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
|
Leng SZ, Fang MJ, Wang YM, Lin ZJ, Li QY, Xu YN, Mai CL, Wan JY, Yu Y, Wei M, Li Y, Zheng YF, Zhang KL, Wang YJ, Zhou LJ, Tan Z, Zhang H. Elevated plasma CXCL12 leads to pain chronicity via positive feedback upregulation of CXCL12/CXCR4 axis in pain synapses. J Headache Pain 2024; 25:213. [PMID: 39627724 PMCID: PMC11616163 DOI: 10.1186/s10194-024-01917-w] [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: 07/09/2024] [Accepted: 11/16/2024] [Indexed: 12/06/2024] Open
Abstract
BACKGROUND Chronic pain poses a clinical challenge due to its associated costly disability and treatment needs. Determining how pain transitions from acute to chronic is crucial for effective management. Upregulation of the chemokine C-X-C motif ligand 12 (CXCL12) in nociceptive pathway is associated with chronic pain. Our previous study has reported that elevated plasma CXCL12 mediates intracerebral neuroinflammation and the comorbidity of cognitive impairment in neuropathic pain, but whether it is also involved in the pathogenesis of pathologic pain has not been investigated. METHODS Intravenous or intrathecal injection (i.v. or i.t.) of recombinant mouse CXCL12, neutralizing antibody (anti-CXCL12) or AMD3100 [an antagonist of its receptor C-X-C chemokine receptor type 4 (CXCR4)] was used to investigate the role of CXCL12 signaling pathway in pain chronicity. Two behavioral tests were used to examine pain changes. ELISA, immunofluorescence staining, Western blot, quantitative Real Time-PCR and Cytokine array were applied to detect the expressions of different molecules. RESULTS We found that increased plasma CXCL12 was positively correlated with pain severity in both chronic pain patients and neuropathic pain model in mice with spared nerve injury (SNI). Neutralizing plasma CXCL12 mitigated SNI-induced hyperalgesia. A single i.v. injection of CXCL12 induced prolonged mechanical hyperalgesia and activation of the nociceptive pathway. Multiple intravenous CXCL12 caused persistent hypersensitivity, enhanced structural plasticity of nociceptors and up-regulation of the CXCL12/CXCR4 axis in the dorsal root ganglion (DRG) and spinal dorsal horn (SDH). However, intrathecal blocking of CXCL12/CXCR4 pathway by CXCL12 antibody or CXCR4 antagonist AMD3100 significantly alleviated CXCL12-induced pain hypersensitivity and pathological changes. CONCLUSIONS Our study provides strong evidence that a sustained increase in plasma CXCL12 contributes to neuropathic pain through a positive feedback loop that enhances nociceptor plasticity, and suggests that targeting CXCL12/CXCR4 axis in plasma or nociceptive pathways has potential value in regulating pain chronicity.
Collapse
Affiliation(s)
- Shi-Ze Leng
- Department of Anesthesiology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China
| | - Mei-Jia Fang
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine and Guangdong Province Key Laboratory of Brain Function and Disease, Sun Yat-Sen University , Guangzhou, 510080, China
| | - Yi-Min Wang
- Department of Anesthesiology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China
| | - Zhen-Jia Lin
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine and Guangdong Province Key Laboratory of Brain Function and Disease, Sun Yat-Sen University , Guangzhou, 510080, China
| | - Qian-Yi Li
- Department of Anesthesiology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China
| | - Ya-Nan Xu
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine and Guangdong Province Key Laboratory of Brain Function and Disease, Sun Yat-Sen University , Guangzhou, 510080, China
| | - Chun-Lin Mai
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine and Guangdong Province Key Laboratory of Brain Function and Disease, Sun Yat-Sen University , Guangzhou, 510080, China
| | - Jun-Ya Wan
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine and Guangdong Province Key Laboratory of Brain Function and Disease, Sun Yat-Sen University , Guangzhou, 510080, China
| | - Yangyinhui Yu
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine and Guangdong Province Key Laboratory of Brain Function and Disease, Sun Yat-Sen University , Guangzhou, 510080, China
| | - Ming Wei
- Department of Anesthesiology and Pain Clinic, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Ying Li
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine and Guangdong Province Key Laboratory of Brain Function and Disease, Sun Yat-Sen University , Guangzhou, 510080, China
| | - Yu-Fan Zheng
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine and Guangdong Province Key Laboratory of Brain Function and Disease, Sun Yat-Sen University , Guangzhou, 510080, China
| | - Kai-Lang Zhang
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine and Guangdong Province Key Laboratory of Brain Function and Disease, Sun Yat-Sen University , Guangzhou, 510080, China
| | - Ya-Juan Wang
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine and Guangdong Province Key Laboratory of Brain Function and Disease, Sun Yat-Sen University , Guangzhou, 510080, China
| | - Li-Jun Zhou
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine and Guangdong Province Key Laboratory of Brain Function and Disease, Sun Yat-Sen University , Guangzhou, 510080, China.
| | - Zhi Tan
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine and Guangdong Province Key Laboratory of Brain Function and Disease, Sun Yat-Sen University , Guangzhou, 510080, China.
| | - Hui Zhang
- Department of Anesthesiology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China.
| |
Collapse
|
3
|
Emvalomenos GM, Kang JWM, Jupp B, Mychasiuk R, Keay KA, Henderson LA. Recent developments and challenges in positron emission tomography imaging of gliosis in chronic neuropathic pain. Pain 2024; 165:2184-2199. [PMID: 38713812 DOI: 10.1097/j.pain.0000000000003247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 03/05/2024] [Indexed: 05/09/2024]
Abstract
ABSTRACT Understanding the mechanisms that underpin the transition from acute to chronic pain is critical for the development of more effective and targeted treatments. There is growing interest in the contribution of glial cells to this process, with cross-sectional preclinical studies demonstrating specific changes in these cell types capturing targeted timepoints from the acute phase and the chronic phase. In vivo longitudinal assessment of the development and evolution of these changes in experimental animals and humans has presented a significant challenge. Recent technological advances in preclinical and clinical positron emission tomography, including the development of specific radiotracers for gliosis, offer great promise for the field. These advances now permit tracking of glial changes over time and provide the ability to relate these changes to pain-relevant symptomology, comorbid psychiatric conditions, and treatment outcomes at both a group and an individual level. In this article, we summarize evidence for gliosis in the transition from acute to chronic pain and provide an overview of the specific radiotracers available to measure this process, highlighting their potential, particularly when combined with ex vivo / in vitro techniques, to understand the pathophysiology of chronic neuropathic pain. These complementary investigations can be used to bridge the existing gap in the field concerning the contribution of gliosis to neuropathic pain and identify potential targets for interventions.
Collapse
Affiliation(s)
- Gaelle M Emvalomenos
- School of Medical Sciences [Neuroscience], and the Brain and Mind Centre, The University of Sydney, Sydney, Australia
| | - James W M Kang
- School of Medical Sciences [Neuroscience], and the Brain and Mind Centre, The University of Sydney, Sydney, Australia
| | - Bianca Jupp
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Australia
| | - Kevin A Keay
- School of Medical Sciences [Neuroscience], and the Brain and Mind Centre, The University of Sydney, Sydney, Australia
| | - Luke A Henderson
- School of Medical Sciences [Neuroscience], and the Brain and Mind Centre, The University of Sydney, Sydney, Australia
| |
Collapse
|
4
|
Morag Y, Kung TA, Soussahn S, Chen Q, Cederna PS. Magnetic Resonance Imaging appearance of regenerative peripheral nerve interface. J Plast Reconstr Aesthet Surg 2024; 99:47-54. [PMID: 39353283 DOI: 10.1016/j.bjps.2024.09.017] [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: 06/28/2024] [Accepted: 09/01/2024] [Indexed: 10/04/2024]
Abstract
PURPOSE To describe the MRI appearance of regenerative peripheral nerve interface (RPNI) and the potential association between the MRI appearance and RPNI revision. MATERIAL AND METHODS A retrospective assessment was undertaken of the MRI appearance of RPNIs performed at our institution between 1/1/2010 and 7/29/2023 with clinical correlation. RESULTS Fourteen patients (8 men and 6 women, age range 31-80 years, median age 51 years) with technically adequate MRI of RPNIs were included in this study including 5 patients with below knee amputation with 5 tibial and 4 common peroneal nerves RPNI, 8 patients with above knee amputations (AKA) with sciatic RPNIs, and 1 patient following forequarter amputation with a brachial plexus RPNI. Two patients underwent revision RPNI surgery thrice (AKA-sciatic nerve) for a total of 6 RPNI revisions. On T1 weighted sequences, all RPNIs were isointense to the muscle and blended with the surrounding scar and muscle tissues whereas on T2 weighted sequences, all RPNIs were hyperintense in signal compared to the muscle. All but 1 RPNI underwent post contrast enhancement in variable patterns. No statistically significant difference in MRI appearance was found between RPNIs with or without a following RPNI revision surgery. CONCLUSION RPNI on MRI typically have a bright and intermediate signal on T2 and T1 weighted sequences, respectively, and typically undergo postcontrast enhancement in variable patterns without a statistically significant difference between the cases with and without follow-up RPNI revision. However, enhancement of RPNI should not be misconstrued as pathological.
Collapse
Affiliation(s)
- Yoav Morag
- Musculoskeletal Imaging Division, Department of Radiology, University of Michigan, Ann Arbor, MI, United States.
| | - Theodore A Kung
- Department of Surgery, Section of Plastic Surgery, University of Michigan, Ann Arbor, MI, United States; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Samer Soussahn
- Musculoskeletal Imaging Division, Department of Radiology, University of Michigan, Ann Arbor, MI, United States
| | - Qiaochu Chen
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, United States
| | - Paul S Cederna
- Department of Surgery, Section of Plastic Surgery, University of Michigan, Ann Arbor, MI, United States; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
5
|
Ran C, Olofsgård FJ, Wellfelt K, Steinberg A, Belin AC. Elevated cytokine levels in the central nervous system of cluster headache patients in bout and in remission. J Headache Pain 2024; 25:121. [PMID: 39044165 PMCID: PMC11267889 DOI: 10.1186/s10194-024-01829-9] [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: 06/20/2024] [Accepted: 07/17/2024] [Indexed: 07/25/2024] Open
Abstract
BACKGROUND Cluster headache is characterized by activation of the trigeminovascular pathway with subsequent pain signalling in the meningeal vessels, and inflammation has been suggested to play a role in the pathophysiology. To further investigate inflammation in cluster headache, inflammatory markers were analysed in patients with cluster headache and controls. METHODS We performed a case-control study, collecting cerebrospinal fluid and serum samples from healthy controls, cluster headache patients in remission, active bout, and during an attack to cover the dynamic range of the cluster headache phenotype. Inflammatory markers were quantified using Target 48 OLINK cytokine panels. RESULTS Altered levels of several cytokines were found in patients with cluster headache compared to controls. CCL8, CCL13, CCL11, CXCL10, CXCL11, HGF, MMP1, TNFSF10 and TNFSF12 levels in cerebrospinal fluid were comparable in active bout and remission, though significantly higher than in controls. In serum samples, CCL11 and CXCL11 displayed decreased levels in patients. Only one cytokine, IL-13 was differentially expressed in serum during attacks. CONCLUSION AND INTERPRETATION Our data shows signs of possible neuroinflammation occurring in biological samples from cluster headache patients. Increased cerebrospinal fluid cytokine levels are detectable in active bout and during remission, indicating neuroinflammation could be considered a marker for cluster headache and is unrelated to the different phases of the disorder.
Collapse
Affiliation(s)
- Caroline Ran
- Centre for Cluster Headache, Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | | | - Katrin Wellfelt
- Centre for Cluster Headache, Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Anna Steinberg
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - Andrea Carmine Belin
- Centre for Cluster Headache, Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
6
|
Li Q, Zhang F, Fu X, Han N. Therapeutic Potential of Mesenchymal Stem Cell-Derived Exosomes as Nanomedicine for Peripheral Nerve Injury. Int J Mol Sci 2024; 25:7882. [PMID: 39063125 PMCID: PMC11277195 DOI: 10.3390/ijms25147882] [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: 05/05/2024] [Revised: 07/02/2024] [Accepted: 07/07/2024] [Indexed: 07/28/2024] Open
Abstract
Peripheral nerve injury (PNI) is a complex and protracted process, and existing therapeutic approaches struggle to achieve effective nerve regeneration. Recent studies have shown that mesenchymal stem cells (MSCs) may be a pivotal choice for treating peripheral nerve injury. MSCs possess robust paracrine capabilities, and exosomes, as the primary secretome of MSCs, are considered crucial regulatory mediators involved in peripheral nerve regeneration. Exosomes, as nanocarriers, can transport various endogenous or exogenous bioactive substances to recipient cells, thereby promoting vascular and axonal regeneration while suppressing inflammation and pain. In this review, we summarize the mechanistic roles of exosomes derived from MSCs in peripheral nerve regeneration, discuss the engineering strategies for MSC-derived exosomes to improve therapeutic potential, and explore the combined effects of MSC-derived exosomes with biomaterials (nerve conduits, hydrogels) in peripheral nerve regeneration.
Collapse
Affiliation(s)
- Qicheng Li
- Department of Trauma and Orthopedics, Peking University People’s Hospital, Beijing 100044, China; (Q.L.); (F.Z.); (X.F.)
- Key Laboratory of Trauma and Neural Regeneration, Peking University, Beijing 100044, China
| | - Fengshi Zhang
- Department of Trauma and Orthopedics, Peking University People’s Hospital, Beijing 100044, China; (Q.L.); (F.Z.); (X.F.)
- Key Laboratory of Trauma and Neural Regeneration, Peking University, Beijing 100044, China
| | - Xiaoyang Fu
- Department of Trauma and Orthopedics, Peking University People’s Hospital, Beijing 100044, China; (Q.L.); (F.Z.); (X.F.)
- Key Laboratory of Trauma and Neural Regeneration, Peking University, Beijing 100044, China
| | - Na Han
- Department of Trauma and Orthopedics, Peking University People’s Hospital, Beijing 100044, China; (Q.L.); (F.Z.); (X.F.)
- Key Laboratory of Trauma and Neural Regeneration, Peking University, Beijing 100044, China
- National Center for Trauma Medicine, Beijing 100044, China
| |
Collapse
|
7
|
Sullivan JM, Bagnell AM, Alevy J, Avila EM, Mihaljević L, Saavedra-Rivera PC, Kong L, Huh JS, McCray BA, Aisenberg WH, Zuberi AR, Bogdanik L, Lutz CM, Qiu Z, Quinlan KA, Searson PC, Sumner CJ. Gain-of-function mutations of TRPV4 acting in endothelial cells drive blood-CNS barrier breakdown and motor neuron degeneration in mice. Sci Transl Med 2024; 16:eadk1358. [PMID: 38776392 PMCID: PMC11316273 DOI: 10.1126/scitranslmed.adk1358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 05/01/2024] [Indexed: 05/25/2024]
Abstract
Blood-CNS barrier disruption is a hallmark of numerous neurological disorders, yet whether barrier breakdown is sufficient to trigger neurodegenerative disease remains unresolved. Therapeutic strategies to mitigate barrier hyperpermeability are also limited. Dominant missense mutations of the cation channel transient receptor potential vanilloid 4 (TRPV4) cause forms of hereditary motor neuron disease. To gain insights into the cellular basis of these disorders, we generated knock-in mouse models of TRPV4 channelopathy by introducing two disease-causing mutations (R269C and R232C) into the endogenous mouse Trpv4 gene. TRPV4 mutant mice exhibited weakness, early lethality, and regional motor neuron loss. Genetic deletion of the mutant Trpv4 allele from endothelial cells (but not neurons, glia, or muscle) rescued these phenotypes. Symptomatic mutant mice exhibited focal disruptions of blood-spinal cord barrier (BSCB) integrity, associated with a gain of function of mutant TRPV4 channel activity in neural vascular endothelial cells (NVECs) and alterations of NVEC tight junction structure. Systemic administration of a TRPV4-specific antagonist abrogated channel-mediated BSCB impairments and provided a marked phenotypic rescue of symptomatic mutant mice. Together, our findings show that mutant TRPV4 channels can drive motor neuron degeneration in a non-cell autonomous manner by precipitating focal breakdown of the BSCB. Further, these data highlight the reversibility of TRPV4-mediated BSCB impairments and identify a potential therapeutic strategy for patients with TRPV4 mutations.
