receptor axis may be a suitable target for strengthening the capillary barrier, removing endoneurial fibrinogen and boosting pain resolution in patients with chronic neuropathic pain.
Collapse
Affiliation(s)
- Beate Hartmannsberger
- Centre for Interdisciplinary Pain Medicine, Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Adel Ben-Kraiem
- Centre for Interdisciplinary Pain Medicine, Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Würzburg, Würzburg, Germany
- Helmholtz Institute for Metabolic, Obesity and Vascular Research, Diet-Induced Metabolic Alterations Group, Leipzig, Germany
| | - Sofia Kramer
- Centre for Interdisciplinary Pain Medicine, Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Carolina Guidolin
- Centre for Interdisciplinary Pain Medicine, Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Ida Kazerani
- Centre for Interdisciplinary Pain Medicine, Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Kathrin Doppler
- Department of Neurology, University Hospital Würzburg, Würzburg, Germany
| | - Dominique Thomas
- Goethe University, Frankfurt, Faculty of Medicine, Institute of Clinical Pharmacology, Frankfurt Am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence of Immune Mediate Diseases CIMD, Frankfurt Am Main, Germany
| | - Robert Gurke
- Goethe University, Frankfurt, Faculty of Medicine, Institute of Clinical Pharmacology, Frankfurt Am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence of Immune Mediate Diseases CIMD, Frankfurt Am Main, Germany
| | - Marco Sisignano
- Goethe University, Frankfurt, Faculty of Medicine, Institute of Clinical Pharmacology, Frankfurt Am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence of Immune Mediate Diseases CIMD, Frankfurt Am Main, Germany
| | - Pranav P Kalelkar
- George W. Woodruff School of Mechanical Engineering, Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, USA
| | - Andrés J García
- George W. Woodruff School of Mechanical Engineering, Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, USA
| | - Paula V Monje
- Department of Neurosurgery, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Michael Sammeth
- Department of Applied Sciences and Health, Coburg University of Applied Sciences and Art, Coburg, Germany
- Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Asma Nusrat
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Alexander Brack
- Centre for Interdisciplinary Pain Medicine, Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Susanne M Krug
- Charité-Universitätsmedizin Berlin, Clinical Physiology/Nutritional Medicine, Berlin, Germany
| | - Claudia Sommer
- Department of Neurology, University Hospital Würzburg, Würzburg, Germany
| | - Heike L Rittner
- Centre for Interdisciplinary Pain Medicine, Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Würzburg, Würzburg, Germany.
| |
Collapse
|
2
|
Neuman K, Zhang X, Lejeune BT, Pizzarella D, Vázquez M, Lewis LH, Koppes AN, Koppes RA. Static Magnetic Stimulation and Magnetic Microwires Synergistically Enhance and Guide Neurite Outgrowth. Adv Healthc Mater 2024:e2403956. [PMID: 39568232 DOI: 10.1002/adhm.202403956] [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: 10/11/2024] [Indexed: 11/22/2024]
Abstract
Axonal growth is heavily influenced by topography and biophysical stimuli including magnetic and electrical fields. Despite extensive investigation, the degree of influence and the underlying genetic mechanisms remain poorly understood. Here, a novel approach to guide neurite growth is undertaken using an innovative ferromagnetic composite material - glass-coated magnetic microwire - to furnish a synergistic combination of magnetic and topographical cues. Whole rat dorsal root ganglia (DRG) are cultured under five different conditions: control, static magnetic field, magnetic microwire, static magnetic field + glass fiber, and static magnetic field + magnetic microwire. DRG outgrowth responses under each condition, including total neurite outgrowth and directionality, are compared. The combination of both magnetic stimulation and topography significantly increases total neurite outgrowth compared to the controls. The combination of magnetic stimulation and magnetic microwire lead to a strong directional bias of growth along the microwire, double what is observed with the glass fiber. Next generation RNA sequencing of DRG exposed to static magnetic field + magnetic microwire reveals the downregulation of genes relating to the immune response, interleukin signaling, and signal transduction. These results set the stage for contemplating future biophysical stimulation for axonal guidance and improved understanding of material-tissue interactions.
Collapse
Affiliation(s)
- Katelyn Neuman
- Dept. of Chemical Engineering, Northeastern University, Boston, MA, 02115, USA
| | - Xiaoyu Zhang
- Dept. of Mechanical and Industrial Engineering, Northeastern University, Boston, MA, 02115, USA
| | - Brian T Lejeune
- Dept. of Chemical Engineering, Northeastern University, Boston, MA, 02115, USA
| | - Dominic Pizzarella
- Dept. of Chemical Engineering, Northeastern University, Boston, MA, 02115, USA
| | - Manuel Vázquez
- Instituto de Ciencia de Materiales de Madrid, CSIC, Madrid, 28049, Spain
| | - Laura H Lewis
- Dept. of Chemical Engineering, Northeastern University, Boston, MA, 02115, USA
- Dept. of Mechanical and Industrial Engineering, Northeastern University, Boston, MA, 02115, USA
| | - Abigail N Koppes
- Dept. of Chemical Engineering, Northeastern University, Boston, MA, 02115, USA
- Dept. of Bioengineering, Northeastern University, Boston, MA, 02115, USA
- Dept. of Biology, Northeastern University, Boston, MA, 02115, USA
| | - Ryan A Koppes
- Dept. of Chemical Engineering, Northeastern University, Boston, MA, 02115, USA
| |
Collapse
|
3
|
Aparicio GI, Quintero JE, Plum L, Deng L, Wanczyk K, Henry M, Lynch E, Murphy M, Gerhardt GA, van Horne CG, Monje PV. Identification of cellular and noncellular components of mature intact human peripheral nerve. J Peripher Nerv Syst 2024; 29:294-314. [PMID: 38973168 DOI: 10.1111/jns.12643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/09/2024] [Accepted: 06/14/2024] [Indexed: 07/09/2024]
Abstract
BACKGROUND AND AIMS The goal of this study was to define basic constituents of the adult peripheral nervous system (PNS) using intact human nerve tissues. METHODS We combined fluorescent and chromogenic immunostaining methods, myelin-selective fluorophores, and routine histological stains to identify common cellular and noncellular elements in aldehyde-fixed nerve tissue sections. We employed Schwann cell (SC)-specific markers, such as S100β, NGFR, Sox10, and myelin protein zero (MPZ), together with axonal, extracellular matrix (collagen IV, laminin, fibronectin), and fibroblast markers to assess the SC's relationship to myelin sheaths, axons, other cell types, and the acellular environment. RESULTS Whereas S100β and Sox10 revealed mature SCs in the absence of other stains, discrimination between myelinating and non-myelinating (Remak) SCs required immunodetection of NGFR along with axonal and/or myelin markers. Surprisingly, our analysis of NGFR+ profiles uncovered the existence of at least 3 different novel populations of NGFR+/S100β- cells, herein referred to as nonglial cells, residing in the stroma and perivascular areas of all nerve compartments. An important proportion of the nerve's cellular content, including circa 30% of endoneurial cells, consisted of heterogenous S100β negative cells that were not associated with axons. Useful markers to identify the localization and diversity of nonglial cell types across different compartments were Thy1, CD34, SMA, and Glut1, a perineurial cell marker. INTERPRETATION Our optimized methods revealed additional detailed information to update our understanding of the complexity and spatial orientation of PNS-resident cell types in humans.
