1
|
Lantermans HC, Ma F, Kuil A, de Rooij MFM, Bergkamp ND, van der Meer W, van Buul JD, Smit MJ, Kersten MJ, Spaargaren M, Pals ST. Presentation of CXCL12γ by heparan sulfate proteoglycans activates CXCR4 without desensitization in normal and malignant B cells. Blood Adv 2025; 9:2307-2320. [PMID: 39969206 DOI: 10.1182/bloodadvances.2024014396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 01/21/2025] [Accepted: 02/03/2025] [Indexed: 02/20/2025] Open
Abstract
ABSTRACT CXCL12-CXCR4 signaling is involved in a wide variety of homeostatic and pathologic processes, but the role of specific CXCL12 isoforms has remained largely unexplored. We have recently shown that the CXCL12γ isoform, which holds an exceptionally high affinity for heparan sulfate (HS), is produced by human bone marrow stromal cells (BMSCs) and remains cell surface immobilized by HS proteoglycans (HSPGs). This HS-bound CXCL12γ is critical for the adhesion of multiple myeloma cells to BMSCs and for BMSC-mediated drug resistance. In this study, we investigated how CXCL12γ activates and regulates CXCR4 by employing a variety of biosensors in HEK293T cells, endogenous CXCR4-expressing B-lymphoma and myeloma cell lines and primary B cells. We showed that CXCL12γ and CXCL12α bind CXCR4 with a similar affinity and that the cumulative activation of CXCR4 over time is equal for both ligands, although CXCL12α activates CXCR4 more rapidly. Although nonbound CXCL12γ and CXCL12α equally induce CXCR4 internalization, cell- or heparin-bound CXCL12γ hardly induces CXCR4 internalization or desensitization. CXCL12γ presented by HSPGs on the membrane of human bone marrow endothelial cells (HBMECs) induces potent cell adhesion to the endothelium under physiological flow, but cells retain the ability to migrate toward CXCL12α when they encounter HBMEC-bound CXCL12γ. Taken together, our data demonstrate that CXCL12γ and CXCL12α differentially modulate CXCR4 trafficking and that CXCL12γ, when immobilized and presented by HSPGs on the cell surface of HBMECs, can efficiently arrest circulating cells without causing CXCR4 internalization or desensitization, thus enabling subsequent cell migration toward a CXCL12α gradient.
Collapse
Affiliation(s)
- Hildo C Lantermans
- Department of Pathology, Amsterdam University Medical Center Location University of Amsterdam, Amsterdam, The Netherlands
- Lymphoma and Myeloma Center Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Target & Therapy Discovery, Amsterdam, The Netherlands
| | - Fangxue Ma
- Department of Pathology, Amsterdam University Medical Center Location University of Amsterdam, Amsterdam, The Netherlands
- Lymphoma and Myeloma Center Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Target & Therapy Discovery, Amsterdam, The Netherlands
| | - Annemieke Kuil
- Department of Pathology, Amsterdam University Medical Center Location University of Amsterdam, Amsterdam, The Netherlands
- Lymphoma and Myeloma Center Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Target & Therapy Discovery, Amsterdam, The Netherlands
| | - Martin F M de Rooij
- Department of Pathology, Amsterdam University Medical Center Location University of Amsterdam, Amsterdam, The Netherlands
- Lymphoma and Myeloma Center Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Target & Therapy Discovery, Amsterdam, The Netherlands
| | - Nick D Bergkamp
- Amsterdam Institute for Molecular and Life Sciences, Division of Medicinal Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Werner van der Meer
- Department of Medical Biochemistry, Vascular Cell Biology Laboratory, Amsterdam University Medical Center Location University of Amsterdam, Amsterdam, The Netherlands
- Section Molecular Cytology at Swammerdam Institute for Life Sciences, Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, Amsterdam, The Netherlands
| | - Jaap D van Buul
- Department of Medical Biochemistry, Vascular Cell Biology Laboratory, Amsterdam University Medical Center Location University of Amsterdam, Amsterdam, The Netherlands