Collapse
Affiliation(s)
- Jeremy M. Sullivan
- Department of Neurology, Johns Hopkins University School of Medicine; Baltimore, MD 21205, USA
| | - Anna M. Bagnell
- Department of Neurology, Johns Hopkins University School of Medicine; Baltimore, MD 21205, USA
| | - Jonathan Alevy
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine; Baltimore, MD 21205, USA
| | - Elvia Mena Avila
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island; Kingston, RI 02881, USA
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island; Kingston, RI 02881, USA
| | - Ljubica Mihaljević
- Department of Physiology, Johns Hopkins University School of Medicine; Baltimore, MD 21205, USA
| | | | - Lingling Kong
- Department of Neurology, Johns Hopkins University School of Medicine; Baltimore, MD 21205, USA
| | - Jennifer S. Huh
- Department of Neurology, Johns Hopkins University School of Medicine; Baltimore, MD 21205, USA
| | - Brett A. McCray
- Department of Neurology, Johns Hopkins University School of Medicine; Baltimore, MD 21205, USA
| | - William H. Aisenberg
- Department of Neurology, Johns Hopkins University School of Medicine; Baltimore, MD 21205, USA
| | | | | | | | - Zhaozhu Qiu
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine; Baltimore, MD 21205, USA
- Department of Physiology, Johns Hopkins University School of Medicine; Baltimore, MD 21205, USA
| | - Katharina A. Quinlan
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island; Kingston, RI 02881, USA
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island; Kingston, RI 02881, USA
| | - Peter C. Searson
- Institute for Nanobiotechnology, Johns Hopkins University; Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University; Baltimore, MD 21218, USA
- Department of Materials Science and Engineering, Johns Hopkins University; Baltimore, MD 21218, USA
| | - Charlotte J. Sumner
- Department of Neurology, Johns Hopkins University School of Medicine; Baltimore, MD 21205, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine; Baltimore, MD 21205, USA
| |
Collapse
|
8
|
Hayashi K, Chwei-Chin Chuang D, Nai-Jen Chang T, Chuieng-Yi Lu J. Functioning Free Muscle Transplantation to Restore Finger Movement for Sequalae of Volkmann Ischemic Contracture. Hand Clin 2024; 40:269-281. [PMID: 38553098 DOI: 10.1016/j.hcl.2023.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/02/2024]
Abstract
Volkmann ischemic contracture (VIC) is a devastating condition that results from neglected compartment syndrome, which leads to prolonged ischemia, irreversible tissue necrosis, and various degrees of muscle and nerve damage, causing serious motor and sensory functional implications for the limb and a spectrum of diseases associated with worsening deformities. A thorough understanding of the anatomy and VIC pathophysiology is needed to plan an appropriate strategy. Functioning free muscle transplantation (FFMT) can restore finger movement in a paralyzed limb but requires a three-staged approach to maximize the benefits of FFMT, leading to meaningful finger extrinsic function.
Collapse
Affiliation(s)
- Kota Hayashi
- Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital, Taipei - Linkou, No. 5, Fuxing Street, Guishan District, Taoyuan City 333, Taiwan
| | - David Chwei-Chin Chuang
- Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital, Taipei - Linkou, No. 5, Fuxing Street, Guishan District, Taoyuan City 333, Taiwan
| | - Tommy Nai-Jen Chang
- Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital, Taipei - Linkou, No. 5, Fuxing Street, Guishan District, Taoyuan City 333, Taiwan
| | - Johnny Chuieng-Yi Lu
- Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital, Taipei - Linkou, No. 5, Fuxing Street, Guishan District, Taoyuan City 333, Taiwan.
| |
Collapse
|
9
|
Smith PA. The Known Biology of Neuropathic Pain and Its Relevance to Pain Management. Can J Neurol Sci 2024; 51:32-39. [PMID: 36799022 DOI: 10.1017/cjn.2023.10] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
Patients with neuropathic pain are heterogeneous in pathophysiology, etiology, and clinical presentation. Signs and symptoms are determined by the nature of the injury and factors such as genetics, sex, prior injury, age, culture, and environment. Basic science has provided general information about pain etiology by studying the consequences of peripheral injury in rodent models. This is associated with the release of inflammatory cytokines, chemokines, and growth factors that sensitize sensory nerve endings, alter gene expression, promote post-translational modification of proteins, and alter ion channel function. This leads to spontaneous activity in primary afferent neurons that is crucial for the onset and persistence of pain and the release of secondary mediators such as colony-stimulating factor 1 from primary afferent terminals. These promote the release of tertiary mediators such as brain-derived neurotrophic factor and interleukin-1β from microglia and astrocytes. Tertiary mediators facilitate the transmission of nociceptive information at the spinal, thalamic, and cortical levels. For the most part, these findings have failed to identify new therapeutic approaches. More recent basic science has better mirrored the clinical situation by addressing the pathophysiology associated with specific types of injury, refinement of methodology, and attention to various contributory factors such as sex. Improved quantification of sensory profiles in each patient and their distribution into defined clusters may improve translation between basic science and clinical practice. If such quantification can be traced back to cellular and molecular aspects of pathophysiology, this may lead to personalized medicine approaches that dictate a rational therapeutic approach for each individual.
Collapse
Affiliation(s)
- Peter A Smith
- Neuroscience and Mental Health Institute and Department of Pharmacology, University of Alberta, Edmonton, Canada
| |
Collapse
|
10
|
Green-Fulgham SM, Lacagnina MJ, Willcox KF, Li J, Harland ME, Ciena AP, Rocha IRC, Ball JB, Dreher RA, Zuberi YA, Dragavon JM, Chacur M, Maier SF, Watkins LR, Grace PM. Voluntary wheel running prevents formation of membrane attack complexes and myelin degradation after peripheral nerve injury. Brain Behav Immun 2024; 115:419-431. [PMID: 37924957 PMCID: PMC10842182 DOI: 10.1016/j.bbi.2023.10.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 10/04/2023] [Accepted: 10/28/2023] [Indexed: 11/06/2023] Open
Abstract
Regular aerobic activity is associated with a reduced risk of chronic pain in humans and rodents. Our previous studies in rodents have shown that prior voluntary wheel running can normalize redox signaling at the site of peripheral nerve injury, attenuating subsequent neuropathic pain. However, the full extent of neuroprotection offered by voluntary wheel running after peripheral nerve injury is unknown. Here, we show that six weeks of voluntary wheel running prior to chronic constriction injury (CCI) reduced the terminal complement membrane attack complex (MAC) at the sciatic nerve injury site. This was associated with increased expression of the MAC inhibitor CD59. The levels of upstream complement components (C3) and their inhibitors (CD55, CR1 and CFH) were altered by CCI, but not increased by voluntary wheel running. Since MAC can degrade myelin, which in turn contributes to neuropathic pain, we evaluated myelin integrity at the sciatic nerve injury site. We found that the loss of myelinated fibers and decreased myelin protein which occurs in sedentary rats following CCI was not observed in rats with prior running. Substitution of prior voluntary wheel running with exogenous CD59 also attenuated mechanical allodynia and reduced MAC deposition at the nerve injury site, pointing to CD59 as a critical effector of the neuroprotective and antinociceptive actions of prior voluntary wheel running. This study links attenuation of neuropathic pain by prior voluntary wheel running with inhibition of MAC and preservation of myelin integrity at the sciatic nerve injury site.
Collapse
Affiliation(s)
- Suzanne M Green-Fulgham
- Department of Psychology and Neuroscience, and the Center for Neuroscience, University of Colorado, Boulder, CO 80309, USA
| | - Michael J Lacagnina
- Laboratories of Neuroimmunology, Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; MD Anderson Pain Research Consortium, Houston, TX 77030, USA
| | - Kendal F Willcox
- Laboratories of Neuroimmunology, Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; MD Anderson Pain Research Consortium, Houston, TX 77030, USA
| | - Jiahe Li
- Laboratories of Neuroimmunology, Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; MD Anderson Pain Research Consortium, Houston, TX 77030, USA
| | - Michael E Harland
- Department of Psychology and Neuroscience, and the Center for Neuroscience, University of Colorado, Boulder, CO 80309, USA
| | - Adriano Polican Ciena
- Laboratory of Morphology and Physical Activity (LAMAF), Institute of Biosciences, São Paulo State University (UNESP), Rio Claro 13506-900, São Paulo, Brazil
| | - Igor R Correia Rocha
- Department of Psychology and Neuroscience, and the Center for Neuroscience, University of Colorado, Boulder, CO 80309, USA; Laboratory of Neuroanatomy Functional of Pain, Departamento de Anatomia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Jayson B Ball
- Department of Psychology and Neuroscience, and the Center for Neuroscience, University of Colorado, Boulder, CO 80309, USA
| | - Renee A Dreher
- Department of Psychology and Neuroscience, and the Center for Neuroscience, University of Colorado, Boulder, CO 80309, USA
| | - Younus A Zuberi
- Laboratories of Neuroimmunology, Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; MD Anderson Pain Research Consortium, Houston, TX 77030, USA
| | - Joseph M Dragavon
- Advanced Light Microscopy Core, BioFrontiers Institute, University of Colorado, Boulder, CO 80309, USA
| | - Marucia Chacur
- Department of Psychology and Neuroscience, and the Center for Neuroscience, University of Colorado, Boulder, CO 80309, USA; Laboratory of Neuroanatomy Functional of Pain, Departamento de Anatomia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Steven F Maier
- Department of Psychology and Neuroscience, and the Center for Neuroscience, University of Colorado, Boulder, CO 80309, USA
| | - Linda R Watkins
- Department of Psychology and Neuroscience, and the Center for Neuroscience, University of Colorado, Boulder, CO 80309, USA
| | - Peter M Grace
- Laboratories of Neuroimmunology, Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; MD Anderson Pain Research Consortium, Houston, TX 77030, USA.
| |
Collapse
|
11
|
Xu T, Wang J, Wu Y, Wu J, Lu W, Liu M, Zhang S, Xie D, Xin W, Xie J. Ac4C Enhances the Translation Efficiency of Vegfa mRNA and Mediates Central Sensitization in Spinal Dorsal Horn in Neuropathic Pain. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303113. [PMID: 37877615 PMCID: PMC10724395 DOI: 10.1002/advs.202303113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 08/25/2023] [Indexed: 10/26/2023]
Abstract
N4-Acetylcytidine (ac4C), a highly conserved post-transcriptional machinery with extensive existence for RNA modification, plays versatile roles in various cellular processes and functions. However, the molecular mechanism by which ac4C modification mediates neuropathic pain remains elusive. Here, it is found that the enhanced ac4C modification promotes the recruitment of polysome in Vegfa mRNA and strengthens the translation efficiency following SNI. Nerve injury increases the expression of NAT10 and the interaction between NAT10 and Vegfa mRNA in the dorsal horn neurons, and the gain and loss of NAT10 function further confirm that NAT10 is involved in the ac4C modification in Vegfa mRNA and pain behavior. Moreover, the ac4C-mediated VEGFA upregulation contributes to the central sensitivity and neuropathic pain induced by SNI or AAV-hSyn-NAT10. Finally, SNI promotes the binding of HNRNPK in Vegfa mRNA and subsequently recruits the NAT10. The enhanced interaction between HNRNPK and NAT10 contributes to the ac4C modification of Vegfa mRNA and neuropathic pain. These findings suggest that the enhanced interaction between HNRNPK and Vegfa mRNA upregulates the ac4C level by recruiting NAT10 and contributes to the central sensitivity and neuropathic pain following SNI. Blocking this cascade may be a novel therapeutic approach in patients with neuropathic pain.
Collapse
Affiliation(s)
- Ting Xu
- Neuroscience ProgramZhongshan School of MedicineThe Fifth Affiliated HospitalGuangdong Province Key Laboratory of Brain Function and DiseaseDepartment of Physiology and Pain Research CenterSun Yat‐Sen UniversityGuangzhou510080China
| | - Jing Wang
- Neuroscience ProgramZhongshan School of MedicineThe Fifth Affiliated HospitalGuangdong Province Key Laboratory of Brain Function and DiseaseDepartment of Physiology and Pain Research CenterSun Yat‐Sen UniversityGuangzhou510080China
- Department of Pain ManagementHenan Provincial People's HospitalZhengzhou UniversityZhengzhou450000China
| | - Yan Wu
- Department of AnesthesiologyThe First Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouGuangdong510062China
| | - Jia‐Yan Wu
- Neuroscience ProgramZhongshan School of MedicineThe Fifth Affiliated HospitalGuangdong Province Key Laboratory of Brain Function and DiseaseDepartment of Physiology and Pain Research CenterSun Yat‐Sen UniversityGuangzhou510080China
| | - Wei‐Cheng Lu
- State Key Laboratory of Oncology in Southern ChinaCollaborative Innovation for Cancer MedicineSun Yat‐Sen University Cancer CenterGuangzhou510060China
| | - Meng Liu
- Department of Anesthesia and Pain MedicineGuangzhou First People's HospitalGuangzhou510180China
| | - Su‐Bo Zhang
- State Key Laboratory of Oncology in Southern ChinaCollaborative Innovation for Cancer MedicineSun Yat‐Sen University Cancer CenterGuangzhou510060China
| | - Dan Xie
- State Key Laboratory of Oncology in Southern ChinaCollaborative Innovation for Cancer MedicineSun Yat‐Sen University Cancer CenterGuangzhou510060China
| | - Wen‐Jun Xin
- Neuroscience ProgramZhongshan School of MedicineThe Fifth Affiliated HospitalGuangdong Province Key Laboratory of Brain Function and DiseaseDepartment of Physiology and Pain Research CenterSun Yat‐Sen UniversityGuangzhou510080China
| | - Jing‐Dun Xie
- State Key Laboratory of Oncology in Southern ChinaCollaborative Innovation for Cancer MedicineSun Yat‐Sen University Cancer CenterGuangzhou510060China
| |
Collapse
|
12
|
Smith PA. Neuropathic pain; what we know and what we should do about it. FRONTIERS IN PAIN RESEARCH 2023; 4:1220034. [PMID: 37810432 PMCID: PMC10559888 DOI: 10.3389/fpain.2023.1220034] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 09/05/2023] [Indexed: 10/10/2023] Open
Abstract
Neuropathic pain can result from injury to, or disease of the nervous system. It is notoriously difficult to treat. Peripheral nerve injury promotes Schwann cell activation and invasion of immunocompetent cells into the site of injury, spinal cord and higher sensory structures such as thalamus and cingulate and sensory cortices. Various cytokines, chemokines, growth factors, monoamines and neuropeptides effect two-way signalling between neurons, glia and immune cells. This promotes sustained hyperexcitability and spontaneous activity in primary afferents that is crucial for onset and persistence of pain as well as misprocessing of sensory information in the spinal cord and supraspinal structures. Much of the current understanding of pain aetiology and identification of drug targets derives from studies of the consequences of peripheral nerve injury in rodent models. Although a vast amount of information has been forthcoming, the translation of this information into the clinical arena has been minimal. Few, if any, major therapeutic approaches have appeared since the mid 1990's. This may reflect failure to recognise differences in pain processing in males vs. females, differences in cellular responses to different types of injury and differences in pain processing in humans vs. animals. Basic science and clinical approaches which seek to bridge this knowledge gap include better assessment of pain in animal models, use of pain models which better emulate human disease, and stratification of human pain phenotypes according to quantitative assessment of signs and symptoms of disease. This can lead to more personalized and effective treatments for individual patients. Significance statement: There is an urgent need to find new treatments for neuropathic pain. Although classical animal models have revealed essential features of pain aetiology such as peripheral and central sensitization and some of the molecular and cellular mechanisms involved, they do not adequately model the multiplicity of disease states or injuries that may bring forth neuropathic pain in the clinic. This review seeks to integrate information from the multiplicity of disciplines that seek to understand neuropathic pain; including immunology, cell biology, electrophysiology and biophysics, anatomy, cell biology, neurology, molecular biology, pharmacology and behavioral science. Beyond this, it underlines ongoing refinements in basic science and clinical practice that will engender improved approaches to pain management.