Collapse
Affiliation(s)
- Gabriela I Aparicio
- Department of Neurosurgery, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
- Neurorestoration Center, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Jorge E Quintero
- Department of Neurosurgery, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
- Neurorestoration Center, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Lauren Plum
- Department of Neurosurgery, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
- Neurorestoration Center, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Lingxiao Deng
- Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Kristen Wanczyk
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Miriam Henry
- Department of Plastic Surgery, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Evan Lynch
- Department of Plastic Surgery, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Michael Murphy
- Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Greg A Gerhardt
- Department of Neurosurgery, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
- Neurorestoration Center, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
- Department of Plastic Surgery, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky, USA
| | - Craig G van Horne
- Department of Neurosurgery, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
- Neurorestoration Center, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Paula V Monje
- Department of Neurosurgery, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
- Neurorestoration Center, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
4
|
Su Y, Huang M, Thomas AG, Maragakis J, Huizar KDJ, Zheng Y, Wu Y, Farah MH, Slusher BS. GCPII Inhibition Promotes Remyelination after Peripheral Nerve Injury in Aged Mice. Int J Mol Sci 2024; 25:6893. [PMID: 39000003 PMCID: PMC11241013 DOI: 10.3390/ijms25136893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 06/18/2024] [Accepted: 06/21/2024] [Indexed: 07/14/2024] Open
Abstract
Peripheral nerve injuries (PNIs) represent a significant clinical challenge, particularly in elderly populations where axonal remyelination and regeneration are impaired. Developing therapies to enhance these processes is crucial for improving PNI repair outcomes. Glutamate carboxypeptidase II (GCPII) is a neuropeptidase that plays a pivotal role in modulating glutamate signaling through its enzymatic cleavage of the abundant neuropeptide N-acetyl aspartyl glutamate (NAAG) to liberate glutamate. Within the PNS, GCPII is expressed in Schwann cells and activated macrophages, and its expression is amplified with aging. In this study, we explored the therapeutic potential of inhibiting GCPII activity following PNI. We report significant GCPII protein and activity upregulation following PNI, which was normalized by the potent and selective GCPII inhibitor 2-(phosphonomethyl)-pentanedioic acid (2-PMPA). In vitro, 2-PMPA robustly enhanced myelination in dorsal root ganglion (DRG) explants. In vivo, using a sciatic nerve crush injury model in aged mice, 2-PMPA accelerated remyelination, as evidenced by increased myelin sheath thickness and higher numbers of remyelinated axons. These findings suggest that GCPII inhibition may be a promising therapeutic strategy to enhance remyelination and potentially improve functional recovery after PNI, which is especially relevant in elderly PNI patients where this process is compromised.
Collapse
Affiliation(s)
- Yu Su
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (Y.S.); (M.H.); (A.G.T.); (J.M.); (K.D.J.H.); (Y.Z.); (Y.W.)
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Meixiang Huang
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (Y.S.); (M.H.); (A.G.T.); (J.M.); (K.D.J.H.); (Y.Z.); (Y.W.)
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ajit G. Thomas
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (Y.S.); (M.H.); (A.G.T.); (J.M.); (K.D.J.H.); (Y.Z.); (Y.W.)
| | - John Maragakis
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (Y.S.); (M.H.); (A.G.T.); (J.M.); (K.D.J.H.); (Y.Z.); (Y.W.)
| | - Kaitlyn D. J. Huizar
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (Y.S.); (M.H.); (A.G.T.); (J.M.); (K.D.J.H.); (Y.Z.); (Y.W.)
| | - Yuxin Zheng
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (Y.S.); (M.H.); (A.G.T.); (J.M.); (K.D.J.H.); (Y.Z.); (Y.W.)
| | - Ying Wu
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (Y.S.); (M.H.); (A.G.T.); (J.M.); (K.D.J.H.); (Y.Z.); (Y.W.)
| | - Mohamed H. Farah
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Barbara S. Slusher
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (Y.S.); (M.H.); (A.G.T.); (J.M.); (K.D.J.H.); (Y.Z.); (Y.W.)
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Psychiatry and Behavioral Science, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
5
|
Trambitas C, Pap T, Niculescu R, Popelea MC, Cotoi OS, Cordoș B, Domnariu HP, Marin A, Feier AM, David C, Vintila C. Biocompatible 3D-Printed Devices With Adipose Stem Cells in the Regenerative Process of Sciatic Nerve Lesions in Rodent Models: An Experimental Study. Cureus 2024; 16:e62412. [PMID: 39011200 PMCID: PMC11248491 DOI: 10.7759/cureus.62412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/14/2024] [Indexed: 07/17/2024] Open
Abstract
INTRODUCTION Peripheral nerve injuries are a significant clinical challenge. The rat sciatic nerve serves as an ideal model for studying nerve regeneration. Extensive research has been conducted to unravel the intricate mechanisms involved in peripheral nerve regeneration, aiming to develop effective therapeutic strategies for nerve injury patients. Research including different types of materials that can be used as nerve guides like synthetic polymers have been investigated for their biocompatibility and molding properties. Among multiple stem cell types, adipose-derived stem cells (ASCs), bone marrow-derived mesenchymal stem cells (BM-MSCs), and induced pluripotent stem cells (iPSCs) have shown neuroprotective and regenerative important properties. METHODS The purposes of our study were to develop a protocol for rat sciatic nerve injury treated with 3D-printed guide and adipose stem cells to investigate nerve regeneration through histologic examination and biomechanical characteristics of muscular tissue. We use 20 (100%) male Wistar rats, measuring between 350 g ± 35 g, who underwent complete transection of the right sciatic nerve, resulting in a 1 cm defect. The group was separated into three subgroups: the first subgroup (n = 8) was treated with a 3D-printed guide with adipose stem cells, the second subgroup (n = 8) was treated with a 3D-printed guide without adipose stem cells, and the third subgroup (n = 4) was the control group. At four, eight, and 12 weeks, we measured with ultrasonography the grade of muscular atrophy. At 12 weeks, we harvested the sciatic nerve and performed a histological examination and mechanical investigation of the tibialis anterior muscle. RESULTS On the examined specimen of the first subgroup, cross-sectioned nerve structures were present, surrounded by a mature fibro-adipose connective tissue, with blood vessels. In the second subgroup, no nerve structure was observed on the examined sections, but in the polymorphic inflammatory infiltrate and control group, no signs of regeneration were found. CONCLUSIONS The present study shows a promising potential when utilizing adipose stem cell-based therapies for promoting peripheral nerve regeneration following large (>1 cm) nerve defects knowing that at this size, regeneration is impossible with known treatments.