- Section Molecular Cytology at Swammerdam Institute for Life Sciences, Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, Amsterdam, The Netherlands
| | - Martine J Smit
- Amsterdam Institute for Molecular and Life Sciences, Division of Medicinal Chemistry, Department of Chemistry and Pharmaceutical Sciences, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Marie José Kersten
- Lymphoma and Myeloma Center Amsterdam, Amsterdam, The Netherlands
- Department of Hematology, Amsterdam University Medical Center Location University of Amsterdam, Amsterdam, The Netherlands
| | - Marcel Spaargaren
- Department of Pathology, Amsterdam University Medical Center Location University of Amsterdam, Amsterdam, The Netherlands
- Lymphoma and Myeloma Center Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Target & Therapy Discovery, Amsterdam, The Netherlands
| | - Steven T Pals
- Department of Pathology, Amsterdam University Medical Center Location University of Amsterdam, Amsterdam, The Netherlands
- Lymphoma and Myeloma Center Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Target & Therapy Discovery, Amsterdam, The Netherlands
| |
Collapse
|
2
|
Belančić A, Janković T, Gkrinia EMM, Kristić I, Rajič Bumber J, Rački V, Pilipović K, Vitezić D, Mršić-Pelčić J. Glial Cells in Spinal Muscular Atrophy: Speculations on Non-Cell-Autonomous Mechanisms and Therapeutic Implications. Neurol Int 2025; 17:41. [PMID: 40137462 PMCID: PMC11944370 DOI: 10.3390/neurolint17030041] [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: 02/09/2025] [Revised: 03/07/2025] [Accepted: 03/11/2025] [Indexed: 03/29/2025] Open
Abstract
Spinal muscular atrophy (SMA) is a neuromuscular disorder caused by homozygous deletions or mutations in the SMN1 gene, leading to progressive motor neuron degeneration. While SMA has been classically viewed as a motor neuron-autonomous disease, increasing evidence indicates a significant role of glial cells-astrocytes, microglia, oligodendrocytes, and Schwann cells-in the disease pathophysiology. Astrocytic dysfunction contributes to motor neuron vulnerability through impaired calcium homeostasis, disrupted synaptic integrity, and neurotrophic factor deficits. Microglia, through reactive gliosis and complement-mediated synaptic stripping, exacerbate neurodegeneration and neuroinflammation. Oligodendrocytes exhibit impaired differentiation and metabolic support, while Schwann cells display abnormalities in myelination, extracellular matrix composition, and neuromuscular junction maintenance, further compromising motor function. Dysregulation of pathways such as NF-κB, Notch, and JAK/STAT, alongside the upregulation of complement proteins and microRNAs, reinforces the non-cell-autonomous nature of SMA. Despite the advances in SMN-restorative therapies, they do not fully mitigate glial dysfunction. Targeting glial pathology, including modulation of reactive astrogliosis, microglial polarization, and myelination deficits, represents a critical avenue for therapeutic intervention. This review comprehensively examines the multifaceted roles of glial cells in SMA and highlights emerging glia-targeted strategies to enhance treatment efficacy and improve patient outcomes.
Collapse
Affiliation(s)
- Andrej Belančić
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (T.J.); (I.K.); (J.R.B.); (K.P.); (D.V.); (J.M.-P.)
| | - Tamara Janković
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (T.J.); (I.K.); (J.R.B.); (K.P.); (D.V.); (J.M.-P.)
| | | | - Iva Kristić
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (T.J.); (I.K.); (J.R.B.); (K.P.); (D.V.); (J.M.-P.)
| | - Jelena Rajič Bumber
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (T.J.); (I.K.); (J.R.B.); (K.P.); (D.V.); (J.M.-P.)
| | - Valentino Rački
- Department of Neurology, Clinical Hospital Centre Rijeka, Krešimirova 42, 51000 Rijeka, Croatia;
| | - Kristina Pilipović
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (T.J.); (I.K.); (J.R.B.); (K.P.); (D.V.); (J.M.-P.)