Collapse
Affiliation(s)
- Peter A. Smith
- Neuroscience and Mental Health Institute and Department of Pharmacology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
13
|
Chen JTC, Hu X, Otto IUC, Schürger C, von Bieberstein BR, Doppler K, Krug SM, Hankir MK, Blasig R, Sommer C, Brack A, Blasig IE, Rittner HL. Myelin barrier breakdown, mechanical hypersensitivity, and painfulness in polyneuropathy with claudin-12 deficiency. Neurobiol Dis 2023; 185:106246. [PMID: 37527762 DOI: 10.1016/j.nbd.2023.106246] [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] [Received: 02/06/2023] [Revised: 05/25/2023] [Accepted: 07/28/2023] [Indexed: 08/03/2023] Open
Abstract
BACKGROUND The blood-nerve and myelin barrier shield peripheral neurons and their axons. These barriers are sealed by tight junction proteins, which control the passage of potentially noxious molecules including proinflammatory cytokines via paracellular pathways. Peripheral nerve barrier breakdown occurs in various neuropathies, such as chronic inflammatory demyelinating polyradiculoneuropathy (CIDP) and traumatic neuropathy. Here, we studied the functional role of the tight junction protein claudin-12 in regulating peripheral nerve barrier integrity and CIDP pathogenesis. METHODS Sections from sural nerve biopsies from 23 patients with CIDP and non-inflammatory idiopathic polyneuropathy (PNP) were analyzed for claudin-12 and -19 immunoreactivity. Cldn12-KO mice were generated and subjected to the chronic constriction injury (CCI) model of neuropathy. These mice were then characterized using a battery of barrier and behavioral tests, histology, immunohistochemistry, and mRNA/protein expression. In phenotype rescue experiments, the proinflammatory cytokine TNFα was neutralized with the anti-TNFα antibody etanercept; the peripheral nerve barrier was stabilized with the sonic hedgehog agonist smoothened (SAG). RESULTS Compared to those without pain, patients with painful neuropathy exhibited reduced claudin-12 expression independently of fiber loss. Accordingly, global Cldn12-KO in male mice, but not fertile female mice, selectively caused mechanical allodynia associated with a leaky myelin barrier, increased TNFα, decreased sonic hedgehog (SHH), and loss of small axons accompanied by reduced peripheral myelin protein 22 (Pmp22). Other barriers and neurological functions remained intact. The Cldn12-KO phenotype could be rescued either by neutralizing TNFα with etanercept or stabilizing the barrier with SAG, which both also upregulated the Schwann cell barrier proteins Cldn19 and Pmp22. CONCLUSION These results point to a critical role for claudin-12 in maintaining the myelin barrier presumably via Pmp22 and highlight restoration of the hedgehog pathway as a potential treatment strategy for painful inflammatory neuropathy.
Collapse
Affiliation(s)
- Jeremy Tsung-Chieh Chen
- University Hospital Würzburg, Center for Interdisciplinary Pain Medicine, Department of Anesthesiology, Intensive Care, Emergency and Pain Medicine, 97080 Würzburg, Germany
| | - Xiawei Hu
- University Hospital Würzburg, Center for Interdisciplinary Pain Medicine, Department of Anesthesiology, Intensive Care, Emergency and Pain Medicine, 97080 Würzburg, Germany
| | - Isabel U C Otto
- University Hospital Würzburg, Center for Interdisciplinary Pain Medicine, Department of Anesthesiology, Intensive Care, Emergency and Pain Medicine, 97080 Würzburg, Germany
| | - Christina Schürger
- University Hospital Würzburg, Center for Interdisciplinary Pain Medicine, Department of Anesthesiology, Intensive Care, Emergency and Pain Medicine, 97080 Würzburg, Germany
| | - Bruno Rogalla von Bieberstein
- University Hospital Würzburg, Center for Interdisciplinary Pain Medicine, Department of Anesthesiology, Intensive Care, Emergency and Pain Medicine, 97080 Würzburg, Germany
| | - Kathrin Doppler
- University Hospital Würzburg, Department of Neurology, 97080 Würzburg, Germany
| | - Susanne M Krug
- Charité-Universitätsmedizin Berlin, Clinical Physiology/Nutritional Medicine, 13125 Berlin, Germany
| | - Mohammed K Hankir
- University Hospital Würzburg, Department of General, Transplantation, Visceral, Vascular and Pediatric Surgery, 97080 Würzburg, Germany
| | - Rosel Blasig
- Leibnitz Institute of Molecular Pharmacology, Departments of Molecular Physiology and Cell Biology, 13125 Berlin, Germany
| | - Claudia Sommer
- University Hospital Würzburg, Department of Neurology, 97080 Würzburg, Germany
| | - Alexander Brack
- University Hospital Würzburg, Center for Interdisciplinary Pain Medicine, Department of Anesthesiology, Intensive Care, Emergency and Pain Medicine, 97080 Würzburg, Germany
| | - Ingolf E Blasig
- Leibnitz Institute of Molecular Pharmacology, Departments of Molecular Physiology and Cell Biology, 13125 Berlin, Germany
| | - Heike L Rittner
- University Hospital Würzburg, Center for Interdisciplinary Pain Medicine, Department of Anesthesiology, Intensive Care, Emergency and Pain Medicine, 97080 Würzburg, Germany.
| |
Collapse
|
14
|
Reinhold AK, Hartmannsberger B, Burek M, Rittner HL. Stabilizing the neural barrier - A novel approach in pain therapy. Pharmacol Ther 2023; 249:108484. [PMID: 37390969 DOI: 10.1016/j.pharmthera.2023.108484] [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: 04/25/2023] [Revised: 06/08/2023] [Accepted: 06/26/2023] [Indexed: 07/02/2023]
Abstract
Chronic and neuropathic pain are a widespread burden. Incomplete understanding of underlying pathomechanisms is one crucial factor for insufficient treatment. Recently, impairment of the blood nerve barrier (BNB) has emerged as one key aspect of pain initiation and maintenance. In this narrative review, we discuss several mechanisms and putative targets for novel treatment strategies. Cells such as pericytes, local mediators like netrin-1 and specialized proresolving mediators (SPMs), will be covered as well as circulating factors including the hormones cortisol and oestrogen and microRNAs. They are crucial in either the BNB or similar barriers and associated with pain. While clinical studies are still scarce, these findings might provide valuable insight into mechanisms and nurture development of therapeutic approaches.
Collapse
Affiliation(s)
- Ann-Kristin Reinhold
- University Hospital Würzburg, Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, Oberdürrbacher Str. 6, 97080 Würzburg, Germany
| | - Beate Hartmannsberger
- University Hospital Würzburg, Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, Oberdürrbacher Str. 6, 97080 Würzburg, Germany
| | - Malgorzata Burek
- University Hospital Würzburg, Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, Oberdürrbacher Str. 6, 97080 Würzburg, Germany
| | - Heike L Rittner
- University Hospital Würzburg, Department of Anaesthesiology, Intensive Care, Emergency and Pain Medicine, Oberdürrbacher Str. 6, 97080 Würzburg, Germany.
| |
Collapse
|
15
|
Robles-Osorio ML, Sabath E. Tight junction disruption and the pathogenesis of the chronic complications of diabetes mellitus: A narrative review. World J Diabetes 2023; 14:1013-1026. [PMID: 37547580 PMCID: PMC10401447 DOI: 10.4239/wjd.v14.i7.1013] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 03/20/2023] [Accepted: 05/23/2023] [Indexed: 07/12/2023] Open
Abstract
The chronic complications of diabetes mellitus constitute a major public health problem. For example, diabetic eye diseases are the most important cause of blindness, and diabetic nephropathy is the most frequent cause of chronic kidney disease worldwide. The cellular and molecular mechanisms of these chronic complications are still poorly understood, preventing the development of effective treatment strategies. Tight junctions (TJs) are epithelial intercellular junctions located at the most apical region of cell-cell contacts, and their main function is to restrict the passage of molecules through the paracellular space. The TJs consist of over 40 proteins, and the most important are occludin, claudins and the zonula occludens. Accumulating evidence suggests that TJ disruption in different organs, such as the brain, nerves, retina and kidneys, plays a fundamental pathophysiological role in the development of chronic complications. Increased permeability of the blood-brain barrier and the blood-retinal barrier has been demonstrated in diabetic neuropathy, brain injury and diabetic retinopathy. The consequences of TJ disruption on kidney function or progression of kidney disease are currently unknown. In the present review, we highlighted the molecular events that lead to barrier dysfunction in diabetes. Further investigation of the mechanisms underlying TJ disruption is expected to provide new insights into therapeutic approaches to ameliorate the chronic complications of diabetes mellitus.
Collapse
Affiliation(s)
| | - Ernesto Sabath
- Renal and Metabolism Unit, Hospital General de Querétaro, Queretaro 76180, Mexico
- Department of Nutrition, Universidad Autónoma de Queretaro, Queretaro 76230, Mexico
| |
Collapse
|
16
|
Hiraoka Y, Matsumura M, Kakei Y, Takeda D, Shigeoka M, Kimoto A, Hasegawa T, Akashi M. Expression of JCAD and EGFR in Perineurial Cell-Cell Junctions of Human Inferior Alveolar Nerve. J Histochem Cytochem 2023; 71:321-332. [PMID: 37309668 PMCID: PMC10315992 DOI: 10.1369/00221554231182193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 05/24/2023] [Indexed: 06/14/2023] Open
Abstract
Although perineurium has an important role in maintenance of the blood-nerve barrier, understanding of perineurial cell-cell junctions is insufficient. The aim of this study was to analyze the expression of junctional cadherin 5 associated (JCAD) and epidermal growth factor receptor (EGFR) in the perineurium of the human inferior alveolar nerve (IAN) and investigate their roles in perineurial cell-cell junctions using cultured human perineurial cells (HPNCs). In human IAN, JCAD was strongly expressed in endoneurial microvessels. JCAD and EGFR were expressed at various intensities in the perineurium. In HPNCs, JCAD was clearly expressed at cell-cell junctions. EGFR inhibitor AG1478 treatment changed cell morphology and the ratio of JCAD-positive cell-cell contacts of HPNCs. Therefore, JCAD and EGFR may have a role in the regulation of perineurial cell-cell junctions.
Collapse
Affiliation(s)
- Yujiro Hiraoka
- Department of Oral and Maxillofacial Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Megumi Matsumura
- Department of Oral and Maxillofacial Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yasumasa Kakei
- Department of Oral and Maxillofacial Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Daisuke Takeda
- Department of Oral and Maxillofacial Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Manabu Shigeoka
- Department of Oral and Maxillofacial Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
- Division of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Akira Kimoto
- Department of Oral and Maxillofacial Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takumi Hasegawa
- Department of Oral and Maxillofacial Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Masaya Akashi
- Department of Oral and Maxillofacial Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
17
|
Malong L, Napoli I, Casal G, White IJ, Stierli S, Vaughan A, Cattin AL, Burden JJ, Hng KI, Bossio A, Flanagan A, Zhao HT, Lloyd AC. Characterization of the structure and control of the blood-nerve barrier identifies avenues for therapeutic delivery. Dev Cell 2023; 58:174-191.e8. [PMID: 36706755 DOI: 10.1016/j.devcel.2023.01.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 10/26/2022] [Accepted: 01/04/2023] [Indexed: 01/27/2023]
Abstract
The blood barriers of the nervous system protect neural environments but can hinder therapeutic accessibility. The blood-brain barrier (BBB) is well characterized, consisting of endothelial cells with specialized tight junctions and low levels of transcytosis, properties conferred by contacting pericytes and astrocytes. In contrast, the blood-nerve barrier (BNB) of the peripheral nervous system is poorly defined. Here, we characterize the structure of the mammalian BNB, identify the processes that confer barrier function, and demonstrate how the barrier can be opened in response to injury. The homeostatic BNB is leakier than the BBB, which we show is due to higher levels of transcytosis. However, the barrier is reinforced by macrophages that specifically engulf leaked materials, identifying a role for resident macrophages as an important component of the BNB. Finally, we demonstrate the exploitation of these processes to effectively deliver RNA-targeting therapeutics to peripheral nerves, indicating new treatment approaches for nervous system pathologies.
Collapse
Affiliation(s)
- Liza Malong
- UCL Laboratory for Molecular Cell Biology and UCL Cancer Institute, University College London, Gower Street, London, WC1E 6BT, UK
| | - Ilaria Napoli
- UCL Laboratory for Molecular Cell Biology and UCL Cancer Institute, University College London, Gower Street, London, WC1E 6BT, UK
| | - Giulia Casal
- UCL Laboratory for Molecular Cell Biology and UCL Cancer Institute, University College London, Gower Street, London, WC1E 6BT, UK
| | - Ian J White
- UCL Laboratory for Molecular Cell Biology and UCL Cancer Institute, University College London, Gower Street, London, WC1E 6BT, UK
| | - Salome Stierli
- UCL Laboratory for Molecular Cell Biology and UCL Cancer Institute, University College London, Gower Street, London, WC1E 6BT, UK
| | - Andrew Vaughan
- UCL Laboratory for Molecular Cell Biology and UCL Cancer Institute, University College London, Gower Street, London, WC1E 6BT, UK
| | - Anne-Laure Cattin
- UCL Laboratory for Molecular Cell Biology and UCL Cancer Institute, University College London, Gower Street, London, WC1E 6BT, UK
| | - Jemima J Burden
- UCL Laboratory for Molecular Cell Biology and UCL Cancer Institute, University College London, Gower Street, London, WC1E 6BT, UK
| | - Keng I Hng
- UCL Laboratory for Molecular Cell Biology and UCL Cancer Institute, University College London, Gower Street, London, WC1E 6BT, UK
| | - Alessandro Bossio
- UCL Laboratory for Molecular Cell Biology and UCL Cancer Institute, University College London, Gower Street, London, WC1E 6BT, UK
| | - Adrienne Flanagan
- UCL Laboratory for Molecular Cell Biology and UCL Cancer Institute, University College London, Gower Street, London, WC1E 6BT, UK
| | - Hien T Zhao
- IONIS, 2855 Gazelle Court, Carlsbad, CA 92010, USA
| | - Alison C Lloyd
- UCL Laboratory for Molecular Cell Biology and UCL Cancer Institute, University College London, Gower Street, London, WC1E 6BT, UK.
| |
Collapse
|
18
|
Zheng G, Ren J, Shang L, Bao Y. Sonic Hedgehog Signaling Pathway: A Role in Pain Processing. Neurochem Res 2023; 48:1611-1630. [PMID: 36738366 DOI: 10.1007/s11064-023-03864-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 01/13/2023] [Accepted: 01/17/2023] [Indexed: 02/05/2023]
Abstract
Pain, as one of the most prevalent clinical symptoms, is a complex physiological and psychological activity. Long-term severe pain can become unbearable to the body. However, existing treatments do not provide satisfactory results. Therefore, new mechanisms and therapeutic targets need to be urgently explored for pain management. The Sonic hedgehog (Shh) signaling pathway is crucial in embryonic development, cell differentiation and proliferation, and nervous system regulation. Here, we review the recent studies on the Shh signaling pathway and its action in multiple pain-related diseases. The Shh signaling pathway is dysregulated under various pain conditions, such as pancreatic cancer pain, bone cancer pain, chronic post-thoracotomy pain, pain caused by degenerative lumbar disc disease, and toothache. Further studies on the Shh signaling pathway may provide new therapeutic options for pain patients.