Collapse
Affiliation(s)
- Cristian Trambitas
- Plastic and Reconstructive Surgery, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Targu Mures, ROU
| | - Timea Pap
- Plastic and Reconstructive Surgery, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Targu Mures, ROU
| | - Raluca Niculescu
- Pathology and Laboratory Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Targu Mures, ROU
| | - Maria Catalina Popelea
- Pathology and Laboratory Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Targu Mures, ROU
| | - Ovidiu S Cotoi
- Physiopathology, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Targu Mures, ROU
| | - Bogdan Cordoș
- Center of Experimental and Imaging Studies, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Targu Mures, ROU
| | - Horatiu-Paul Domnariu
- Plastic and Reconstructive Surgery, University of Oradea, Lucian Blaga University of Sibiu, Oradea, ROU
| | - Andrei Marin
- Plastic and Reconstructive Surgery, Carol Davila University of Medicine and Pharmacy, Bucuresti, ROU
| | - Andrei Marian Feier
- Orthopaedics, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Targu Mures, ROU
| | - Camelia David
- Plastic and Reconstructive Surgery, Emergency County Hospital Targu Mures, Targu Mures, ROU
| | - Cristian Vintila
- Plastic and Reconstructive Surgery, Emergency County Hospital Targu Mures, Targu Mures, ROU
| |
Collapse
|
6
|
Zhang H, Zhang Z, Lin H. Research progress on the reduced neural repair ability of aging Schwann cells. Front Cell Neurosci 2023; 17:1228282. [PMID: 37545880 PMCID: PMC10398339 DOI: 10.3389/fncel.2023.1228282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 07/06/2023] [Indexed: 08/08/2023] Open
Abstract
Peripheral nerve injury (PNI) is associated with delayed repair of the injured nerves in elderly patients, resulting in loss of nerve function, chronic pain, muscle atrophy, and permanent disability. Therefore, the mechanism underlying the delayed repair of peripheral nerves in aging patients should be investigated. Schwann cells (SCs) play a crucial role in repairing PNI and regulating various nerve-repair genes after injury. SCs also promote peripheral nerve repair through various modalities, including mediating nerve demyelination, secreting neurotrophic factors, establishing Büngner bands, clearing axon and myelin debris, and promoting axon remyelination. However, aged SCs undergo structural and functional changes, leading to demyelination and dedifferentiation disorders, decreased secretion of neurotrophic factors, impaired clearance of axonal and myelin debris, and reduced capacity for axon remyelination. As a result, aged SCs may result in delayed repair of nerves after injury. This review article aimed to examine the mechanism underlying the diminished neural repair ability of aging SCs.
Collapse
|
7
|
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
|
8
|
Gale JR, Gedeon JY, Donnelly CJ, Gold MS. Local translation in primary afferents and its contribution to pain. Pain 2022; 163:2302-2314. [PMID: 35438669 PMCID: PMC9579217 DOI: 10.1097/j.pain.0000000000002658] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 04/08/2022] [Indexed: 02/06/2023]
Abstract
ABSTRACT Chronic pain remains a significant problem due to its prevalence, impact, and limited therapeutic options. Progress in addressing chronic pain is dependent on a better understanding of underlying mechanisms. Although the available evidence suggests that changes within the central nervous system contribute to the initiation and maintenance of chronic pain, it also suggests that the primary afferent plays a critical role in all phases of the manifestation of chronic pain in most of those who suffer. Most notable among the changes in primary afferents is an increase in excitability or sensitization. A number of mechanisms have been identified that contribute to primary afferent sensitization with evidence for both increases in pronociceptive signaling molecules, such as voltage-gated sodium channels, and decreases in antinociceptive signaling molecules, such as voltage-dependent or calcium-dependent potassium channels. Furthermore, these changes in signaling molecules seem to reflect changes in gene expression as well as posttranslational processing. A mechanism of sensitization that has received far less attention, however, is the local or axonal translation of these signaling molecules. A growing body of evidence indicates that this process not only is dynamically regulated but also contributes to the initiation and maintenance of chronic pain. Here, we review the biology of local translation in primary afferents and its relevance to pain pathobiology.