| | - Dinko Vitezić
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (T.J.); (I.K.); (J.R.B.); (K.P.); (D.V.); (J.M.-P.)
| | - Jasenka Mršić-Pelčić
- Department of Basic and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia; (T.J.); (I.K.); (J.R.B.); (K.P.); (D.V.); (J.M.-P.)
| |
Collapse
|
3
|
Soendenbroe C, Schjerling P, Bechshøft CJL, Svensson RB, Schaeffer L, Kjaer M, Chazaud B, Jacquier A, Mackey AL. Muscle fibroblasts and stem cells stimulate motor neurons in an age and exercise-dependent manner. Aging Cell 2025; 24:e14413. [PMID: 39555723 PMCID: PMC11896526 DOI: 10.1111/acel.14413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 10/24/2024] [Accepted: 11/02/2024] [Indexed: 11/19/2024] Open
Abstract
Exercise preserves neuromuscular function in aging through unknown mechanisms. Skeletal muscle fibroblasts (FIB) and stem cells (MuSC) are abundant in skeletal muscle and reside close to neuromuscular junctions, but their relative roles in motor neuron maintenance remain undescribed. Using direct cocultures of embryonic rat motor neurons with either human MuSC or FIB, RNA sequencing revealed profound differential regulation of the motor neuron transcriptome, with FIB generally favoring neuron growth and cell migration and MuSC favoring production of ribosomes and translational machinery. Conditioned medium from FIB was superior to MuSC in preserving motor neurons and increasing their maturity. Lastly, we established the importance of donor age and exercise status and found an age-related distortion of motor neuron and muscle cell interaction that was fully mitigated by lifelong physical activity. In conclusion, we show that human muscle FIB and MuSC synergistically stimulate the growth and viability of motor neurons, which is further amplified by regular exercise.
Collapse
Affiliation(s)
- Casper Soendenbroe
- Department of Orthopedic SurgeryInstitute of Sports Medicine Copenhagen, Copenhagen University Hospital—Bispebjerg and FrederiksbergCopenhagenDenmark
- Department of Clinical Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
- Department of Clinical Biochemistry, Faculty of Health Sciences, Bispebjerg HospitalUniversity of CopenhagenCopenhagenDenmark
| | - Peter Schjerling
- Department of Orthopedic SurgeryInstitute of Sports Medicine Copenhagen, Copenhagen University Hospital—Bispebjerg and FrederiksbergCopenhagenDenmark
- Department of Clinical Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Cecilie J. L. Bechshøft
- Department of Orthopedic SurgeryInstitute of Sports Medicine Copenhagen, Copenhagen University Hospital—Bispebjerg and FrederiksbergCopenhagenDenmark
- Department of Clinical Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Rene B. Svensson
- Department of Orthopedic SurgeryInstitute of Sports Medicine Copenhagen, Copenhagen University Hospital—Bispebjerg and FrederiksbergCopenhagenDenmark
- Department of Clinical Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Laurent Schaeffer
- Institut NeuroMyoGèneUnité Physiopathologie et Génétique du Neurone et du MuscleLyonFrance
- Centre de Biotechnologie Cellulaire, CBC BiotecCHU de Lyon—Hospices Civils de Lyon (HCL) Groupement EstBronFrance
| | - Michael Kjaer
- Department of Orthopedic SurgeryInstitute of Sports Medicine Copenhagen, Copenhagen University Hospital—Bispebjerg and FrederiksbergCopenhagenDenmark
- Department of Clinical Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Bénédicte Chazaud
- Institut NeuroMyoGèneUnité Physiopathologie et Génétique du Neurone et du MuscleLyonFrance
| | - Arnaud Jacquier
- Institut NeuroMyoGèneUnité Physiopathologie et Génétique du Neurone et du MuscleLyonFrance
- Centre de Biotechnologie Cellulaire, CBC BiotecCHU de Lyon—Hospices Civils de Lyon (HCL) Groupement EstBronFrance