Collapse
Affiliation(s)
- Guangda Zheng
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beixiange 5, Xicheng District, Beijing, 100053, China
| | - Juanxia Ren
- Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, Liaoning Province, China
| | - Lu Shang
- Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, Liaoning Province, China
| | - Yanju Bao
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beixiange 5, Xicheng District, Beijing, 100053, China.
| |
Collapse
|
19
|
Astrocyte reactivity in the glia limitans superficialis of the rat medial prefrontal cortex following sciatic nerve injury. Histochem Cell Biol 2023; 159:185-198. [PMID: 36326875 DOI: 10.1007/s00418-022-02161-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/14/2022] [Indexed: 11/06/2022]
Abstract
The glia limitans superficialis (GLS) on the rodent cortical surface consists of astrocyte bodies intermingled with their cytoplasmic processes. Many studies have observed astrocyte reactivity in the medial prefrontal cortex (mPFC) parenchyma induced by a peripheral nerve injury, while the response of GLS astrocytes is still not fully understood. The aim of our study was to identify the reactivity of rat GLS astrocytes in response to sciatic nerve compression (SNC) over different time periods. The alteration of GLS astrocyte reactivity was monitored using immunofluorescence (IF) intensities of glial fibrillary acidic protein (GFAP), glutamine synthetase (GS), and NFκBp65. Our results demonstrated that SNC induced GLS astrocyte reactivity seen as increased intensities of GFAP-IF, and longer extensions of cytoplasmic processes into lamina I. First significant increase of GFAP-IF was observed on post-operation day 7 (POD7) after SNC with further increases on POD14 and POD21. In contrast, dynamic alteration of the extension of cytoplasmic processes into lamina I was detected as early as POD1 and continued throughout the monitored survival periods of both sham and SNC operations. The reactivity of GLS astrocytes was not associated with their proliferation. In addition, GLS astrocytes also displayed a significant decrease in GS immunofluorescence (GS-IF) and NFκB immunofluorescence (NFκB-IF) in response to sham and SNC operation compared with naïve control rats. These results suggest that damaged peripheral tissues (following sham operation as well as peripheral nerve lesions) may induce significant changes in GLS astrocyte reactivity. The signaling mechanism from injured peripheral tissue and nerve remains to be elucidated.
Collapse
|
20
|
Zhou LX, Lin SW, Qiu RH, Lin L, Guo YF, Luo DS, Li YQ, Wang F. Blood-nerve barrier disruption and coagulation system activation induced by mechanical compression injury participate in the peripheral sensitization of trigeminal neuralgia. Front Mol Neurosci 2022; 15:1059980. [PMID: 36618827 PMCID: PMC9810503 DOI: 10.3389/fnmol.2022.1059980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 11/28/2022] [Indexed: 12/24/2022] Open
Abstract
Introduction The aim of this study was to investigate the effect and possible mechanisms of the blood-nerve barrier (BNB) and the coagulation-anticoagulation system in modulating the mechanical allodynia in a trigeminal neuralgia (TN) rat model induced by chronic compression of the trigeminal root entry zone (TREZ). Methods Von Frey filaments were applied to determine the orofacial mechanical allodynia threshold. The BNB permeability was evaluated by Evans blue extravasation test. Immunohistochemical staining and laser confocal microscopy were used to measure the length of the depletion zones of the nodes of Ranvier in the TREZ, the diameter of nerve fibers and the length of the nodal gap. The transcriptional levels of prothrombin and endogenous thrombin inhibitor protease nexin-1 (PN-1) in the TREZ of TN rats were assessed by RT-qPCR. A Western blotting assay was performed to detect the expression of paranodal proteins neurofascin-155 (NF155) and neurofascin-125 (NF125) in the TREZ. The spatiotemporal expression pattern of thrombin activated receptor (i.e. protease activated receptor 1, PAR1) in TREZ were defined by immunostaining and immunoblotting assays. PAR1 receptor inhibitors SCH79797 were administrated to TN rats to analyze the effect of thrombin-PAR1 on orofacial hyperalgesia. Results A compression injury of a rat's TREZ successfully induced TN-like behavior and was accompanied by the destruction of the permeability of the BNB and the promotion of prothrombin and thrombin inhibitor protease nexin-1 (PN-1) expression. The expression of the paranodal proteins neurofascin-155 (NF155) and neurofascin-125 (NF125) was increased, while the nodal gap length of the nodes of Ranvier was widened and the length of node-depleted zones was shortened. Moreover, the expression of PAR1 within the TREZ was upregulated at an early stage of TN, and administration of the PAR1 antagonist SCH79797 effectively ameliorated orofacial mechanical allodynia. Conclusion A compression injury of the TREZ increased the permeability of the BNB and induced disturbances in the local coagulation-anticoagulation system, concomitant with the structural changes in the nodes of Ranvier, thrombin-PAR1 may play a critical role in modulating orofacial mechanical hyperalgesia in a TN rat model.
Collapse
Affiliation(s)
- Lu-Xi Zhou
- Laboratory of Clinical Applied Anatomy, Key Laboratory of Brain Aging and Neurodegenerative Diseases, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Shao-Wei Lin
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Rong-Hui Qiu
- Laboratory of Clinical Applied Anatomy, Key Laboratory of Brain Aging and Neurodegenerative Diseases, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Ling Lin
- Laboratory of Clinical Applied Anatomy, Key Laboratory of Brain Aging and Neurodegenerative Diseases, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, China,Public Technology Service Center, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Yue-Feng Guo
- Laboratory of Clinical Applied Anatomy, Key Laboratory of Brain Aging and Neurodegenerative Diseases, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Dao-Shu Luo
- Laboratory of Clinical Applied Anatomy, Key Laboratory of Brain Aging and Neurodegenerative Diseases, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, China,Dao-Shu Luo,
| | - Yun-Qing Li
- Laboratory of Clinical Applied Anatomy, Key Laboratory of Brain Aging and Neurodegenerative Diseases, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, China,Public Technology Service Center, Fujian Medical University, Fuzhou, Fujian Province, China,Yun-Qing Li,
| | - Feng Wang
- Laboratory of Clinical Applied Anatomy, Key Laboratory of Brain Aging and Neurodegenerative Diseases, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, China,*Correspondence: Feng Wang,
| |
Collapse
|
21
|
Pathophysiology of Post-Traumatic Trigeminal Neuropathic Pain. Biomolecules 2022; 12:biom12121753. [PMID: 36551181 PMCID: PMC9775491 DOI: 10.3390/biom12121753] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 11/11/2022] [Accepted: 11/12/2022] [Indexed: 11/29/2022] Open
Abstract
Trigeminal nerve injury is one of the causes of chronic orofacial pain. Patients suffering from this condition have a significantly reduced quality of life. The currently available management modalities are associated with limited success. This article reviews some of the common causes and clinical features associated with post-traumatic trigeminal neuropathic pain (PTNP). A cascade of events in the peripheral and central nervous system function is involved in the pathophysiology of pain following nerve injuries. Central and peripheral processes occur in tandem and may often be co-dependent. Due to the complexity of central mechanisms, only peripheral events contributing to the pathophysiology have been reviewed in this article. Future investigations will hopefully help gain insight into trigeminal-specific events in the pathophysiology of the development and maintenance of neuropathic pain secondary to nerve injury and enable the development of new therapeutic modalities.
Collapse
|
22
|
The Impact of P-Glycoprotein on Opioid Analgesics: What's the Real Meaning in Pain Management and Palliative Care? Int J Mol Sci 2022; 23:ijms232214125. [PMID: 36430602 PMCID: PMC9695906 DOI: 10.3390/ijms232214125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/01/2022] [Accepted: 11/11/2022] [Indexed: 11/18/2022] Open
Abstract
Opioids are widely used in cancer and non-cancer pain management. However, many transporters at the blood-brain barrier (BBB), such as P-glycoprotein (P-gp, ABCB1/MDR1), may impair their delivery to the brain, thus leading to opioid tolerance. Nonetheless, opioids may regulate P-gp expression, thus altering the transport of other compounds, namely chemotherapeutic agents, resulting in pharmacoresistance. Other kinds of painkillers (e.g., acetaminophen, dexamethasone) and adjuvant drugs used for neuropathic pain may act as P-gp substrates and modulate its expression, thus making pain management challenging. Inflammatory conditions are also believed to upregulate P-gp. The role of P-gp in drug-drug interactions is currently under investigation, since many P-gp substrates may also act as substrates for the cytochrome P450 enzymes, which metabolize a wide range of xenobiotics and endobiotics. Genetic variability of the ABCB1/MDR1 gene may be accountable for inter-individual variation in opioid-induced analgesia. P-gp also plays a role in the management of opioid-induced adverse effects, such as constipation. Peripherally acting mu-opioid receptors antagonists (PAMORAs), such as naloxegol and naldemedine, are substrates of P-gp, which prevent their penetration in the central nervous system. In our review, we explore the interactions between P-gp and opioidergic drugs, with their implications in clinical practice.
Collapse
|
23
|
Tseng KY, Wang HC, Cheng KF, Wang YH, Chang LL, Cheng KI. Sciatic Nerve Intrafascicular Injection Induces Neuropathy by Activating the Matrix Modulators MMP-9 and TIMP-1. Front Pharmacol 2022; 13:859982. [PMID: 35694244 PMCID: PMC9178525 DOI: 10.3389/fphar.2022.859982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 04/19/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Peripheral nerve block (PNB) under echo guidance may not prevent intrafascicular anesthetic injection-induced nerve injury. This study investigated whether unintended needle piercing alone, or the intrafascicular nerve injectant could induce neuropathy. Methods: 120 adult male Sprague-Dawley rats were divided into four groups: 1) group S, only the left sciatic nerve was exposed; 2) group InF-P, the left sciatic nerve was exposed and pierced with a 30 G needle; 3) group InF-S, left sciatic nerve was exposed and injected with saline (0.9% NaCl 30 µL); 4) group InF-R, left sciatic nerve was exposed and injected with 0.5% (5 mg/mL, 30 µL) ropivacaine. Behaviors of thermal and mechanical stimuli responses from hindpaws, sciatic nerve vascular permeability and tight junction protein expression, and macrophage infiltration were assessed. Pro-inflammatory cytokine expression and TIMP-1 and MMP-9 activation at the injection site and the swollen, and distal sites of the sciatic nerve were measured by cytokine array, western blotting, and immunofluorescence of POh14 and POD3. Results: Intrafascicular saline and ropivacaine into the sciatic nerve, but not needle piercing alone, significantly induced mechanical allodynia that lasted for seven days. In addition, the prior groups increased vascular permeability and macrophage infiltration, especially in the swollen site of the sciatic nerve. Thermal hypersensitivity was induced and lasted for only 3 days after intrafascicular saline injection. Obvious upregulation of TIMP-1 and MMP-9 on POh6 and POh14 occurred regardless of intrafascicular injection or needle piercing. Compared to the needle piercing group, the ratio of MMP-9/TIMP-1 was significantly higher in the intrafascicular injectant groups at the injected and swollen sites of the sciatic nerve. Although no gross changes in the expressions of tight junction proteins (TJPs) claudin-5 and ZO-1, the TJPs turned to apparent fragmentation and fenestration-like degenerative change in swollen endothelial cells and thickened microvessels. Conclusion: Intrafascicular nerve injection is a distinct mechanism that induces neuropathy. It is likely that the InF nerve injection-induced neuropathy was largely due to dramatic, but transient, increases in enzymatic activities of MMP-9 and activating TIMP-1 in the operated nerves. The changes in enzymatic activities then contributed to certain levels of extracellular matrix degradation, which leads to increases in endoneurial vascular permeability.
Collapse
Affiliation(s)
- Kuang-Yi Tseng
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Anesthesiology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hung-Chen Wang
- Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Kai-Feng Cheng
- Department of Microbiology and Immunology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yi-Hsuan Wang
- Department of Microbiology and Immunology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Lin-Li Chang
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Microbiology and Immunology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Kuang-I Cheng
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Anesthesiology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
24
|
Reuschel V, Scherlach C, Pfeifle C, Krause M, Struck MF, Hoffmann KT, Schob S. Treatment Effect of CT-Guided Periradicular Injections in Context of Different Contrast Agent Distribution Patterns. Diagnostics (Basel) 2022; 12:diagnostics12040787. [PMID: 35453835 PMCID: PMC9028051 DOI: 10.3390/diagnostics12040787] [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: 02/06/2022] [Revised: 03/08/2022] [Accepted: 03/18/2022] [Indexed: 02/01/2023] Open
Abstract
Acutely manifesting radicular pain syndromes associated with degenerations of the lower spine are frequent ailments with a high rate of recurrence. Part of the conservative management are periradicular infiltrations of analgesics and steroids. The purpose of this study is to evaluate the dependence of the clinical efficacy of CT-guided periradicular injections on the pattern of contrast distribution and to identify the best distribution pattern that is associated with the most effective pain relief. Using a prospective study design, 161 patients were included in this study, ensuring ethical standards. Statistical analysis was performed, with the level of statistical significance set at p = 0.05. A total of 37.9% of patients experienced significant but not long-lasting (four weeks on average) complete pain relief. A total of 44.1% of patients experienced prolonged, subjectively satisfying pain relief of more than four weeks to three months. A total of 18% of patients had complete and sustained relief for more than six months. A significant correlation exists between circumferential, large area contrast distribution including the zone of action between the disc and affected nerve root contrast distribution pattern with excellent pain relief. Our results support the value of CT-guided contrast injection for achieving a good efficacy, and, if necessary, indicative repositioning of the needle to ensure a circumferential distribution pattern of corticosteroids for the sufficient treatment of radicular pain in degenerative spine disease.
Collapse
Affiliation(s)
- Vera Reuschel
- Institut für Neuroradiologie, Universitätsklinikum Leipzig AöR, Liebigstr. 20, 04103 Leipzig, Germany; (V.R.); (C.S.); (K.-T.H.)
- Institut für Diagnostische und Interventionelle Neuroradiologie, Universitätsmedizin Göttingen, Robert-Koch-Str. 40, 37075 Göttingen, Germany
| | - Cordula Scherlach
- Institut für Neuroradiologie, Universitätsklinikum Leipzig AöR, Liebigstr. 20, 04103 Leipzig, Germany; (V.R.); (C.S.); (K.-T.H.)
| | - Christian Pfeifle
- Klinik und Poliklinik für Orthopädie, Unfallchirurgie und Plastische Chirurgie, Universitätsklinikum Leipzig AöR, Liebigstr. 20, 04103 Leipzig, Germany;
| | - Matthias Krause
- Klinik und Poliklinik für Neurochirurgie, Universitätsklinikum Leipzig AöR, Liebigstr. 20, 04103 Leipzig, Germany;
| | - Manuel Florian Struck
- Klinik und Poliklinik für Anästhesiologie und Intensivtherapie, Universitätsklinikum Leipzig AöR, Liebigstr. 20, 04103 Leipzig, Germany;
| | - Karl-Titus Hoffmann
- Institut für Neuroradiologie, Universitätsklinikum Leipzig AöR, Liebigstr. 20, 04103 Leipzig, Germany; (V.R.); (C.S.); (K.-T.H.)
| | - Stefan Schob
- Abteilung für Neuroradiologie, Universitätsklinik und Poliklinik für Radiologie, Universitätsklinikum Halle (Saale) Ernst-Grube-Str. 40, 06120 Halle (Saale), Germany
- Correspondence: ; Tel.: +49-34-5557-2432
| |
Collapse
|
25
|
Lee J, Choi H, Park C, Jeon S, Yune T. Jmjd3 Mediates Neuropathic Pain by Inducing Macrophage Infiltration and Activation in Lumbar Spinal Stenosis Animal Model. Int J Mol Sci 2021; 22:ijms222413426. [PMID: 34948220 PMCID: PMC8707917 DOI: 10.3390/ijms222413426] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 12/09/2021] [Accepted: 12/13/2021] [Indexed: 01/14/2023] Open
Abstract
Lumbar spinal stenosis (LSS) is a major cause of chronic neuropathic back and/or leg pain. Recently, we demonstrated that a significant number of macrophages infiltrated into the cauda equina after compression injury, causing neuroinflammation, and consequently mediating neuropathic pain development and/or maintenance. However, the molecular mechanisms underlying macrophage infiltration and activation have not been elucidated. Here, we demonstrated the critical role of histone H3K27 demethylase Jmjd3 in blood-nerve barrier dysfunction following macrophage infiltration and activation in LSS rats. The LSS rat model was induced by cauda equina compression using a silicone block within the epidural spaces of the L5-L6 vertebrae with neuropathic pain developing 4 weeks after compression. We found that Jmjd3 was induced in the blood vessels and infiltrated macrophages in a rat model of neuropathic pain. The blood-nerve barrier permeability in the cauda equina was increased after compression and significantly attenuated by the Jmjd3 demethylase inhibitor, GSK-J4. GSK-J4 also inhibited the expression and activation of MMP-2 and MMP-9 and significantly alleviated the loss of tight junction proteins and macrophage infiltration. Furthermore, the activation of a macrophage cell line, RAW 264.7, by LPS was significantly alleviated by GSK-J4. Finally, GSK-J4 and a potential Jmjd3 inhibitor, gallic acid, significantly inhibited mechanical allodynia in LSS rats. Thus, our findings suggest that Jmjd3 mediates neuropathic pain development and maintenance by inducing macrophage infiltration and activation after cauda equina compression and thus may serve as a potential therapeutic target for LSS-induced neuropathic pain.