Collapse
Affiliation(s)
- Jenna R Gale
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| | - Jeremy Y Gedeon
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| | | | - Michael S Gold
- Corresponding author: Michael S Gold, PhD, Department of Neurobiology, University of Pittsburgh School of Medicine, 200 Lothrop Street, Pittsburgh, PA 15213, P: 412-383-5367,
| |
Collapse
|
9
|
Berner J, Weiss T, Sorger H, Rifatbegovic F, Kauer M, Windhager R, Dohnal A, Ambros PF, Ambros IM, Boztug K, Steinberger P, Taschner‐Mandl S. Human repair-related Schwann cells adopt functions of antigen-presenting cells in vitro. Glia 2022; 70:2361-2377. [PMID: 36054432 PMCID: PMC9804420 DOI: 10.1002/glia.24257] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 07/16/2022] [Accepted: 07/25/2022] [Indexed: 01/05/2023]
Abstract
The plastic potential of Schwann cells (SCs) is increasingly recognized to play a role after nerve injury and in diseases of the peripheral nervous system. Reports on the interaction between immune cells and SCs indicate their involvement in inflammatory processes. However, the immunocompetence of human SCs has been primarily deduced from neuropathies, but whether after nerve injury SCs directly regulate an adaptive immune response is unknown. Here, we performed comprehensive analysis of immunomodulatory capacities of human repair-related SCs (hrSCs), which recapitulate SC response to nerve injury in vitro. We used our well-established culture model of primary hrSCs from human peripheral nerves and analyzed the transcriptome, secretome, and cell surface proteins for pathways and markers relevant in innate and adaptive immunity, performed phagocytosis assays, and monitored T-cell subset activation in allogeneic co-cultures. Our findings show that hrSCs are phagocytic, which is in line with high MHCII expression. Furthermore, hrSCs express co-regulatory proteins, such as CD40, CD80, B7H3, CD58, CD86, and HVEM, release a plethora of chemoattractants, matrix remodeling proteins and pro- as well as anti-inflammatory cytokines, and upregulate the T-cell inhibiting PD-L1 molecule upon pro-inflammatory stimulation with IFNγ. In contrast to monocytes, hrSC alone are not sufficient to trigger allogenic CD4+ and CD8+ T-cells, but limit number and activation status of exogenously activated T-cells. This study demonstrates that hrSCs possess features and functions typical for professional antigen-presenting cells in vitro, and suggest a new role of these cells as negative regulators of T-cell immunity during nerve regeneration.
Collapse
Affiliation(s)
- Jakob Berner
- St. Anna Children's Cancer Research Institute (CCRI)ViennaAustria
- St. Anna Children's HospitalViennaAustria
| | - Tamara Weiss
- St. Anna Children's Cancer Research Institute (CCRI)ViennaAustria
- Department of Plastic, Reconstructive and Aesthetic SurgeryMedical University of Vienna
| | - Helena Sorger
- St. Anna Children's Cancer Research Institute (CCRI)ViennaAustria
| | | | - Max Kauer
- St. Anna Children's Cancer Research Institute (CCRI)ViennaAustria
| | - Reinhard Windhager
- Department of Orthopedics and Trauma SurgeryMedical University of ViennaViennaAustria
| | - Alexander Dohnal
- St. Anna Children's Cancer Research Institute (CCRI)ViennaAustria
| | - Peter F. Ambros
- St. Anna Children's Cancer Research Institute (CCRI)ViennaAustria
| | - Inge M. Ambros
- St. Anna Children's Cancer Research Institute (CCRI)ViennaAustria
| | - Kaan Boztug
- St. Anna Children's Cancer Research Institute (CCRI)ViennaAustria
- St. Anna Children's HospitalViennaAustria
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases (LBI‐RUD)ViennaAustria
- Center for Molecular Medicine (CeMM)ViennaAustria
| | | | | |
Collapse
|
10
|
Chau MJ, Quintero JE, Blalock E, Byrum S, Mackintosh SG, Samaan C, Gerhardt GA, van Horne CG. Transection injury differentially alters the proteome of the human sural nerve. PLoS One 2022; 17:e0260998. [PMID: 36417411 PMCID: PMC9683555 DOI: 10.1371/journal.pone.0260998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 09/09/2022] [Indexed: 11/25/2022] Open
Abstract
Regeneration after severe peripheral nerve injury is often poor. Knowledge of human nerve regeneration and the growth microenvironment is greatly lacking. We aimed to identify the regenerative proteins in human peripheral nerve by comparing the proteome before and after a transection injury. In a unique study design, we collected closely matched samples of naïve and injured sural nerve. Naïve and injured (two weeks after injury) samples were analyzed using mass spectrometry and immunoassays. We found significantly altered levels following the nerve injury. Mass spectrometry revealed that injury samples had 568 proteins significantly upregulated and 471 significantly downregulated compared to naïve samples (q-value ≤ 0.05 and Z ≥ |2| (log2)). We used Gene Ontology (GO) pathway overrepresentation analysis to highlight groups of proteins that were significantly upregulated or downregulated with injury-induced degeneration and regeneration. Significant protein changes in key pathways were identified including growth factor levels, Schwann cell de-differentiation, myelination downregulation, epithelial-mesenchymal transition (EMT), and axonal regeneration pathways. The proteomes of the uninjured nerve compared to the degenerating/regenerating nerve may reveal biomarkers to aid in the development of repair strategies such as infusing supplemental trophic factors and in monitoring neural tissue regeneration.
Collapse
Affiliation(s)
- Monica J. Chau
- Brain Restoration Center, College of Medicine, University of Kentucky, Lexington, KY, United States of America
- Department of Neurosurgery, College of Medicine, University of Kentucky, Lexington, KY, United States of America
| | - Jorge E. Quintero
- Brain Restoration Center, College of Medicine, University of Kentucky, Lexington, KY, United States of America
- Department of Neurosurgery, College of Medicine, University of Kentucky, Lexington, KY, United States of America
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, United States of America
| | - Eric Blalock
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY, United States of America
| | - Stephanie Byrum
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, United States of America
| | - Samuel G. Mackintosh
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, United States of America
| | - Christopher Samaan
- Brain Restoration Center, College of Medicine, University of Kentucky, Lexington, KY, United States of America
- Department of Neurosurgery, College of Medicine, University of Kentucky, Lexington, KY, United States of America