| | - Abigail L. Mackey
- Department of Orthopedic SurgeryInstitute of Sports Medicine Copenhagen, Copenhagen University Hospital—Bispebjerg and FrederiksbergCopenhagenDenmark
- Department of Clinical Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| |
Collapse
|
4
|
Gould TW, Ko CP, Willison H, Robitaille R. Perisynaptic Schwann Cells: Guardians of Neuromuscular Junction Integrity and Function in Health and Disease. Cold Spring Harb Perspect Biol 2025; 17:a041362. [PMID: 38858074 PMCID: PMC11694759 DOI: 10.1101/cshperspect.a041362] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2024]
Abstract
The neuromuscular junction (NMJ) is a highly reliable synapse to carry the control of the motor commands of the nervous system over the muscles. Its development, organization, and synaptic properties are highly structured and regulated to support such reliability and efficacy. Yet, the NMJ is also highly plastic, able to react to injury, and able to adapt to changes. This balance between structural stability and synaptic efficacy on one hand and structural plasticity and repair on another hand is made possible by perisynaptic Schwann cells (PSCs), glial cells at this synapse. They regulate synaptic efficacy and structural plasticity of the NMJ in a dynamic, bidirectional manner owing to their ability to decode synaptic transmission and by their interactions with trophic-related factors. Alteration of these fundamental roles of PSCs is also important in the maladapted response of NMJs in various diseases and in aging.
Collapse
Affiliation(s)
- Thomas W Gould
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, Nevada 89557, USA
| | - Chien-Ping Ko
- Section of Neurobiology, Department of Biological Sciences, University of Southern California, Los Angeles, California 90089-2520, USA
| | - Hugh Willison
- School of Infection and Immunity, University of Glasgow, Glasgow G12 8TA, Scotland
| | - Richard Robitaille
- Département de neurosciences, Université de Montréal, Montréal, Québec H3C 3J7, Canada
- Centre Interdisciplinaire de Recherche sur le Cerveau et l'apprentissage, Université de Montréal, Montréal, Québec H3C 3J7, Canada
| |
Collapse
|
5
|
Moss KR, Saxena S. Schwann Cells in Neuromuscular Disorders: A Spotlight on Amyotrophic Lateral Sclerosis. Cells 2025; 14:47. [PMID: 39791748 PMCID: PMC11719703 DOI: 10.3390/cells14010047] [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: 11/14/2024] [Revised: 12/23/2024] [Accepted: 01/01/2025] [Indexed: 01/12/2025] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a complex neurodegenerative disease primarily affecting motor neurons, leading to progressive muscle atrophy and paralysis. This review explores the role of Schwann cells in ALS pathogenesis, highlighting their influence on disease progression through mechanisms involving demyelination, neuroinflammation, and impaired synaptic function. While Schwann cells have been traditionally viewed as peripheral supportive cells, especially in motor neuron disease, recent evidence indicates that they play a significant role in ALS by impacting motor neuron survival and plasticity, influencing inflammatory responses, and altering myelination processes. Furthermore, advancements in understanding Schwann cell pathology in ALS combined with lessons learned from studying Charcot-Marie-Tooth disease Type 1 (CMT1) suggest potential therapeutic strategies targeting these cells may support nerve repair and slow disease progression. Overall, this review aims to provide comprehensive insights into Schwann cell classification, physiology, and function, underscoring the critical pathological contributions of Schwann cells in ALS and suggests new avenues for targeted therapeutic interventions aimed at modulating Schwann cell function in ALS.