Collapse
Affiliation(s)
- Jeeyoun Lee
- Age-Related and Brain Diseases Research Center, Kyung Hee University, Seoul 02447, Korea; (J.L.); (H.C.); (C.P.)
| | - Haeyoung Choi
- Age-Related and Brain Diseases Research Center, Kyung Hee University, Seoul 02447, Korea; (J.L.); (H.C.); (C.P.)
| | - Chansol Park
- Age-Related and Brain Diseases Research Center, Kyung Hee University, Seoul 02447, Korea; (J.L.); (H.C.); (C.P.)
| | - Sangryong Jeon
- Department of Neurological Surgery, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Korea;
| | - Taeyoung Yune
- Age-Related and Brain Diseases Research Center, Kyung Hee University, Seoul 02447, Korea; (J.L.); (H.C.); (C.P.)
- Department Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
- Correspondence: ; Tel.: +82-2-961-0968; Fax: +82-2-969-6343
| |
Collapse
|
26
|
Avraham O, Feng R, Ewan EE, Rustenhoven J, Zhao G, Cavalli V. Profiling sensory neuron microenvironment after peripheral and central axon injury reveals key pathways for neural repair. eLife 2021; 10:e68457. [PMID: 34586065 PMCID: PMC8480984 DOI: 10.7554/elife.68457] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 09/12/2021] [Indexed: 12/19/2022] Open
Abstract
Sensory neurons with cell bodies in dorsal root ganglia (DRG) represent a useful model to study axon regeneration. Whereas regeneration and functional recovery occurs after peripheral nerve injury, spinal cord injury or dorsal root injury is not followed by regenerative outcomes. Regeneration of sensory axons in peripheral nerves is not entirely cell autonomous. Whether the DRG microenvironment influences the different regenerative capacities after injury to peripheral or central axons remains largely unknown. To answer this question, we performed a single-cell transcriptional profiling of mouse DRG in response to peripheral (sciatic nerve crush) and central axon injuries (dorsal root crush and spinal cord injury). Each cell type responded differently to the three types of injuries. All injuries increased the proportion of a cell type that shares features of both immune cells and glial cells. A distinct subset of satellite glial cells (SGC) appeared specifically in response to peripheral nerve injury. Activation of the PPARα signaling pathway in SGC, which promotes axon regeneration after peripheral nerve injury, failed to occur after central axon injuries. Treatment with the FDA-approved PPARα agonist fenofibrate increased axon regeneration after dorsal root injury. This study provides a map of the distinct DRG microenvironment responses to peripheral and central injuries at the single-cell level and highlights that manipulating non-neuronal cells could lead to avenues to promote functional recovery after CNS injuries or disease.
Collapse
Affiliation(s)
- Oshri Avraham
- Department of Neuroscience, Washington University School of MedicineSaint LouisUnited States
| | - Rui Feng
- Department of Neuroscience, Washington University School of MedicineSaint LouisUnited States
| | - Eric Edward Ewan
- Department of Neuroscience, Washington University School of MedicineSaint LouisUnited States
| | - Justin Rustenhoven
- Department of Pathology and Immunology, Washington University School of MedicineSt LouisUnited States
- Center for Brain Immunology and Glia (BIG), Washington University School of MedicineSt LouisUnited States
| | - Guoyan Zhao
- Department of Neuroscience, Washington University School of MedicineSaint LouisUnited States
- Department of Pathology and Immunology, Washington University School of MedicineSt LouisUnited States
| | - Valeria Cavalli
- Department of Neuroscience, Washington University School of MedicineSaint LouisUnited States
- Center of Regenerative Medicine, Washington University School of MedicineSt. LouisUnited States
- Hope Center for Neurological Disorders, Washington University School of MedicineSt. LouisUnited States
| |
Collapse
|
27
|
Chen JTC, Schmidt L, Schürger C, Hankir MK, Krug SM, Rittner HL. Netrin-1 as a Multitarget Barrier Stabilizer in the Peripheral Nerve after Injury. Int J Mol Sci 2021; 22:ijms221810090. [PMID: 34576252 PMCID: PMC8466625 DOI: 10.3390/ijms221810090] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/14/2021] [Accepted: 09/16/2021] [Indexed: 02/06/2023] Open
Abstract
The blood–nerve barrier and myelin barrier normally shield peripheral nerves from potentially harmful insults. They are broken down during nerve injury, which contributes to neuronal damage. Netrin-1 is a neuronal guidance protein with various established functions in the peripheral and central nervous systems; however, its role in regulating barrier integrity and pain processing after nerve injury is poorly understood. Here, we show that chronic constriction injury (CCI) in Wistar rats reduced netrin-1 protein and the netrin-1 receptor neogenin-1 (Neo1) in the sciatic nerve. Replacement of netrin-1 via systemic or local administration of the recombinant protein rescued injury-induced nociceptive hypersensitivity. This was prevented by siRNA-mediated knockdown of Neo1 in the sciatic nerve. Mechanistically, netrin-1 restored endothelial and myelin, but not perineural, barrier function as measured by fluorescent dye or fibrinogen penetration. Netrin-1 also reversed the decline in the tight junction proteins claudin-5 and claudin-19 in the sciatic nerve caused by CCI. Our findings emphasize the role of the endothelial and myelin barriers in pain processing after nerve damage and reveal that exogenous netrin-1 restores their function to mitigate CCI-induced hypersensitivity via Neo1. The netrin-1-neogenin-1 signaling pathway may thus represent a multi-target barrier protector for the treatment of neuropathic pain.
Collapse
Affiliation(s)
- Jeremy Tsung-Chieh Chen
- Center for Interdisciplinary Pain Medicine, Department of Anesthesiology, Intensive Care, Emergency Medicine and Pain Therapy, University Hospital of Würzburg, 97080 Würzburg, Germany; (J.T.-C.C.); (L.S.); (C.S.)
| | - Lea Schmidt
- Center for Interdisciplinary Pain Medicine, Department of Anesthesiology, Intensive Care, Emergency Medicine and Pain Therapy, University Hospital of Würzburg, 97080 Würzburg, Germany; (J.T.-C.C.); (L.S.); (C.S.)
| | - Christina Schürger
- Center for Interdisciplinary Pain Medicine, Department of Anesthesiology, Intensive Care, Emergency Medicine and Pain Therapy, University Hospital of Würzburg, 97080 Würzburg, Germany; (J.T.-C.C.); (L.S.); (C.S.)
| | - Mohammed K. Hankir
- Department of Experimental Surgery, University Hospital of Würzburg, 97080 Würzburg, Germany;
| | - Susanne M. Krug
- Clinical Physiology/Nutritional Medicine, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, 12203 Berlin, Germany;
| | - Heike L. Rittner
- Center for Interdisciplinary Pain Medicine, Department of Anesthesiology, Intensive Care, Emergency Medicine and Pain Therapy, University Hospital of Würzburg, 97080 Würzburg, Germany; (J.T.-C.C.); (L.S.); (C.S.)
- Correspondence: ; Tel.: +49-931-201-30251
| |
Collapse
|
28
|
Ben-Kraiem A, Sauer RS, Norwig C, Popp M, Bettenhausen AL, Atalla MS, Brack A, Blum R, Doppler K, Rittner HL. Selective blood-nerve barrier leakiness with claudin-1 and vessel-associated macrophage loss in diabetic polyneuropathy. J Mol Med (Berl) 2021; 99:1237-1250. [PMID: 34018017 PMCID: PMC8367905 DOI: 10.1007/s00109-021-02091-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 04/26/2021] [Accepted: 05/10/2021] [Indexed: 12/19/2022]
Abstract
Diabetic polyneuropathy (DPN) is the most common complication in diabetes and can be painful in up to 26% of all diabetic patients. Peripheral nerves are shielded by the blood-nerve barrier (BNB) consisting of the perineurium and endoneurial vessels. So far, there are conflicting results regarding the role and function of the BNB in the pathophysiology of DPN. In this study, we analyzed the spatiotemporal tight junction protein profile, barrier permeability, and vessel-associated macrophages in Wistar rats with streptozotocin-induced DPN. In these rats, mechanical hypersensitivity developed after 2 weeks and loss of motor function after 8 weeks, while the BNB and the blood-DRG barrier were leakier for small, but not for large molecules after 8 weeks only. The blood-spinal cord barrier remained sealed throughout the observation period. No gross changes in tight junction protein or cytokine expression were observed in all barriers to blood. However, expression of Cldn1 mRNA in perineurium was specifically downregulated in conjunction with weaker vessel-associated macrophage shielding of the BNB. Our results underline the role of specific tight junction proteins and BNB breakdown in DPN maintenance and differentiate DPN from traumatic nerve injury. Targeting claudins and sealing the BNB could stabilize pain and prevent further nerve damage. KEY MESSAGES: • In diabetic painful neuropathy in rats: • Blood nerve barrier and blood DRG barrier are leaky for micromolecules. • Perineurial Cldn1 sealing the blood nerve barrier is specifically downregulated. • Endoneurial vessel-associated macrophages are also decreased. • These changes occur after onset of hyperalgesia thereby maintaining rather than inducing pain.
Collapse
Affiliation(s)
- Adel Ben-Kraiem
- Center for Interdisciplinary Pain Medicine, Department of Anesthesiology, University Hospital of Würzburg, 97080, Würzburg, Germany
| | - Reine-Solange Sauer
- Center for Interdisciplinary Pain Medicine, Department of Anesthesiology, University Hospital of Würzburg, 97080, Würzburg, Germany
| | - Carla Norwig
- Center for Interdisciplinary Pain Medicine, Department of Anesthesiology, University Hospital of Würzburg, 97080, Würzburg, Germany
| | - Maria Popp
- Center for Interdisciplinary Pain Medicine, Department of Anesthesiology, University Hospital of Würzburg, 97080, Würzburg, Germany
| | - Anna-Lena Bettenhausen
- Center for Interdisciplinary Pain Medicine, Department of Anesthesiology, University Hospital of Würzburg, 97080, Würzburg, Germany
| | - Mariam Sobhy Atalla
- Center for Interdisciplinary Pain Medicine, Department of Anesthesiology, University Hospital of Würzburg, 97080, Würzburg, Germany
| | - Alexander Brack
- Center for Interdisciplinary Pain Medicine, Department of Anesthesiology, University Hospital of Würzburg, 97080, Würzburg, Germany
| | - Robert Blum
- Institute of Clinical Neurobiology, University Hospital of Würzburg, 97078, Würzburg, Germany
- Department of Neurology, University Hospital of Würzburg, 97080, Würzburg, Germany
| | - Kathrin Doppler
- Department of Neurology, University Hospital of Würzburg, 97080, Würzburg, Germany
| | - Heike Lydia Rittner
- Center for Interdisciplinary Pain Medicine, Department of Anesthesiology, University Hospital of Würzburg, 97080, Würzburg, Germany.
| |
Collapse
|
29
|
Li W, Gong Y, Liu J, Guo Y, Tang H, Qin S, Zhao Y, Wang S, Xu Z, Chen B. Peripheral and Central Pathological Mechanisms of Chronic Low Back Pain: A Narrative Review. J Pain Res 2021; 14:1483-1494. [PMID: 34079363 PMCID: PMC8166276 DOI: 10.2147/jpr.s306280] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 05/01/2021] [Indexed: 12/31/2022] Open
Abstract
Chronic low back pain (CLBP), lasting >3 months, is the end result of multiple pathogenic factors. Unfortunately, little is known about CLBP pathogenesis, which limits its advancements in clinical therapy and disease management. This paper summarizes the known pathological axes of CLBP, involving both peripheral and central systems. In particular, this paper details injurious nerve stimulation, inflammation-induced peripheral pathway, and central sensitization. Lumbar components, such as intervertebral disc (IVD), facet joints, muscles, fascia, ligaments, and joint capsules, contain pain receptors called nociceptors. Degeneration of the aforementioned lumbar components activates inflammatory pathways, which can directly damage nerves, lower nociceptor threshold to fire action potentials (AP), and cause pain. Additionally, damaged lumbar IVDs and endplates can also lead to the pathologic invasion of nerve growth and innervation, followed by the compression of herniated IVDs on nerve roots, thereby causing traumatic neuropathic pain. The central mechanism of CLBP involves alteration of the sensory processing of the brain and malfunction of the descending pain modulatory system, which facilitates pain amplification in the center nervous system (CNS). Lastly, abnormalities in the brain biochemical metabolism, activation of glial cells, and subsequent inflammation also play important roles in CLBP development. Taken together, inflammation plays an important role in both peripheral and central sensitization of CLBP. Due to the heterogeneity of CLBP, its pathological mechanism remains complex and difficult to understand. Therefore, it is a worthy field for future research into the subcomponents of CLBP pathogenesis, in order to distinguish the specific form of the disease, identify its origins, and develop corresponding highly effective comprehensive therapy against CLBP.
Collapse
Affiliation(s)
- Wei Li
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Yinan Gong
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Jingyi Liu
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Yongming Guo
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China.,School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, People's Republic of China
| | - Huiling Tang
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Siru Qin
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Yadan Zhao
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Songtao Wang
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Zhifang Xu
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China.,School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, People's Republic of China
| | - Bo Chen
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China.,School of Acupuncture & Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, People's Republic of China
| |
Collapse
|
30
|
Montnach J, De Waard S, Nicolas S, Burel S, Osorio N, Zoukimian C, Mantegazza M, Boukaiba R, Béroud R, Partiseti M, Delmas P, Marionneau C, De Waard M. Fluorescent- and tagged-protoxin II peptides: potent markers of the Na v 1.7 channel pain target. Br J Pharmacol 2021; 178:2632-2650. [PMID: 33742442 DOI: 10.1111/bph.15453] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 01/26/2021] [Accepted: 02/09/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND AND PURPOSE Protoxin II (ProTx II) is a high affinity gating modifier that is thought to selectively block the Nav 1.7 voltage-dependent Na+ channel, a major therapeutic target for the control of pain. We aimed at producing ProTx II analogues entitled with novel functionalities for cell distribution studies and biochemical characterization of its Nav channel targets. EXPERIMENTAL APPROACH We took advantage of the high affinity properties of the peptide, combined to its slow off rate, to design a number of new tagged analogues useful for imaging and biochemistry purposes. We used high-throughput automated patch-clamp to identify the analogues best matching the native properties of ProTx II and validated them on various Nav -expressing cells in pull-down and cell distribution studies. KEY RESULTS Two of the produced ProTx II analogues, Biot-ProTx II and ATTO488-ProTx II, best emulate the pharmacological properties of unlabelled ProTx II, whereas other analogues remain high affinity blockers of Nav 1.7. The biotinylated version of ProTx II efficiently works for the pull-down of several Nav isoforms tested in a concentration-dependent manner, whereas the fluorescent ATTO488-ProTx II specifically labels the Nav 1.7 channel over other Nav isoforms tested in various experimental conditions. CONCLUSIONS AND IMPLICATIONS The properties of these ProTx II analogues as tools for Nav channel purification and cell distribution studies pave the way for a better understanding of ProTx II channel receptors in pain and their pathophysiological implications in sensory neuronal processing. The new fluorescent ProTx II should also be useful in the design of new drug screening strategies.