| | - Greg A. Gerhardt
- Brain Restoration Center, College of Medicine, University of Kentucky, Lexington, KY, United States of America
- Department of Neurosurgery, College of Medicine, University of Kentucky, Lexington, KY, United States of America
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, United States of America
- Department of Neurology, College of Medicine, University of Kentucky, Lexington, KY, United States of America
| | - Craig G. van Horne
- Brain Restoration Center, College of Medicine, University of Kentucky, Lexington, KY, United States of America
- Department of Neurosurgery, College of Medicine, University of Kentucky, Lexington, KY, United States of America
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, United States of America
- * E-mail:
| |
Collapse
|
11
|
Li WY, Li ZG, Fu XM, Wang XY, Lv ZX, Sun P, Zhu XF, Wang Y. Transgenic Schwann cells overexpressing POU6F1 promote sciatic nerve regeneration within acellular nerve allografts. J Neural Eng 2022; 19. [PMID: 36317259 DOI: 10.1088/1741-2552/ac9e1e] [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: 07/13/2022] [Accepted: 10/27/2022] [Indexed: 11/11/2022]
Abstract
Objective.Acellular nerve allograft (ANA) is an effective surgical approach used to bridge the sciatic nerve gap. The molecular regulators of post-surgical recovery are not well-known. Here, we explored the effect of transgenic Schwann cells (SCs) overexpressing POU domain class 6, transcription factor 1 (POU6F1) on sciatic nerve regeneration within ANAs. We explored the functions of POU6F1 in nerve regeneration by using a cell model of H2O2-induced SCs injury and transplanting SCs overexpressing POU6F1 into ANA to repair sciatic nerve gaps.Approach.Using RNA-seq, Protein-Protein Interaction network analysis, gene ontology enrichment, and Kyoto Encyclopedia of Genes and Genomes pathway analysis, we identified a highly and differentially expressed transcription factor, POU6F1, following ANA treatment of sciatic nerve gap. Expressing a high degree of connectivity, POU6F1 was predicted to play a role in peripheral nervous system myelination.Main results.To test the role of POU6F1 in nerve regeneration after ANA, we infected SCs with adeno-associated virus-POU6F1, demonstrating that POU6F1 overexpression promotes proliferation, anti-apoptosis, and migration of SCsin vitro. We also found that POU6F1 significantly upregulated JNK1/2 and c-Jun phosphorylation and that selective JNK1/2 inhibition attenuated the effects of POU6F1 on proliferation, survival, migration, and JNK1/2 and c-Jun phosphorylation. The direct interaction of POU6F1 and activated JNK1/2 was subsequently confirmed by co-immunoprecipitation. In rat sciatic nerve injury model with a 10 mm gap, we confirmed the pattern of POU6F1 upregulation and co-localization with transplanted SCs. ANAs loaded with POU6F1-overexpressing SCs demonstrated the enhanced survival of transplanted SCs, axonal regeneration, myelination, and functional motor recovery compared to the ANA group loaded by SCs-only in line within vitrofindings.Significance.This study identifies POU6F1 as a novel regulator of post-injury sciatic nerve repair, acting through JNK/c-Jun signaling in SCs to optimize therapeutic outcomes in the ANA surgical approach.
Collapse
Affiliation(s)
- Wen-Yuan Li
- Institute of Neural Tissue Engineering, Mudanjiang Medical University, Mudanjiang, Heilongjiang Province, People's Republic of China
| | - Zhi-Gang Li
- The Second Department of General Surgery, Hongqi Hospital, Mudanjiang Medical University, Mudanjiang, Heilongjiang Province, People's Republic of China
| | - Xiu-Mei Fu
- Department of Anatomy, College of Basic Medical Sciences, Chengde Medical College, Chengde 067000, People's Republic of China.,Hebei Key Laboratory of Nerve Injury and Repair, Chengde 067000, People's Republic of China
| | - Xiao-Yu Wang
- Institute of Neural Tissue Engineering, Mudanjiang Medical University, Mudanjiang, Heilongjiang Province, People's Republic of China
| | - Zhong-Xiao Lv
- Institute of Neural Tissue Engineering, Mudanjiang Medical University, Mudanjiang, Heilongjiang Province, People's Republic of China
| | - Ping Sun
- Institute of Neural Tissue Engineering, Mudanjiang Medical University, Mudanjiang, Heilongjiang Province, People's Republic of China
| | - Xiao-Feng Zhu
- Institute of Neural Tissue Engineering, Mudanjiang Medical University, Mudanjiang, Heilongjiang Province, People's Republic of China
| | - Ying Wang
- Institute of Neural Tissue Engineering, Mudanjiang Medical University, Mudanjiang, Heilongjiang Province, People's Republic of China
| |
Collapse
|
12
|
Membrane Progesterone Receptor α (mPRα/PAQR7) Promotes Survival and Neurite Outgrowth of Human Neuronal Cells by a Direct Action and Through Schwann Cell-like Stem Cells. J Mol Neurosci 2022; 72:2067-2080. [PMID: 35974286 DOI: 10.1007/s12031-022-02057-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 08/05/2022] [Indexed: 10/15/2022]
Abstract
We recently showed that membrane progesterone receptor α (mPRα/PAQR7) promotes pro-regenerative effects in Schwann cell-like adipose stem cells (SCL-ASC), an alternative model to Schwann cells for the promotion of peripheral nerve regeneration. In this study, we investigated how mPRα activation with the mPR-specific agonist Org OD 02-0 in SCL-ASC affected regenerative parameters in two neuronal cell lines, IMR-32 and SH-SY-5Y. In a series of conditioned medium experiments, we found that mPR activation of SCL-ASC led to increased neurite outgrowth, protection from cell death and increased expression of peripheral nerve regeneration markers (CREB3, ATF3, GAP43) in neuronal cell lines. These effects were stronger than the ones observed with the conditioned medium from untreated SCL-ASC. The addition of Org OD 02-0 to the untreated cell medium mimicked the effects of mPR activation of SCL-ASC on cell death, but not on neurite outgrowth. Therefore, the effect of Org OD 02-0 on neurite outgrowth is SCL-ASC-dependent, while its effect on cell survivability is likely due to the direct activation of mPRs on neuronal cells. SCL-ASC transfection with mPRα siRNA showed that this isoform is responsible for the beneficial effect on neurite outgrowth. Further experiments showed that SCL-ASC-dependent outcomes likely involved the release of BDNF and IGF-2 from these cells. The beneficial mPRα effect on neurite outgrowth was confirmed in co-culture conditions. These findings strengthen the hypothesis that mPRα could play a pro-regenerative role in SCL-ASC and be a therapeutic target for the promotion of peripheral nerve regeneration.