Collapse
Affiliation(s)
- Kathryn R. Moss
- Department of Physical Medicine and Rehabilitation, University of Missouri School of Medicine, Columbia, MO 65211, USA
- NextGen Precision Health, University of Missouri, Columbia, MO 65211, USA
| | - Smita Saxena
- Department of Physical Medicine and Rehabilitation, University of Missouri School of Medicine, Columbia, MO 65211, USA
- NextGen Precision Health, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
6
|
Tosolini AP, Abatecola F, Negro S, Sleigh JN, Schiavo G. The node of Ranvier influences the in vivo axonal transport of mitochondria and signaling endosomes. iScience 2024; 27:111158. [PMID: 39524336 PMCID: PMC11544082 DOI: 10.1016/j.isci.2024.111158] [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: 06/24/2024] [Revised: 09/15/2024] [Accepted: 10/09/2024] [Indexed: 11/16/2024] Open
Abstract
Efficient long-range axonal transport is essential for maintaining neuronal function, and perturbations in this process underlie severe neurological diseases. Nodes of Ranvier (NoR) are short, specialized unmyelinated axonal domains with a unique molecular and structural composition. Currently, it remains unresolved how the distinct molecular structures of the NoR impact axonal transport dynamics. Using intravital time-lapse microscopy of sciatic nerves in live, anesthetized mice, we reveal (1) similar morphologies of the NoR in fast and slow motor axons, (2) signaling endosomes and mitochondria accumulate specifically at the distal node, and (3) unique axonal transport profiles of signaling endosomes and mitochondria transiting through the NoR. Collectively, these findings provide important insights into the fundamental physiology of peripheral nerve axons, motor neuron subtypes, and diverse organelle dynamics at the NoR. Furthermore, this work has relevance for several pathologies affecting peripheral nerves and the NoR.
Collapse
Affiliation(s)
- Andrew P. Tosolini
- Department of Neuromuscular Diseases and UCL Queen Square Motor Neuron Disease Centre, Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, QLD 4067, Australia
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4067, Australia
| | - Federico Abatecola
- Department of Neuromuscular Diseases and UCL Queen Square Motor Neuron Disease Centre, Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Samuele Negro
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy
- U.O.C. Clinica Neurologica, Azienda Ospedale, University of Padua, 35128 Padua, Italy
| | - James N. Sleigh
- Department of Neuromuscular Diseases and UCL Queen Square Motor Neuron Disease Centre, Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
- UK Dementia Research Institute, University College London, London WC1E 6BT, UK
| | - Giampietro Schiavo
- Department of Neuromuscular Diseases and UCL Queen Square Motor Neuron Disease Centre, Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
- UK Dementia Research Institute, University College London, London WC1E 6BT, UK
| |
Collapse
|
7
|
Hosoki H, Asahi T, Nozaki C. Cannabinoid CB2 receptors enhance high-fat diet evoked peripheral neuroinflammation. Life Sci 2024; 355:123002. [PMID: 39173999 DOI: 10.1016/j.lfs.2024.123002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 08/11/2024] [Accepted: 08/19/2024] [Indexed: 08/24/2024]
Abstract
It is known that the cannabinoid type 2 (CB2) receptor has an anti-inflammatory role. Therefore, animals without CB2 receptors show enhanced inflammation and pain in the model of chronic pain, e.g., neuropathic pain. We previously proposed the upregulated leptin signaling at the peripheral nerve as one of the underlying molecular mechanisms of pain exacerbation in nerve-injured CB2 knockouts, as they displayed robust upregulation of leptin receptors and leptin signaling in the peripheral nerve. Due to these past results, we hypothesized that CB2 receptor deficiency might also modify the peripheral neuroinflammation led by chronic exposure to a high-fat diet (HFD). Interestingly, CB2 knockout animals showed significant resistance to HFD-induced neuroinflammation. Namely, 5-week feeding of HFD induced substantial hypersensitivity in WT animals, while tactile sensitivity of HFD-fed CB2 knockouts remained intact. HFD-fed WT animals also displayed the robust upregulation of chemokine CXCR4 expression with increased macrophage infiltration, which was never observed in HFD-fed CB2 knockout mice. Moreover, 5-week HFD exposure led significant increase of CD11b+Ly6G-Ly6Chigh cells and a decrease of CD11b+Ly6G+Ly6Clow cells in the spleen of WT animals, which was also not found in either HFD-fed CB2 knockouts or standard diet-fed WT and CB2 animals. Together with past reports, these results suggest that CB2 receptors might have a double-sided regulatory role in the context of inflammation development or, more widely, immune system regulation. We propose that CB2 signaling is not always anti-inflammatory and could take a pro-inflammatory role depending on the cause of the inflammation.