Collapse
Affiliation(s)
- Jérôme Montnach
- LabEx "Ion Channels, Science & Therapeutics", l'institut du thorax, INSERM, CNRS, UNIV NANTES, Nantes, France
| | - Stephan De Waard
- LabEx "Ion Channels, Science & Therapeutics", l'institut du thorax, INSERM, CNRS, UNIV NANTES, Nantes, France
| | - Sébastien Nicolas
- LabEx "Ion Channels, Science & Therapeutics", l'institut du thorax, INSERM, CNRS, UNIV NANTES, Nantes, France
| | - Sophie Burel
- LabEx "Ion Channels, Science & Therapeutics", l'institut du thorax, INSERM, CNRS, UNIV NANTES, Nantes, France
| | - Nancy Osorio
- Laboratory of Cognitive Neuroscience, UMR 7291, CNRS, Aix-Marseille University, Marseille, France
| | | | - Massimo Mantegazza
- Université Cote d'Azur, CNRS UMR 7275, Institute of Molecular and Cellular Pharmacology, Valbonne-Sophia Antipolis, France
| | - Rachid Boukaiba
- Sanofi R&D, Integrated Drug Discovery - High Content Biology, Vitry-sur-Seine, France
| | | | - Michel Partiseti
- Sanofi R&D, Integrated Drug Discovery - High Content Biology, Vitry-sur-Seine, France
| | - Patrick Delmas
- Laboratory of Cognitive Neuroscience, UMR 7291, CNRS, Aix-Marseille University, Marseille, France
| | - Céline Marionneau
- LabEx "Ion Channels, Science & Therapeutics", l'institut du thorax, INSERM, CNRS, UNIV NANTES, Nantes, France
| | - Michel De Waard
- LabEx "Ion Channels, Science & Therapeutics", l'institut du thorax, INSERM, CNRS, UNIV NANTES, Nantes, France.,Smartox Biotechnology, Saint-Egrève, France
| |
Collapse
|
31
|
Differential expression of cerebrospinal fluid neuroinflammatory mediators depending on osteoarthritis pain phenotype. Pain 2021; 161:2142-2154. [PMID: 32384383 PMCID: PMC7431139 DOI: 10.1097/j.pain.0000000000001903] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 04/21/2020] [Indexed: 12/15/2022]
Abstract
Supplemental Digital Content is Available in the Text. Distinct cerebrospinal fluid neuroinflammatory profiles may be associated with different objective characteristics of persistent pain in osteoarthritis patients undergoing total hip arthroplasty. Neuroinflammation is implicated in the development and maintenance of persistent pain states, but there are limited data linking cerebrospinal fluid (CSF) inflammatory mediators with neurophysiological pain processes in humans. In a prospective observational study, CSF inflammatory mediators were compared between patients with osteoarthritis (OA) who were undergoing total hip arthroplasty due to disabling pain symptoms (n = 52) and pain-free comparison controls (n = 30). In OA patients only, detailed clinical examination and quantitative sensory testing were completed. Cerebrospinal fluid samples were analyzed for 10 proinflammatory mediators using Meso Scale Discovery platform. Compared to controls, OA patients had higher CSF levels of interleukin 8 (IL-8) (P = 0.002), intercellular adhesion molecule 1 (P = 0.007), and vascular cell adhesion molecule 1 (P = 0.006). Osteoarthritis patients with central sensitization possibly indicated by arm pressure pain detection threshold <250 kPa showed significantly higher CSF levels of Fms-related tyrosine kinase 1 (Flt-1) (P = 0.044) and interferon gamma-induced protein 10 (IP-10) (P = 0.024), as compared to subjects with PPDT above that threshold. In patients reporting pain numerical rating scale score ≥3/10 during peripheral venous cannulation, Flt-1 was elevated (P = 0.025), and in patients with punctate stimulus wind-up ratio ≥2, CSF monocyte chemoattractant protein 1 was higher (P = 0.011). Multiple logistic regression models showed that increased Flt-1 was associated with central sensitization, assessed by remote-site PPDT and peripheral venous cannulation pain, and monocyte chemoattractant protein-1 with temporal summation in the area of maximum pain. Multiple proinflammatory mediators measured in CSF are associated with persistent hip OA-related pain. Pain phenotype may be influenced by specific CSF neuroinflammatory profiles.
Collapse
|
32
|
Uversky VN, Elrashdy F, Aljadawi A, Ali SM, Khan RH, Redwan EM. Severe acute respiratory syndrome coronavirus 2 infection reaches the human nervous system: How? J Neurosci Res 2021; 99:750-777. [PMID: 33217763 PMCID: PMC7753416 DOI: 10.1002/jnr.24752] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 10/19/2020] [Accepted: 10/21/2020] [Indexed: 02/06/2023]
Abstract
Without protective and/or therapeutic agents the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection known as coronavirus disease 2019 is quickly spreading worldwide. It has surprising transmissibility potential, since it could infect all ages, gender, and human sectors. It attacks respiratory, gastrointestinal, urinary, hepatic, and endovascular systems and can reach the peripheral nervous system (PNS) and central nervous system (CNS) through known and unknown mechanisms. The reports on the neurological manifestations and complications of the SARS-CoV-2 infection are increasing exponentially. Herein, we enumerate seven candidate routes, which the mature or immature SARS-CoV-2 components could use to reach the CNS and PNS, utilizing the within-body cross talk between organs. The majority of SARS-CoV-2-infected patients suffer from some neurological manifestations (e.g., confusion, anosmia, and ageusia). It seems that although the mature virus did not reach the CNS or PNS of the majority of patients, its unassembled components and/or the accompanying immune-mediated responses may be responsible for the observed neurological symptoms. The viral particles and/or its components have been specifically documented in endothelial cells of lung, kidney, skin, and CNS. This means that the blood-endothelial barrier may be considered as the main route for SARS-CoV-2 entry into the nervous system, with the barrier disruption being more logical than barrier permeability, as evidenced by postmortem analyses.
Collapse
Affiliation(s)
- Vladimir N. Uversky
- Biological Science DepartmentFaculty of ScienceKing Abdulaziz UniversityJeddahSaudi Arabia
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of MedicineUniversity of South FloridaTampaFLUSA
- Institute for Biological Instrumentation of the Russian Academy of SciencesFederal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”PushchinoRussia
| | - Fatma Elrashdy
- Department of Endemic Medicine and HepatogastroenterologyKasr Alainy School of MedicineCairo UniversityCairoEgypt
| | - Abdullah Aljadawi
- Biological Science DepartmentFaculty of ScienceKing Abdulaziz UniversityJeddahSaudi Arabia
| | - Syed Moasfar Ali
- Interdisciplinary Biotechnology UnitAligarh Muslim UniversityAligarhIndia
| | - Rizwan Hasan Khan
- Interdisciplinary Biotechnology UnitAligarh Muslim UniversityAligarhIndia
| | - Elrashdy M. Redwan
- Biological Science DepartmentFaculty of ScienceKing Abdulaziz UniversityJeddahSaudi Arabia
| |
Collapse
|
33
|
Shreckengost J, Halder M, Mena-Avila E, Garcia-Ramirez DL, Quevedo J, Hochman S. Nicotinic receptor modulation of primary afferent excitability with selective regulation of Aδ-mediated spinal actions. J Neurophysiol 2020; 125:568-585. [PMID: 33326305 DOI: 10.1152/jn.00228.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Somatosensory input strength can be modulated by primary afferent depolarization (PAD) generated predominantly via presynaptic GABAA receptors on afferent terminals. We investigated whether ionotropic nicotinic acetylcholine receptors (nAChRs) also provide modulatory actions, focusing on myelinated afferent excitability in in vitro murine spinal cord nerve-attached models. Primary afferent stimulation-evoked synaptic transmission was recorded in the deep dorsal horn as extracellular field potentials (EFPs), whereas concurrently recorded dorsal root potentials (DRPs) were used as an indirect measure of PAD. Changes in afferent membrane excitability were simultaneously measured as direct current (DC)-shifts in membrane polarization recorded in dorsal roots or peripheral nerves. The broad nAChR antagonist d-tubocurarine (d-TC) selectively and strongly depressed Aδ-evoked synaptic EFPs (36% of control) coincident with similarly depressed A-fiber DRP (43% of control), whereas afferent electrical excitability remained unchanged. In comparison, acetylcholine (ACh) and the nAChR agonists, epibatidine and nicotine, reduced afferent excitability by generating coincident depolarizing DC-shifts in peripheral axons and intraspinally. Progressive depolarization corresponded temporally with the emergence of spontaneous axonal spiking and reductions in the DRP and all afferent-evoked synaptic actions (31%-37% of control). Loss of evoked response was long-lasting, independent of DC repolarization, and likely due to mechanisms initiated by spontaneous C-fiber activity. DC-shifts were blocked with d-TC but not GABAA receptor blockers and retained after tetrodotoxin block of voltage-gated Na+ channels. Notably, actions tested were comparable between three mouse strains, in rat, and when performed in different labs. Thus, nAChRs can regulate afferent excitability via two distinct mechanisms: by central Aδ-afferent actions, and by transient extrasynaptic axonal activation of high-threshold primary afferents.NEW & NOTEWORTHY Primary afferents express many nicotinic ACh receptor (nAChR) subtypes but whether activation is linked to presynaptic inhibition, facilitation, or more complex and selective activity modulation is unknown. Recordings of afferent-evoked responses in the lumbar spinal cord identified two nAChR-mediated modulatory actions: 1) selective control of Aδ afferent transmission and 2) robust changes in axonal excitability initiated via extrasynaptic shifts in DC polarization. This work broadens the diversity of presynaptic modulation of primary afferents by nAChRs.
Collapse
Affiliation(s)
| | - Mallika Halder
- Department of Physiology, Emory University, Atlanta, Georgia
| | - Elvia Mena-Avila
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, México City, México
| | - David Leonardo Garcia-Ramirez
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, México City, México
| | - Jorge Quevedo
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, México City, México
| | - Shawn Hochman
- Department of Physiology, Emory University, Atlanta, Georgia
| |
Collapse
|
34
|
Nerve Lesions in Volkmann Ischemic Contracture. J Hand Surg Am 2020; 45:746-757. [PMID: 32600789 DOI: 10.1016/j.jhsa.2020.03.027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 07/29/2019] [Accepted: 03/09/2020] [Indexed: 02/02/2023]
Abstract
Volkmann ischemic contracture (VIC) is a late sequela of acute compartment syndrome and consists of extensive muscle necrosis, fibrosis, contracture, and variable degrees of neural dysfunction. The outcome depends on successful restoration of muscle and neural function. The timing of surgery is often determined by the development of contractures and is delayed in the interest of observing some spontaneous recovery and infarct maturation. This period of observation may be detrimental to nerve function with gradual formation of scar tissue and worsening constrictive neuropathy. Early intervention appears to be more effective in preventing further nerve damage and restoring protective sensation. In this article, common features of compartment syndrome, frequently seen nerve lesions, and the effect of timing of surgical intervention on the outcome of VIC were reviewed in the light of the current basic and clinical science literature.
Collapse
|
35
|
Lee GW, Son JY, Lee AR, Ju JS, Bae YC, Ahn DK. Central VEGF-A pathway plays a key role in the development of trigeminal neuropathic pain in rats. Mol Pain 2020; 15:1744806919872602. [PMID: 31397622 PMCID: PMC6719481 DOI: 10.1177/1744806919872602] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The study reported here investigated the role of the central vascular endothelial growth factor-A (VEGF-A) pathway in the development of trigeminal neuropathic pain following nerve injury. A Sprague-Dawley rat model of trigeminal neuropathic pain was produced using malpositioned dental implants. The left mandibular second molar was extracted under anesthesia and replaced with a miniature dental implant to induce injury to the inferior alveolar nerve. The inferior alveolar nerve injury produced a significant upregulation of astrocytic VEGF-A expression in the medullary dorsal horn. The nerve injury-induced mechanical allodynia was inhibited by an intracisternal infusion of VEGF-A164 antibody. Although both VEGF-A Receptor 1 (VEGF-A R1; colocalized with the blood–brain barrier) and VEGF-A Receptor 2 (VEGF-A R2; colocalized with astrocytes) participated in the development of trigeminal neuropathic pain following nerve injury, only the intracisternal infusion of a VEGF-A R1 antibody, and not that of a VEGF-A R2 antibody, inhibited the increased blood–brain barrier permeability produced by nerve injury. Finally, we confirmed the participation of the central VEGF-A pathway in the development of trigeminal neuropathic pain by reducing VEGF-A expression using VEGF-A164 siRNA. This suppression of VEGF-A produced significant prolonged anti-allodynic effects. These results suggest that the central VEGF-A pathway plays a key role in the development of trigeminal neuropathic pain following nerve injury through two separate pathways: VEGF-A R1 and VEGF-A R2. Hence, a blockade of the central VEGF-A pathway provides a new therapeutic avenue for the treatment of trigeminal neuropathic pain.
Collapse
Affiliation(s)
- Geun W Lee
- 1 Department of Oral Physiology, School of Dentistry, Kyungpook National University, Daegu, Korea
| | - Jo Y Son
- 1 Department of Oral Physiology, School of Dentistry, Kyungpook National University, Daegu, Korea
| | - Ah R Lee
- 1 Department of Oral Physiology, School of Dentistry, Kyungpook National University, Daegu, Korea
| | - Jin S Ju
- 1 Department of Oral Physiology, School of Dentistry, Kyungpook National University, Daegu, Korea
| | - Yong C Bae
- 2 Oral Anatomy, School of Dentistry, Kyungpook National University, Daegu, Korea
| | - Dong K Ahn
- 1 Department of Oral Physiology, School of Dentistry, Kyungpook National University, Daegu, Korea
| |
Collapse
|
36
|
The geriatric pain experience in mice: intact cutaneous thresholds but altered responses to tonic and chronic pain. Neurobiol Aging 2020; 89:1-11. [DOI: 10.1016/j.neurobiolaging.2019.12.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 11/29/2019] [Accepted: 12/19/2019] [Indexed: 11/23/2022]
|
37
|
Fujino K, Yokota A, Ohno K, Hirofuji S, Neo M. Impairment and restoration of the blood-nerve barrier and its correlation with pain following gradual nerve elongation of the rat sciatic nerve. Int J Neurosci 2020; 131:254-263. [PMID: 32167000 DOI: 10.1080/00207454.2020.1738430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Purpose: This study aimed to evaluate the time course of impairment and restoration of the blood-nerve barrier (BNB) following gradual elongation of the sciatic nerve and to clarify its association with nociception.Materials and Methods: The right femur was lengthened at a rate of 1.5 mm/day for 10 days. Von Frey tests were performed until 50 days after lengthening. Compound muscle action potentials (CMAPs) were measured to assess gross dysfunction of the elongated nerve. Evans blue-albumin tracing and immunohistochemistry for endothelial barrier antigen (EBA), rat endothelial cell antigen-1 (RECA-1), and CD68 for qualitative and quantitative analysis of the BNB and macrophage infiltration were performed for up to 50 days after cessation of lengthening in three segments of the sciatic nerves.Results: Paw-withdrawal threshold was significantly decreased at 7 days from initiation and began to recover from day 25 after lengthening. CMAPs showed delayed latency and attenuated amplitude but recovered at day 30 after cessation. On days 10 and 30 after cessation, spotted leakage of Evans blue-albumin in the endoneurium was observed, and the ratio of EBA/RECA-1-positive microvessels was significantly decreased, which subsequently recovered simultaneously in all segments on day 50 after cessation. Macrophages did not infiltrate the BNB at any time point.Conclusion: The restoration of BNB function following gradual nerve elongation was associated with the resolution of mechanical allodynia. Our findings provide insight into the association between nerve stretch injury and chronic nociception in adult male rats, which are potentially relevant to human orthopedic procedures and chronic neuropathic pain.