Collapse
|
13
|
An RNA-sequencing transcriptome of the rodent Schwann cell response to peripheral nerve injury. J Neuroinflammation 2022; 19:105. [PMID: 35501870 PMCID: PMC9063194 DOI: 10.1186/s12974-022-02462-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 04/13/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The important contribution of glia to mechanisms of injury and repair of the nervous system is increasingly recognized. In stark contrast to the central nervous system (CNS), the peripheral nervous system (PNS) has a remarkable capacity for regeneration after injury. Schwann cells are recognized as key contributors to PNS regeneration, but the molecular underpinnings of the Schwann cell response to injury and how they interact with the inflammatory response remain incompletely understood. METHODS We completed bulk RNA-sequencing of Schwann cells purified acutely using immunopanning from the naïve and injured rodent sciatic nerve at 3, 5, and 7 days post-injury. We used qRT-PCR and in situ hybridization to assess cell purity and probe dataset integrity. Finally, we used bioinformatic analysis to probe Schwann cell-specific injury-induced modulation of cellular pathways. RESULTS Our data confirm Schwann cell purity and validate RNAseq dataset integrity. Bioinformatic analysis identifies discrete modules of genes that follow distinct patterns of regulation in the 1st days after injury and their corresponding molecular pathways. These findings enable improved differentiation of myeloid and glial components of neuroinflammation after peripheral nerve injury and highlight novel molecular aspects of the Schwann cell injury response such as acute downregulation of the AGE/RAGE pathway and of secreted molecules Sparcl1 and Sema5a. CONCLUSIONS We provide a helpful resource for further deciphering the Schwann cell injury response and a depth of transcriptional data that can complement the findings of recent single cell sequencing approaches. As more data become available on the response of CNS glia to injury, we anticipate that this dataset will provide a valuable platform for understanding key differences in the PNS and CNS glial responses to injury and for designing approaches to ameliorate CNS regeneration.
Collapse
|
14
|
Chau MJ, Quintero JE, Monje PV, Voss SR, Welleford AS, Gerhardt GA, van Horne CG. Using a Transection Paradigm to Enhance the Repair Mechanisms of an Investigational Human Cell Therapy. Cell Transplant 2022; 31:9636897221123515. [PMID: 36169034 PMCID: PMC9523845 DOI: 10.1177/09636897221123515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 08/16/2022] [Indexed: 12/02/2022] Open
Abstract
One promising strategy in cell therapies for Parkinson's disease (PD) is to harness a patient's own cells to provide neuroprotection in areas of the brain affected by neurodegeneration. No treatment exists to replace cells in the brain. Thus, our goal has been to support sick neurons and slow neurodegeneration by transplanting living repair tissue from the peripheral nervous system into the substantia nigra of those with PD. Our group has pioneered the transplantation of transection-activated sural nerve fascicles into the brain of human subjects with PD. Our experience in sural nerve transplantation has supported the safety and feasibility of this approach. As part of a paradigm to assess the reparative properties of human sural nerve following a transection injury, we collected nerve tissue approximately 2 weeks after sural nerve transection for immunoassays from 15 participants, and collected samples from two additional participants for single nuclei RNA sequencing. We quantified the expression of key neuroprotective and select anti-apoptotic genes along with their corresponding protein levels using immunoassays. The single nuclei data clustered into 10 distinctive groups defined on the basis of previously published cell type-specific genes. Transection-induced reparative peripheral nerve tissue showed RNA expression of neuroprotective factors and anti-apoptotic factors across multiple cell types after nerve injury induction. Key proteins of interest (BDNF, GDNF, beta-NGF, PDGFB, and VEGF) were upregulated in reparative tissue. These results provide insight on this repair tissue's utility as a neuroprotective cell therapy.
Collapse
Affiliation(s)
- Monica J. Chau
- Brain Restoration Center, College of Medicine, University of Kentucky, Lexington, KY, USA
- Department of Neurosurgery, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Jorge E. Quintero
- Brain Restoration Center, College of Medicine, University of Kentucky, Lexington, KY, USA
- Department of Neurosurgery, College of Medicine, University of Kentucky, Lexington, KY, USA
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Paula V. Monje
- Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Stephen Randal Voss
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Andrew S. Welleford
- Department of Neurology, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Greg A. Gerhardt
- Brain Restoration Center, College of Medicine, University of Kentucky, Lexington, KY, USA
- Department of Neurosurgery, College of Medicine, University of Kentucky, Lexington, KY, USA
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, USA
- Department of Neurology, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Craig G. van Horne
- Brain Restoration Center, College of Medicine, University of Kentucky, Lexington, KY, USA
- Department of Neurosurgery, College of Medicine, University of Kentucky, Lexington, KY, USA
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
15
|
Chen P, Wang B, Li M, Cui C, Liu F, Gao Y. Celastrol inhibits the proliferation and migration of MCF-7 cells through the leptin-triggered PI3K/AKT pathway. Comput Struct Biotechnol J 2022; 20:3173-3181. [PMID: 35782744 PMCID: PMC9234344 DOI: 10.1016/j.csbj.2022.06.042] [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: 02/11/2022] [Revised: 06/15/2022] [Accepted: 06/16/2022] [Indexed: 11/16/2022] Open
Abstract
Leptin is the pivotal modulator in the onset and progression of breast cancer and obesity. Celastrol, which is extracted from the roots of Tripterygium wilfordi plants, exerts various anticancer bioactivities and has recently emerged as a candidate to treat obesity by improving leptin sensitivity. However, the relationship between leptin and celastrol in the treatment of breast cancer is unknown. Here, the growth and migration of MCF-7 cells induced by leptin were tested to demonstrate the antineoplastic activity of celastrol. Transcriptomic analysis and western blotting were conducted to explore the biological roles of leptin in treating breast cancer with celastrol. The present findings showed that celastrol remarkably reversed leptin-triggered cell proliferation and migration in MCF-7 cells. Fifty-two mRNAs with fivefold higher counts and 149 mRNAs with fivefold lower counts were identified in the celastrol-treated MCF-7 cells. According to the GO and KEGG analyses, the effects of celastrol on MCF-7 cells forced lipid metabolism and the endocrine system. Moreover, leptin treatment induced phosphorylation of leptin receptor and PI3K/AKT in MCF-7 cells, whereas pretreatment with celastrol partly abrogated leptin activation. The binding of celastrol to the leptin receptor was also confirmed by molecular docking. The antitumor effect of celastrol is proposed to be mediated by its binding to the leptin receptor and controlled downregulation of the PI3K/AKT pathway.