Collapse
Affiliation(s)
- Haruka Hosoki
- School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Toru Asahi
- School of Advanced Science and Engineering, Waseda University, Tokyo, Japan; Comprehensive Research Organization, Waseda University, Tokyo, Japan; Research Organization for Nano & Life Innovation, Waseda University, Tokyo, Japan
| | - Chihiro Nozaki
- School of Advanced Science and Engineering, Waseda University, Tokyo, Japan; Global Center for Science and Engineering, Waseda University, Tokyo, Japan.
| |
Collapse
|
8
|
Montecucco C, Bano L. A possible cause and therapy for equine grass sickness. Toxicon 2024; 245:107790. [PMID: 38821320 DOI: 10.1016/j.toxicon.2024.107790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 06/02/2024]
Affiliation(s)
- Cesare Montecucco
- Institute of Neuroscience, National Research Council and Department of Biomedical Sciences, University of Padova, 35131, Padua, Italy.
| | - Luca Bano
- Microbiology and Veterinary Diagnostic Laboratory, Istituto Zooprofilattico Sperimentale delle Venezie, 35020, Legnaro, Italy.
| |
Collapse
|
9
|
Stassart RM, Gomez-Sanchez JA, Lloyd AC. Schwann Cells as Orchestrators of Nerve Repair: Implications for Tissue Regeneration and Pathologies. Cold Spring Harb Perspect Biol 2024; 16:a041363. [PMID: 38199866 PMCID: PMC11146315 DOI: 10.1101/cshperspect.a041363] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2024]
Abstract
Peripheral nerves exist in a stable state in adulthood providing a rapid bidirectional signaling system to control tissue structure and function. However, following injury, peripheral nerves can regenerate much more effectively than those of the central nervous system (CNS). This multicellular process is coordinated by peripheral glia, in particular Schwann cells, which have multiple roles in stimulating and nurturing the regrowth of damaged axons back to their targets. Aside from the repair of damaged nerves themselves, nerve regenerative processes have been linked to the repair of other tissues and de novo innervation appears important in establishing an environment conducive for the development and spread of tumors. In contrast, defects in these processes are linked to neuropathies, aging, and pain. In this review, we focus on the role of peripheral glia, especially Schwann cells, in multiple aspects of nerve regeneration and discuss how these findings may be relevant for pathologies associated with these processes.
Collapse
Affiliation(s)
- Ruth M Stassart
- Paul-Flechsig-Institute of Neuropathology, University Clinic Leipzig, Leipzig 04103, Germany
| | - Jose A Gomez-Sanchez
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante 03010, Spain
- Instituto de Neurociencias CSIC-UMH, Sant Joan de Alicante 03550, Spain
| | - Alison C Lloyd
- UCL Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, United Kingdom
| |
Collapse
|
10
|
Li A, Yi J, Li X, Dong L, Ostrow LW, Ma J, Zhou J. Distinct transcriptomic profile of satellite cells contributes to preservation of neuromuscular junctions in extraocular muscles of ALS mice. eLife 2024; 12:RP92644. [PMID: 38661532 PMCID: PMC11045223 DOI: 10.7554/elife.92644] [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] [Indexed: 04/26/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neuromuscular disorder characterized by progressive weakness of almost all skeletal muscles, whereas extraocular muscles (EOMs) are comparatively spared. While hindlimb and diaphragm muscles of end-stage SOD1G93A (G93A) mice (a familial ALS mouse model) exhibit severe denervation and depletion of Pax7+satellite cells (SCs), we found that the pool of SCs and the integrity of neuromuscular junctions (NMJs) are maintained in EOMs. In cell sorting profiles, SCs derived from hindlimb and diaphragm muscles of G93A mice exhibit denervation-related activation, whereas SCs from EOMs of G93A mice display spontaneous (non-denervation-related) activation, similar to SCs from wild-type mice. Specifically, cultured EOM SCs contain more abundant transcripts of axon guidance molecules, including Cxcl12, along with more sustainable renewability than the diaphragm and hindlimb counterparts under differentiation pressure. In neuromuscular co-culture assays, AAV-delivery of Cxcl12 to G93A-hindlimb SC-derived myotubes enhances motor neuron axon extension and innervation, recapitulating the innervation capacity of EOM SC-derived myotubes. G93A mice fed with sodium butyrate (NaBu) supplementation exhibited less NMJ loss in hindlimb and diaphragm muscles. Additionally, SCs derived from G93A hindlimb and diaphragm muscles displayed elevated expression of Cxcl12 and improved renewability following NaBu treatment in vitro. Thus, the NaBu-induced transcriptomic changes resembling the patterns of EOM SCs may contribute to the beneficial effects observed in G93A mice. More broadly, the distinct transcriptomic profile of EOM SCs may offer novel therapeutic targets to slow progressive neuromuscular functional decay in ALS and provide possible 'response biomarkers' in pre-clinical and clinical studies.