Collapse
Affiliation(s)
- Keitaro Fujino
- Department of Orthopedic Surgery, Osaka Medical College, Osaka, Japan
| | - Atsushi Yokota
- Department of Orthopedic Surgery, Osaka Medical College, Osaka, Japan
| | - Katsunori Ohno
- Department of Orthopedic Surgery, Osaka Medical College, Osaka, Japan
| | - Shinji Hirofuji
- Department of Orthopedic Surgery, Osaka Medical College, Osaka, Japan
| | - Masashi Neo
- Department of Orthopedic Surgery, Osaka Medical College, Osaka, Japan
| |
Collapse
|
38
|
Biology of the human blood-nerve barrier in health and disease. Exp Neurol 2020; 328:113272. [PMID: 32142802 DOI: 10.1016/j.expneurol.2020.113272] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 02/29/2020] [Accepted: 03/02/2020] [Indexed: 12/13/2022]
Abstract
A highly regulated endoneurial microenvironment is required for normal axonal function in peripheral nerves and nerve roots, which structurally consist of an outer collagenous epineurium, inner perineurium consisting of multiple concentric layers of specialized epithelioid myofibroblasts that surround the innermost endoneurium, which consists of myelinated and unmyelinated axons embedded in a looser mesh of collagen fibers. Endoneurial homeostasis is achieved by tight junction-forming endoneurial microvessels that control ion, solute, water, nutrient, macromolecule and leukocyte influx and efflux between the bloodstream and endoneurium, and the innermost layers of the perineurium that control interstitial fluid component flux between the freely permeable epineurium and endoneurium. Strictly speaking, endoneurial microvascular endothelium should be considered the blood-nerve barrier (BNB) due to direct communication with circulating blood. The mammalian BNB is considered the second most restrictive vascular system after the blood-brain barrier (BBB) based on classic in situ permeability studies. Structural alterations in endoneurial microvessels or interactions with hematogenous leukocytes have been described in several human peripheral neuropathies; however major advances in BNB biology in health and disease have been limited over the past 50 years. Guided by transcriptome and proteome studies of normal and pathologic human peripheral nerves, purified primary and immortalized human endoneurial endothelial cells that form the BNB and leukocytes from patients with well-characterized peripheral neuropathies, validated by in situ or ex vivo protein expression studies, data are emerging on the molecular and functional characteristics of the human BNB in health and in specific peripheral neuropathies, as well as chronic neuropathic pain. These early advancements have the potential to not only increase our understanding of how the BNB works and adapts or fails to adapt to varying insult, but provide insights relevant to pathogenic leukocyte trafficking, with translational potential and specific therapeutic application for chronic peripheral neuropathies and neuropathic pain.
Collapse
|
39
|
Abstract
Chronic pain is a major clinical problem of which the mechanisms are incompletely understood. Here, we describe the concept that PI16, a protein of unknown function mainly produced by fibroblasts, controls neuropathic pain. The spared nerve injury (SNI) model of neuropathic pain increases PI16 protein levels in fibroblasts in dorsal root ganglia (DRG) meninges and in the epi/perineurium of the sciatic nerve. We did not detect PI16 expression in neurons or glia in spinal cord, DRG, and nerve. Mice deficient in PI16 are protected against neuropathic pain. In vitro, PI16 promotes transendothelial leukocyte migration. In vivo, Pi16 -/- mice show reduced endothelial barrier permeability, lower leukocyte infiltration and reduced activation of the endothelial barrier regulator MLCK, and reduced phosphorylation of its substrate MLC2 in response to SNI. In summary, our findings support a model in which PI16 promotes neuropathic pain by mediating a cross-talk between fibroblasts and the endothelial barrier leading to barrier opening, cellular influx, and increased pain. Its key role in neuropathic pain and its limited cellular and tissue distribution makes PI16 an attractive target for pain management.
Collapse
|
40
|
Reinhold AK, Rittner HL. Characteristics of the nerve barrier and the blood dorsal root ganglion barrier in health and disease. Exp Neurol 2020; 327:113244. [PMID: 32057794 DOI: 10.1016/j.expneurol.2020.113244] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 01/17/2020] [Accepted: 02/09/2020] [Indexed: 12/14/2022]
Abstract
A variety of barriers ensures the protection of the peripheral nervous system from noxious blood-borne or surrounding stimuli. In this review, anatomy and functioning of the blood nerve barrier (BNB) and the blood DRG barrier (BDB) will be presented and key tight junction proteins described: ZO-1, claudin-1, -3, -5, -11, -12, -19, occludin, and tricellulin. Different diseases can lead to or be accompanied by nerve barrier disruption; impairment of nerve barriers in turn worsens pathology. Peripheral nerve injury, diabetic neuropathy and inflammatory polyneuropathy cause an increased permeability of BNB and BDB. Knowledge and understanding of these mechanisms might ultimately lead to the invention of drugs to control barrier function and help ameliorating neurological diseases.
Collapse
Affiliation(s)
- A K Reinhold
- Dept Anesthesiology, Center for Interdisciplinary Pain Medicine, University Hospitals Wuerzburg, Germany
| | - H L Rittner
- Dept Anesthesiology, Center for Interdisciplinary Pain Medicine, University Hospitals Wuerzburg, Germany.
| |
Collapse
|
41
|
Trevisan AJ, Bauer MB, Brindley RL, Currie KPM, Carter BD. Jedi-1 deficiency increases sensory neuron excitability through a non-cell autonomous mechanism. Sci Rep 2020; 10:1300. [PMID: 31992767 PMCID: PMC6987110 DOI: 10.1038/s41598-020-57971-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 12/12/2019] [Indexed: 12/14/2022] Open
Abstract
The dorsal root ganglia (DRG) house the primary afferent neurons responsible for somatosensation, including pain. We previously identified Jedi-1 (PEAR1/MEGF12) as a phagocytic receptor expressed by satellite glia in the DRG involved in clearing apoptotic neurons during development. Here, we further investigated the function of this receptor in vivo using Jedi-1 null mice. In addition to satellite glia, we found Jedi-1 expression in perineurial glia and endothelial cells, but not in sensory neurons. We did not detect any morphological or functional changes in the glial cells or vasculature of Jedi-1 knockout mice. Surprisingly, we did observe changes in DRG neuron activity. In neurons from Jedi-1 knockout (KO) mice, there was an increase in the fraction of capsaicin-sensitive cells relative to wild type (WT) controls. Patch-clamp electrophysiology revealed an increase in excitability, with a shift from phasic to tonic action potential firing patterns in KO neurons. We also found alterations in the properties of voltage-gated sodium channel currents in Jedi-1 null neurons. These results provide new insight into the expression pattern of Jedi-1 in the peripheral nervous system and indicate that loss of Jedi-1 alters DRG neuron activity indirectly through an intercellular interaction between non-neuronal cells and sensory neurons.
Collapse
Affiliation(s)
- Alexandra J Trevisan
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Mary Beth Bauer
- Anesthesiology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Rebecca L Brindley
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, USA
| | - Kevin P M Currie
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, USA.
| | - Bruce D Carter
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA.
- Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, TN, USA.
| |
Collapse
|
42
|
Regional Differences in Tight Junction Protein Expression in the Blood-DRG Barrier and Their Alterations after Nerve Traumatic Injury in Rats. Int J Mol Sci 2019; 21:ijms21010270. [PMID: 31906086 PMCID: PMC6981987 DOI: 10.3390/ijms21010270] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 12/23/2019] [Accepted: 12/28/2019] [Indexed: 12/15/2022] Open
Abstract
The nervous system is shielded by special barriers. Nerve injury results in blood–nerve barrier breakdown with downregulation of certain tight junction proteins accompanying the painful neuropathic phenotype. The dorsal root ganglion (DRG) consists of a neuron-rich region (NRR, somata of somatosensory and nociceptive neurons) and a fibre-rich region (FRR), and their putative epi-/perineurium (EPN). Here, we analysed blood–DRG barrier (BDB) properties in these physiologically distinct regions in Wistar rats after chronic constriction injury (CCI). Cldn5, Cldn12, and Tjp1 (rats) mRNA were downregulated 1 week after traumatic nerve injury. Claudin-1 immunoreactivity (IR) found in the EPN, claudin-19-IR in the FRR, and ZO-1-IR in FRR-EPN were unaltered after CCI. However, laser-assisted, vessel specific qPCR, and IR studies confirmed a significant loss of claudin-5 in the NRR. The NRR was three-times more permeable compared to the FRR for high and low molecular weight markers. NRR permeability was not further increased 1-week after CCI, but significantly more CD68+ macrophages had migrated into the NRR. In summary, NRR and FRR are different in naïve rats. Short-term traumatic nerve injury leaves the already highly permeable BDB in the NRR unaltered for small and large molecules. Claudin-5 is downregulated in the NRR. This could facilitate macrophage invasion, and thereby neuronal sensitisation and hyperalgesia. Targeting the stabilisation of claudin-5 in microvessels and the BDB barrier could be a future approach for neuropathic pain therapy.
Collapse
|
43
|
Madison RD, Robinson GA. Muscle-Derived Extracellular Vesicles Influence Motor Neuron Regeneration Accuracy. Neuroscience 2019; 419:46-59. [PMID: 31454553 DOI: 10.1016/j.neuroscience.2019.08.028] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 08/13/2019] [Accepted: 08/14/2019] [Indexed: 01/06/2023]
Abstract
Extracellular vesicles are lipid bilayer-enclosed extracellular structures. Although the term extracellular vesicles is quite inclusive, it generally refers to exosomes (<200 nm), and microvesicles (~100-1000 nm). Such vesicles are resistant to degradation and can contain proteins, lipids, and nucleic acids. Although it was previously thought that the primary purpose of such vesicles was to rid cells of unwanted components, it is now becoming increasingly clear that they can function as intercellular messengers, sometimes operating over long distances. As such, there is now intense interest in extracellular vesicles in fields as diverse as immunology, cell biology, cancer, and more recently, neuroscience. The influence that such extracellular vesicles might exert on peripheral nerve regeneration is just beginning to be investigated. In the current studies we show that muscle-derived extracellular vesicles significantly influence the anatomical accuracy of motor neuron regeneration in the rat femoral nerve. These findings suggest a basic cellular mechanism by which target end-organs could guide their own reinnervation following nerve injury.
Collapse
Affiliation(s)
- Roger D Madison
- Research Service of the Veterans Affairs Medical Center, Durham, NC 27705, USA; Department of Neurosurgery, Duke University Medical Center, Durham, NC 27710, USA.
| | - Grant A Robinson
- Department of Neurosurgery, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
44
|
Krock E, Millecamps M, Anderson KM, Srivastava A, Reihsen TE, Hari P, Sun YR, Jang SH, Wilcox GL, Belani KG, Beebe DS, Ouellet J, Pinto MR, Kehl LJ, Haglund L, Stone LS. Interleukin-8 as a therapeutic target for chronic low back pain: Upregulation in human cerebrospinal fluid and pre-clinical validation with chronic reparixin in the SPARC-null mouse model. EBioMedicine 2019; 43:487-500. [PMID: 31047862 PMCID: PMC6558025 DOI: 10.1016/j.ebiom.2019.04.032] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 03/22/2019] [Accepted: 04/17/2019] [Indexed: 12/18/2022] Open
Abstract
Background Low back pain (LBP) is the leading global cause of disability and is associated with intervertebral disc degeneration (DD) in some individuals. However, many adults have DD without LBP. Understanding why DD is painful in some and not others may unmask novel therapies for chronic LBP. The objectives of this study were to a) identify factors in human cerebrospinal fluid (CSF) associated with chronic LBP and b) examine their therapeutic utility in a proof-of-concept pre-clinical study. Methods Pain-free human subjects without DD, pain-free human subjects with DD, and patients with chronic LBP linked to DD were recruited and lumbar MRIs, pain and disability levels were obtained. CSF was collected and analyzed by multiplex cytokine assay. Interleukin-8 (IL-8) expression was confirmed by ELISA in CSF and in intervertebral discs. The SPARC-null mouse model of progressive, age-dependent DD and chronic LBP was used for pre-clinical validation. Male SPARC-null and control mice received systemic Reparixin, a CXCR1/2 (receptors for IL-8 and murine analogues) inhibitor, for 8 weeks. Behavioral signs of axial discomfort and radiating pain were assessed. Following completion of the study, discs were excised and cultured, and conditioned media was evaluated with a protein array. Findings IL-8 was elevated in CSF of chronic LBP patients with DD compared to pain-free subjects with or without DD. Chronic inhibition with reparixin alleviated low back pain behaviors and attenuated disc inflammation in SPARC-null mice. Interpretation These studies suggest that the IL-8 signaling pathway is a viable therapy for chronic LBP. Fund Supported by NIH, MMF, CIHR and FRQS.
Collapse
Affiliation(s)
- Emerson Krock
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, Quebec H3A 0G1, Canada; McGill Scoliosis and Spine Research Group, McGill University, Montreal, Quebec H3A 1G1, Canada; Faculty of Medicine, Department of Surgery, Orthopaedic Research Lab, McGill University, Montreal, Quebec H3A 1G1, Canada.
| | - Magali Millecamps
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, Quebec H3A 0G1, Canada; McGill Scoliosis and Spine Research Group, McGill University, Montreal, Quebec H3A 1G1, Canada; Faculty of Dentistry, McGill University, Montreal, Quebec H3A 1G1, Canada.
| | - Kathleen M Anderson
- Division of Physical Therapy, Department of Rehabilitation Medicine, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Akanksha Srivastava
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, Quebec H3A 0G1, Canada; Faculty of Dentistry, McGill University, Montreal, Quebec H3A 1G1, Canada.
| | - Troy E Reihsen
- Department of Anesthesiology, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Pawan Hari
- Department of Epidemiology, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Yue Ran Sun
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, Quebec H3A 0G1, Canada; Faculty of Dentistry, McGill University, Montreal, Quebec H3A 1G1, Canada.
| | - Seon Ho Jang
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, Quebec H3A 0G1, Canada; Faculty of Dentistry, McGill University, Montreal, Quebec H3A 1G1, Canada.
| | - George L Wilcox
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Kumar G Belani
- Department of Anesthesiology, University of Minnesota, Minneapolis, MN 55455, USA.
| | - David S Beebe
- Department of Anesthesiology, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Jean Ouellet
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, Quebec H3A 0G1, Canada; McGill Scoliosis and Spine Research Group, McGill University, Montreal, Quebec H3A 1G1, Canada; Faculty of Medicine, Department of Surgery, Orthopaedic Research Lab, McGill University, Montreal, Quebec H3A 1G1, Canada; Shriner's Hospital for Children, 1003 Decarie Blvd, Montreal, Quebec H4A 0A9, Canada
| | | | - Lois J Kehl
- Minnesota Head & Neck Pain Clinic, St. Paul, MN 55114, USA.
| | - Lisbet Haglund
- McGill Scoliosis and Spine Research Group, McGill University, Montreal, Quebec H3A 1G1, Canada; Faculty of Medicine, Department of Surgery, Orthopaedic Research Lab, McGill University, Montreal, Quebec H3A 1G1, Canada; Shriner's Hospital for Children, 1003 Decarie Blvd, Montreal, Quebec H4A 0A9, Canada.
| | - Laura S Stone
- Alan Edwards Centre for Research on Pain, McGill University, Montreal, Quebec H3A 0G1, Canada; McGill Scoliosis and Spine Research Group, McGill University, Montreal, Quebec H3A 1G1, Canada; Faculty of Dentistry, McGill University, Montreal, Quebec H3A 1G1, Canada; Faculty of Medicine, Anesthesia Research Unit, Montreal, Montreal, Quebec H3A 1G1, Canada; Faculty of Medicine, Department of Pharmacology and Therapeutics, Montreal, Quebec H3A 1G1, Canada.
| |
Collapse
|
45
|
Escaloni J, Young I, Loss J. Cupping with neural glides for the management of peripheral neuropathic plantar foot pain: a case study. J Man Manip Ther 2019; 27:54-61. [PMID: 30692843 DOI: 10.1080/10669817.2018.1514355] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Background/purpose: Plantar foot pain of neural origin is a challenging diagnosis to identify and treat. The purpose of this paper is to illustrate the novel way in which cupping was utilized in conjunction with neural glides to better diagnose and manage a patient who presented with symptoms of peripheral neuropathic plantar foot pain. Case description: A 65-year-old male presented to physical therapy with the diagnosis of plantar fasciitis by an orthopedic surgeon. The presentation included a diffuse area of pain toward the medial border of the foot with a peripheral neuropathic pain description. Cupping was used to identify pain in the saphenous nerve distribution and aided in resolving symptoms with the concomitant use of lower quarter neural glides. Outcome: At discharge and 1-year follow-up, the patient had a full resolution of symptoms and a return to prior level of function. Self-report outcomes included the numeric pain rating scale and the lower extremity functional scale. Discussion: This case is the first to describe the use of cupping combined with neural glides in the diagnosis and management of peripheral neuropathic pain from the saphenous nerve that was previously diagnosed as plantar fasciitis. The proposed mechanisms behind this treatment are also reviewed. Conclusion: In patients that present with symptoms of plantar fasciitis, testing neural glides combined with cupping may be warranted to confirm or refute the presence of a peripheral neuropathic pain source. Further studies are necessary to determine the mechanisms and further utility of the combined interventions in well controlled trials. Level of Evidence: Level IV.