Collapse
|
16
|
Cintron-Colon AF, Almeida-Alves G, VanGyseghem JM, Spitsbergen JM. GDNF to the rescue: GDNF delivery effects on motor neurons and nerves, and muscle re-innervation after peripheral nerve injuries. Neural Regen Res 2021; 17:748-753. [PMID: 34472460 PMCID: PMC8530131 DOI: 10.4103/1673-5374.322446] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Peripheral nerve injuries commonly occur due to trauma, like a traffic accident. Peripheral nerves get severed, causing motor neuron death and potential muscle atrophy. The current golden standard to treat peripheral nerve lesions, especially lesions with large (≥ 3 cm) nerve gaps, is the use of a nerve autograft or reimplantation in cases where nerve root avulsions occur. If not tended early, degeneration of motor neurons and loss of axon regeneration can occur, leading to loss of function. Although surgical procedures exist, patients often do not fully recover, and quality of life deteriorates. Peripheral nerves have limited regeneration, and it is usually mediated by Schwann cells and neurotrophic factors, like glial cell line-derived neurotrophic factor, as seen in Wallerian degeneration. Glial cell line-derived neurotrophic factor is a neurotrophic factor known to promote motor neuron survival and neurite outgrowth. Glial cell line-derived neurotrophic factor is upregulated in different forms of nerve injuries like axotomy, sciatic nerve crush, and compression, thus creating great interest to explore this protein as a potential treatment for peripheral nerve injuries. Exogenous glial cell line-derived neurotrophic factor has shown positive effects in regeneration and functional recovery when applied in experimental models of peripheral nerve injuries. In this review, we discuss the mechanism of repair provided by Schwann cells and upregulation of glial cell line-derived neurotrophic factor, the latest findings on the effects of glial cell line-derived neurotrophic factor in different types of peripheral nerve injuries, delivery systems, and complementary treatments (electrical muscle stimulation and exercise). Understanding and overcoming the challenges of proper timing and glial cell line-derived neurotrophic factor delivery is paramount to creating novel treatments to tend to peripheral nerve injuries to improve patients’ quality of life.
Collapse
Affiliation(s)
| | | | | | - John M Spitsbergen
- Biological Sciences Department, Western Michigan University, Kalamazoo, MI, USA
| |
Collapse
|
17
|
Ma C, Zhang W, Cao M. Role of the Peripheral Nervous System in PD Pathology, Diagnosis, and Treatment. Front Neurosci 2021; 15:598457. [PMID: 33994915 PMCID: PMC8119739 DOI: 10.3389/fnins.2021.598457] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 03/30/2021] [Indexed: 12/13/2022] Open
Abstract
Studies on Parkinson disease (PD) have mostly focused on the central nervous system—specifically, on the loss of mesencephalic dopaminergic neurons and associated motor dysfunction. However, the peripheral nervous system (PNS) is gaining prominence in PD research, with increasing clinical attention being paid to non-motor symptoms. Researchers found abnormal deposition of α-synuclein and neuroinflammation in the PNS. Attempts have been made to use these pathological changes during the clinical diagnosis of PD. Animal studies demonstrated that combined transplantation of autologous peripheral nerves and cells with tyrosine hydroxylase activity can reduce dopaminergic neuronal damage, and similar effects were observed in some clinical trials. In this review, we will systematically explain PNS performance in PD pathology and its clinical diagnostic research, describe PNS experimental results [especially Schwann cell (SC) transplantation in the treatment of PD animal models] and the results of clinical trials, and discuss future directions. The mechanism by which SCs produce such a therapeutic effect and the safety of transplantation therapy are briefly described.
Collapse
Affiliation(s)
- Chengxiao Ma
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, China.,Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Wen Zhang
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, China.,Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Maohong Cao
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
18
|
Gera G, Guduru Z, Yamasaki T, Gurwell JA, Chau MJ, Krotinger A, Schmitt FA, Slevin JT, Gerhardt GA, van Horne C, Quintero JE. Gait and Balance Changes with Investigational Peripheral Nerve Cell Therapy during Deep Brain Stimulation in People with Parkinson's Disease. Brain Sci 2021; 11:brainsci11040500. [PMID: 33921079 PMCID: PMC8071359 DOI: 10.3390/brainsci11040500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/25/2021] [Accepted: 04/10/2021] [Indexed: 12/02/2022] Open
Abstract
Background: The efficacy of deep brain stimulation (DBS) and dopaminergic therapy is known to decrease over time. Hence, a new investigational approach combines implanting autologous injury-activated peripheral nerve grafts (APNG) at the time of bilateral DBS surgery to the globus pallidus interna. Objectives: In a study where APNG was unilaterally implanted into the substantia nigra, we explored the effects on clinical gait and balance assessments over two years in 14 individuals with Parkinson’s disease. Methods: Computerized gait and balance evaluations were performed without medication, and stimulation was in the off state for at least 12 h to best assess the role of APNG implantation alone. We hypothesized that APNG might improve gait and balance deficits associated with PD. Results: While people with a degenerative movement disorder typically worsen with time, none of the gait parameters significantly changed across visits in this 24 month study. The postural stability item in the UPDRS did not worsen from baseline to the 24-month follow-up. However, we measured gait and balance improvements in the two most affected individuals, who had moderate PD. In these two individuals, we observed an increase in gait velocity and step length that persisted over 6 and 24 months. Conclusions: Participants did not show worsening of gait and balance performance in the off therapy state two years after surgery, while the two most severely affected participants showed improved performance. Further studies may better address the long-term maintanenace of these results.
Collapse
Affiliation(s)
- Geetanjali Gera
- Department of Physical Therapy, College of Health Sciences, University of Kentucky, 204L 900 South Limestone Street, Lexington, KY 40536, USA
- Brain Restoration Center, University of Kentucky, Lexington, KY 40536, USA; (Z.G.); (T.Y.); (J.A.G.); (M.J.C.); (F.A.S.); (J.T.S.); (G.A.G.); (C.v.H.); (J.E.Q.)
- Correspondence: ; Tel.: +1-859-218-0547
| | - Zain Guduru
- Brain Restoration Center, University of Kentucky, Lexington, KY 40536, USA; (Z.G.); (T.Y.); (J.A.G.); (M.J.C.); (F.A.S.); (J.T.S.); (G.A.G.); (C.v.H.); (J.E.Q.)
- Neurology, University of Kentucky, Lexington, KY 40536, USA
| | - Tritia Yamasaki
- Brain Restoration Center, University of Kentucky, Lexington, KY 40536, USA; (Z.G.); (T.Y.); (J.A.G.); (M.J.C.); (F.A.S.); (J.T.S.); (G.A.G.); (C.v.H.); (J.E.Q.)
- Neurology, University of Kentucky, Lexington, KY 40536, USA
- Neuroscience, University of Kentucky, Lexington, KY 40536, USA
- Veterans Affairs Medical Center, Lexington, KY 40502, USA
| | - Julie A. Gurwell
- Brain Restoration Center, University of Kentucky, Lexington, KY 40536, USA; (Z.G.); (T.Y.); (J.A.G.); (M.J.C.); (F.A.S.); (J.T.S.); (G.A.G.); (C.v.H.); (J.E.Q.)
- Neurology, University of Kentucky, Lexington, KY 40536, USA
| | - Monica J. Chau
- Brain Restoration Center, University of Kentucky, Lexington, KY 40536, USA; (Z.G.); (T.Y.); (J.A.G.); (M.J.C.); (F.A.S.); (J.T.S.); (G.A.G.); (C.v.H.); (J.E.Q.)
- Neurosurgery, University of Kentucky, Lexington, KY 40536, USA
| | - Anna Krotinger
- Department of Neuroscience, Wesleyan University, Middletown, CT 06459, USA;
| | - Frederick A. Schmitt
- Brain Restoration Center, University of Kentucky, Lexington, KY 40536, USA; (Z.G.); (T.Y.); (J.A.G.); (M.J.C.); (F.A.S.); (J.T.S.); (G.A.G.); (C.v.H.); (J.E.Q.)
- Neurology, University of Kentucky, Lexington, KY 40536, USA
| | - John T. Slevin
- Brain Restoration Center, University of Kentucky, Lexington, KY 40536, USA; (Z.G.); (T.Y.); (J.A.G.); (M.J.C.); (F.A.S.); (J.T.S.); (G.A.G.); (C.v.H.); (J.E.Q.)
- Neurology, University of Kentucky, Lexington, KY 40536, USA
- Veterans Affairs Medical Center, Lexington, KY 40502, USA
| | - Greg A. Gerhardt
- Brain Restoration Center, University of Kentucky, Lexington, KY 40536, USA; (Z.G.); (T.Y.); (J.A.G.); (M.J.C.); (F.A.S.); (J.T.S.); (G.A.G.); (C.v.H.); (J.E.Q.)
- Neurology, University of Kentucky, Lexington, KY 40536, USA
- Neuroscience, University of Kentucky, Lexington, KY 40536, USA
- Neurosurgery, University of Kentucky, Lexington, KY 40536, USA
| | - Craig van Horne
- Brain Restoration Center, University of Kentucky, Lexington, KY 40536, USA; (Z.G.); (T.Y.); (J.A.G.); (M.J.C.); (F.A.S.); (J.T.S.); (G.A.G.); (C.v.H.); (J.E.Q.)
- Neuroscience, University of Kentucky, Lexington, KY 40536, USA
- Neurosurgery, University of Kentucky, Lexington, KY 40536, USA
| | - Jorge E. Quintero
- Brain Restoration Center, University of Kentucky, Lexington, KY 40536, USA; (Z.G.); (T.Y.); (J.A.G.); (M.J.C.); (F.A.S.); (J.T.S.); (G.A.G.); (C.v.H.); (J.E.Q.)
- Neuroscience, University of Kentucky, Lexington, KY 40536, USA
- Neurosurgery, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
19
|
Nazareth L, St John J, Murtaza M, Ekberg J. Phagocytosis by Peripheral Glia: Importance for Nervous System Functions and Implications in Injury and Disease. Front Cell Dev Biol 2021; 9:660259. [PMID: 33898462 PMCID: PMC8060502 DOI: 10.3389/fcell.2021.660259] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 03/17/2021] [Indexed: 12/30/2022] Open
Abstract
The central nervous system (CNS) has very limited capacity to regenerate after traumatic injury or disease. In contrast, the peripheral nervous system (PNS) has far greater capacity for regeneration. This difference can be partly attributed to variances in glial-mediated functions, such as axon guidance, structural support, secretion of growth factors and phagocytic activity. Due to their growth-promoting characteristic, transplantation of PNS glia has been trialed for neural repair. After peripheral nerve injuries, Schwann cells (SCs, the main PNS glia) phagocytose myelin debris and attract macrophages to the injury site to aid in debris clearance. One peripheral nerve, the olfactory nerve, is unique in that it continuously regenerates throughout life. The olfactory nerve glia, olfactory ensheathing cells (OECs), are the primary phagocytes within this nerve, continuously clearing axonal debris arising from the normal regeneration of the nerve and after injury. In contrast to SCs, OECs do not appear to attract macrophages. SCs and OECs also respond to and phagocytose bacteria, a function likely critical for tackling microbial invasion of the CNS via peripheral nerves. However, phagocytosis is not always effective; inflammation, aging and/or genetic factors may contribute to compromised phagocytic activity. Here, we highlight the diverse roles of SCs and OECs with the focus on their phagocytic activity under physiological and pathological conditions. We also explore why understanding the contribution of peripheral glia phagocytosis may provide us with translational strategies for achieving axonal regeneration of the injured nervous system and potentially for the treatment of certain neurological diseases.
Collapse
Affiliation(s)
- Lynn Nazareth
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia.,Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia
| | - James St John
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia.,Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia.,Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD, Australia
| | - Mariyam Murtaza
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia.,Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia.,Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD, Australia
| | - Jenny Ekberg
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia.,Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia.,Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD, Australia
| |
Collapse
|
20
|
Schwann Cell Role in Selectivity of Nerve Regeneration. Cells 2020; 9:cells9092131. [PMID: 32962230 PMCID: PMC7563640 DOI: 10.3390/cells9092131] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 09/16/2020] [Accepted: 09/17/2020] [Indexed: 12/13/2022] Open
Abstract
Peripheral nerve injuries result in the loss of the motor, sensory and autonomic functions of the denervated segments of the body. Neurons can regenerate after peripheral axotomy, but inaccuracy in reinnervation causes a permanent loss of function that impairs complete recovery. Thus, understanding how regenerating axons respond to their environment and direct their growth is essential to improve the functional outcome of patients with nerve lesions. Schwann cells (SCs) play a crucial role in the regeneration process, but little is known about their contribution to specific reinnervation. Here, we review the mechanisms by which SCs can differentially influence the regeneration of motor and sensory axons. Mature SCs express modality-specific phenotypes that have been associated with the promotion of selective regeneration. These include molecular markers, such as L2/HNK-1 carbohydrate, which is differentially expressed in motor and sensory SCs, or the neurotrophic profile after denervation, which differs remarkably between SC modalities. Other important factors include several molecules implicated in axon-SC interaction. This cell–cell communication through adhesion (e.g., polysialic acid) and inhibitory molecules (e.g., MAG) contributes to guiding growing axons to their targets. As many of these factors can be modulated, further research will allow the design of new strategies to improve functional recovery after peripheral nerve injuries.
Collapse
|