Collapse
Affiliation(s)
| | - Ian Young
- CORA Physical Therapy, Savannah, GA, USA
| | | |
Collapse
|
46
|
Richner M, Ferreira N, Dudele A, Jensen TS, Vaegter CB, Gonçalves NP. Functional and Structural Changes of the Blood-Nerve-Barrier in Diabetic Neuropathy. Front Neurosci 2019; 12:1038. [PMID: 30692907 PMCID: PMC6339909 DOI: 10.3389/fnins.2018.01038] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 12/21/2018] [Indexed: 01/21/2023] Open
Abstract
The incidence of diabetes mellitus is approaching global epidemic proportions and should be considered a major health-care problem of modern societies in the twenty-first century. Diabetic neuropathy is a common chronic complication of diabetes and, although an adequate glycemic control can reduce the frequency of diabetic neuropathy in type 1 diabetes, the majority of type 2 diabetic patients will develop this complication. The underlying cellular and molecular mechanisms are still poorly understood, preventing the development of effective treatment strategies. However, accumulating evidence suggests that breakdown of the blood-nerve barrier (BNB) plays a pivotal pathophysiological role in diabetic neuropathy. In the present review, we highlight the structural and functional significance of the BNB in health and disease, focusing on the pathological molecular events leading to BNB dysfunction in diabetic neuropathy. In addition, we discuss potential molecular targets involved in BNB homeostasis that may pave the way toward novel therapeutic strategies for treating diabetic neuropathy.
Collapse
Affiliation(s)
- Mette Richner
- Danish Research Institute of Translational Neuroscience, Nordic EMBL Partnership for Molecular Medicine, Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Nelson Ferreira
- Danish Research Institute of Translational Neuroscience, Nordic EMBL Partnership for Molecular Medicine, Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Anete Dudele
- The International Diabetic Neuropathy Consortium, Aarhus University Hospital, Aarhus, Denmark.,Center of Functionally Integrative Neuroscience, Institute of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Troels S Jensen
- The International Diabetic Neuropathy Consortium, Aarhus University Hospital, Aarhus, Denmark.,Department of Neurology, Danish Pain Research Center, Aarhus University, Aarhus, Denmark
| | - Christian B Vaegter
- Danish Research Institute of Translational Neuroscience, Nordic EMBL Partnership for Molecular Medicine, Department of Biomedicine, Aarhus University, Aarhus, Denmark.,The International Diabetic Neuropathy Consortium, Aarhus University Hospital, Aarhus, Denmark
| | - Nádia P Gonçalves
- Danish Research Institute of Translational Neuroscience, Nordic EMBL Partnership for Molecular Medicine, Department of Biomedicine, Aarhus University, Aarhus, Denmark.,The International Diabetic Neuropathy Consortium, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
47
|
Reinhold AK, Yang S, Chen JTC, Hu L, Sauer RS, Krug SM, Mambretti EM, Fromm M, Brack A, Rittner HL. Tissue plasminogen activator and neuropathy open the blood-nerve barrier with upregulation of microRNA-155-5p in male rats. Biochim Biophys Acta Mol Basis Dis 2019; 1865:1160-1169. [PMID: 30625382 DOI: 10.1016/j.bbadis.2019.01.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 11/30/2018] [Accepted: 01/04/2019] [Indexed: 12/14/2022]
Abstract
The blood-nerve barrier (BNB) consisting of the perineurium and endoneurial vessels is sealed by tight junction proteins. BNB alterations are a crucial factor in the pathogenesis of peripheral neuropathies. However, barrier opening, e.g. by tissue plasminogen activator (tPA), can also facilitate topical application of analgesics. Here, we examined tPA both in the pathophysiology of neuropathy-induced BNB opening or via exogenous application and its effect on the cytoplasmatic tight junction protein anchoring protein, zona occludens-1 (ZO-1), the adherens molecule JAM-C and microRNA(miR)-155-5p. Specifically, we investigated whether tPA alone and barrier opening lead to pain behavioral changes, i.e. hyperalgesia, or whether these effects require further factors. Male Wistar rats underwent chronic constriction injury (CCI) or were treated by a single perisciatic application of recombinant (r)tPA. CCI elicited mechanical allodynia, tPA mRNA upregulation, macrophage invasion, BNB leakage for large molecule tracers, downregulation of ZO-1 and JAM-C mRNA/protein, and a loss of immunoreactivity of both in perineurium and endoneurial cells. Similarly, after perisciatic rtPA injection, ZO-1 and JAM-C mRNA as well as cytosolic/membrane protein and ZO-1 immunoreactivity were downregulated, and the BNB was opened. Neither mechanical hypersensitivity nor macrophage infiltration was observed after rtPA in contrast to CCI. Mechanistically, miR-155-5p, which is known to destabilize barriers and tight junction proteins like claudin-1 and ZO-1, was increased in CCI and to lesser extent after rtPA application. In summary, tPA transiently opens the BNB possibly via miR-155-5p. However, tPA does not provoke allodynia in the absence of a neuropathic stimulus like a ligation or inflammation.
Collapse
Affiliation(s)
- Ann-Kristin Reinhold
- Dept. of Anesthesiology, University Hospital of Würzburg, 97080 Würzburg, Germany
| | - Shaobing Yang
- Dept. of Anesthesiology, University Hospital of Würzburg, 97080 Würzburg, Germany; Dept. of Anesthesiology and Pain Medicine, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China
| | | | - Liu Hu
- Dept. of Anesthesiology, University Hospital of Würzburg, 97080 Würzburg, Germany; Dept. of Anesthesiology and Pain Medicine, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China
| | - Reine-Solange Sauer
- Dept. of Anesthesiology, University Hospital of Würzburg, 97080 Würzburg, Germany
| | - Susanne M Krug
- Institute of Clinical Physiology, Charité - Universitätsmedizin Berlin, Campus Benjamin Franklin, 12200 Berlin, Germany
| | - Egle M Mambretti
- Dept. of Anesthesiology, University Hospital of Würzburg, 97080 Würzburg, Germany
| | - Michael Fromm
- Institute of Clinical Physiology, Charité - Universitätsmedizin Berlin, Campus Benjamin Franklin, 12200 Berlin, Germany
| | - Alexander Brack
- Dept. of Anesthesiology, University Hospital of Würzburg, 97080 Würzburg, Germany
| | - Heike L Rittner
- Dept. of Anesthesiology, University Hospital of Würzburg, 97080 Würzburg, Germany.
| |
Collapse
|
48
|
Reinhold AK, Schwabe J, Lux TJ, Salvador E, Rittner HL. Quantitative and Microstructural Changes of the Blood-Nerve Barrier in Peripheral Neuropathy. Front Neurosci 2018; 12:936. [PMID: 30618565 PMCID: PMC6305433 DOI: 10.3389/fnins.2018.00936] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 11/27/2018] [Indexed: 12/14/2022] Open
Abstract
Peripheral neuropathy is accompanied by changes in the neuronal environment. The blood-nerve barrier (BNB) is crucial in protecting the neural homeostasis: Tight junctions (TJ) seal paracellular spaces and thus prevent external stimuli from entering. In different models of neuropathic pain, the BNB is impaired, thus contributing to local damage, immune cell invasion and, ultimately, the development of neuropathy with its symptoms. In this study, we examined changes in expression and microstructural localization of two key tight junction proteins (TJP), claudin-1 and the cytoplasmic anchoring ZO-1, in the sciatic nerve of mice subjected to chronic constriction injury (CCI). Via qPCR and analysis of fluorescence immunohistochemistry, a marked downregulation of mRNA as well as decreased fluorescence intensity were observed in the nerve for both proteins. Moreover, a distinct zig-zag structure for both proteins located at cell-cell contacts, indicative of the localization of TJs, was observed in the perineurial compartment of sham-operated animals. This microstructural location in cell-cell-contacts was lost in neuropathy as semiquantified via computational analysis, based on a novel algorithm. In summary, we provide evidence that peripheral neuropathy is not only associated with decrease in relevant TJPs but also exhibits alterations in TJP arrangement and loss in barrier tightness, presumably due to internalization. Specifically, semiquantification of TJP in cell-cell-contacts of microcompartments could be used in the future for routine clinical samples of patients with neuropathy.
Collapse
Affiliation(s)
- Ann Kristin Reinhold
- Department of Anaesthesiology, University Hospitals Würzburg, Wüerzburg, Germany
| | - Joachim Schwabe
- Department of Anaesthesiology, University Hospitals Würzburg, Wüerzburg, Germany
| | - Thomas J Lux
- Department of Anaesthesiology, University Hospitals Würzburg, Wüerzburg, Germany
| | - Ellaine Salvador
- Department of Anaesthesiology, University Hospitals Würzburg, Wüerzburg, Germany
| | - Heike L Rittner
- Department of Anaesthesiology, University Hospitals Würzburg, Wüerzburg, Germany
| |
Collapse
|
49
|
Lee S, Shi XQ, Fan A, West B, Zhang J. Targeting macrophage and microglia activation with colony stimulating factor 1 receptor inhibitor is an effective strategy to treat injury-triggered neuropathic pain. Mol Pain 2018; 14:1744806918764979. [PMID: 29546785 PMCID: PMC5858622 DOI: 10.1177/1744806918764979] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Introduction Neuropathic pain is a debilitating condition. The importance of neuroimmune interactions in neuropathic pain has been evidenced by the involvement of different immune cells in peripheral and central sensitization of pathological pain. Macrophages and microglia are the most abundant immune cells activated in injured nerves and spinal cord, respectively. Several lines of evidence showed that macrophage/microglia survival, activation, proliferation, and differentiation require the involvement of macrophage-colony stimulating factor. In this study, we investigated whether blocking macrophage-colony stimulating factor/colony stimulating factor 1 receptor signaling can be effective in relieving neuropathic pain. Materials and methods Partial sciatic nerve ligation was performed in mice to induce neuropathic pain behavior. Mice were orally treated with a selective colony stimulating factor 1 receptor inhibitor, PLX5622, daily in both preventive (two days prior to surgery until D14 post-partial sciatic nerve ligation) and reversal paradigms (D28-D33 post-partial sciatic nerve ligation). Animal neuropathic pain behavior was monitored using von Frey hairs and acetone application. Phenotype of macrophages in injured nerves was analyzed at D3 and D33 post-injury using flow cytometry analysis. The effect of PLX5622 on microglia activation in lumbar spinal cord was further examined by immunohistochemistry using Iba-1 antibody. Results Significant alleviation of both mechanical and cold allodynia was observed in PLX5622-treated animals, both in preventive and reversal paradigms. PLX5622 treatment reduced the total number of macrophages in injured nerves, it appears colony stimulating factor 1 receptor inhibition affected more specifically CD86+ (M1 like) macrophages. Consequently, the expression of various pro-inflammatory cytokines (TNF-α, IL-1β) was reduced. Microglia activation in dorsal horn of lumbar spinal cord following partial sciatic nerve ligation was significantly inhibited with PLX5622 treatment in both preventive and reversal paradigms. Conclusion Macrophages in peripheral nerve and microglia in the spinal cord are required in the generation and maintenance of injury-associated neuropathic pain. Blocking macrophage-colony stimulating factor/colony stimulating factor 1 receptor signaling on these myeloid cells along the pain transmission pathway is an effective strategy to alleviate neuropathic pain.
Collapse
Affiliation(s)
- SeungHwan Lee
- 1 Faculty of Dentistry, 5620 McGill University , Montreal, QC, Canada.,2 The Alan Edwards Centre for Research on Pain, 5620 McGill University , Montreal, QC, Canada
| | - Xiang Qun Shi
- 1 Faculty of Dentistry, 5620 McGill University , Montreal, QC, Canada.,2 The Alan Edwards Centre for Research on Pain, 5620 McGill University , Montreal, QC, Canada
| | - Anni Fan
- 1 Faculty of Dentistry, 5620 McGill University , Montreal, QC, Canada.,2 The Alan Edwards Centre for Research on Pain, 5620 McGill University , Montreal, QC, Canada
| | | | - Ji Zhang
- 1 Faculty of Dentistry, 5620 McGill University , Montreal, QC, Canada.,2 The Alan Edwards Centre for Research on Pain, 5620 McGill University , Montreal, QC, Canada.,4 Department of Neurology and Neurosurgery, 5620 McGill University , Montreal, QC, Canada
| |
Collapse
|
50
|
Moreau N, Mauborgne A, Couraud PO, Romero IA, Weksler BB, Villanueva L, Pohl M, Boucher Y. Could an endoneurial endothelial crosstalk between Wnt/β-catenin and Sonic Hedgehog pathways underlie the early disruption of the infra-orbital blood-nerve barrier following chronic constriction injury? Mol Pain 2018; 13:1744806917727625. [PMID: 28814148 PMCID: PMC5574482 DOI: 10.1177/1744806917727625] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background Blood–nerve barrier disruption is pivotal in the development of neuroinflammation, peripheral sensitization, and neuropathic pain after peripheral nerve injury. Activation of toll-like receptor 4 and inactivation of Sonic Hedgehog signaling pathways within the endoneurial endothelial cells are key events, resulting in the infiltration of harmful molecules and immunocytes within the nerve parenchyma. However, we showed in a previous study that preemptive inactivation of toll-like receptor 4 signaling or sustained activation of Sonic Hedgehog signaling did not prevent the local alterations observed following peripheral nerve injury, suggesting the implication of another signaling pathway. Methods Using a classical neuropathic pain model, the infraorbital nerve chronic constriction injury (IoN-CCI), we investigated the role of the Wnt/β-catenin pathway in chronic constriction injury-mediated blood–nerve barrier disruption and in its interactions with the toll-like receptor 4 and Sonic Hedgehog pathways. In the IoN-CCI model versus control, mRNA expression levels and/or immunochemical detection of major Wnt/Sonic Hedgehog pathway (Frizzled-7, vascular endothelial-cadherin, Patched-1 and Gli-1) and/or tight junction proteins (Claudin-1, Claudin-5, and Occludin) readouts were assessed. Vascular permeability was assessed by sodium fluorescein extravasation. Results IoN-CCI induced early alterations in the vascular endothelial-cadherin/β-catenin/Frizzled-7 complex, shown to participate in local blood–nerve barrier disruption via a β-catenin-dependent tight junction protein downregulation. Wnt pathway also mediated a crosstalk between toll-like receptor 4 and Sonic Hedgehog signaling within endoneurial endothelial cells. Nevertheless, preemptive inhibition of Wnt/β-catenin signaling before IoN-CCI could not prevent the downregulation of key Sonic Hedgehog pathway readouts or the disruption of the infraorbital blood–nerve barrier, suggesting that Sonic Hedgehog pathway inhibition observed following IoN-CCI is an independent event responsible for blood–nerve barrier disruption. Conclusion A crosstalk between Wnt/β-catenin- and Sonic Hedgehog-mediated signaling pathways within endoneurial endothelial cells could mediate the chronic disruption of the blood–nerve barrier following IoN-CCI, resulting in increased irreversible endoneurial vascular permeability and neuropathic pain development.
Collapse
Affiliation(s)
- Nathan Moreau
- Centre de Psychiatrie et Neurosciences, INSERM UMR 894, Paris, France
| | - Annie Mauborgne
- Centre de Psychiatrie et Neurosciences, INSERM UMR 894, Paris, France
| | | | - Ignacio A Romero
- Department of Biological Sciences, The Open University, Walton Hall, Milton Keynes MK7 6BJ, UK
| | - Babette B Weksler
- Weill Medical College of Cornell University, New York, NY 10065, USA
| | - Luis Villanueva
- 1Centre de Psychiatrie et Neurosciences, INSERM UMR 894, Paris, France
| | - Michel Pohl
- 1Centre de Psychiatrie et Neurosciences, INSERM UMR 894, Paris, France
| | | |
Collapse
|