Collapse
Affiliation(s)
- Ang Li
- Department of Kinesiology, College of Nursing and Health Innovation, The University of Texas at ArlingtonArlingtonUnited States
| | - Jianxun Yi
- Department of Kinesiology, College of Nursing and Health Innovation, The University of Texas at ArlingtonArlingtonUnited States
| | - Xuejun Li
- Department of Kinesiology, College of Nursing and Health Innovation, The University of Texas at ArlingtonArlingtonUnited States
| | - Li Dong
- Department of Kinesiology, College of Nursing and Health Innovation, The University of Texas at ArlingtonArlingtonUnited States
| | - Lyle W Ostrow
- Department of Neurology, Lewis Katz School of Medicine at Temple UniversityPhiladelphiaUnited States
| | - Jianjie Ma
- Department of Surgery, Division of Surgical Sciences, University of VirginiaCharlottesvilleUnited States
| | - Jingsong Zhou
- Department of Kinesiology, College of Nursing and Health Innovation, The University of Texas at ArlingtonArlingtonUnited States
| |
Collapse
|
11
|
Li A, Yi J, Li X, Dong L, Ostrow LW, Ma J, Zhou J. Distinct transcriptomic profile of satellite cells contributes to preservation of neuromuscular junctions in extraocular muscles of ALS mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.02.12.528218. [PMID: 36824725 PMCID: PMC9949002 DOI: 10.1101/2023.02.12.528218] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neuromuscular disorder characterized by progressive weakness of almost all skeletal muscles, whereas extraocular muscles (EOMs) are comparatively spared. While hindlimb and diaphragm muscles of end-stage SOD1G93A (G93A) mice (a familial ALS mouse model) exhibit severe denervation and depletion of Pax7 + satellite cells (SCs), we found that the pool of SCs and the integrity of neuromuscular junctions (NMJs) are maintained in EOMs. In cell sorting profiles, SCs derived from hindlimb and diaphragm muscles of G93A mice exhibit denervation-related activation, whereas SCs from EOMs of G93A mice display spontaneous (non-denervation-related) activation, similar to SCs from wild-type mice. Specifically, cultured EOM SCs contain more abundant transcripts of axon guidance molecules, including Cxcl12 , along with more sustainable renewability than the diaphragm and hindlimb counterparts under differentiation pressure. In neuromuscular co-culture assays, AAV-delivery of Cxcl12 to G93A-hindlimb SC-derived myotubes enhances motor neuron axon extension and innervation, recapitulating the innervation capacity of EOM SC-derived myotubes. G93A mice fed with sodium butyrate (NaBu) supplementation exhibited less NMJ loss in hindlimb and diaphragm muscles. Additionally, SCs derived from G93A hindlimb and diaphragm muscles displayed elevated expression of Cxcl12 and improved renewability following NaBu treatment in vitro . Thus, the NaBu-induced transcriptomic changes resembling the patterns of EOM SCs may contribute to the beneficial effects observed in G93A mice. More broadly, the distinct transcriptomic profile of EOM SCs may offer novel therapeutic targets to slow progressive neuromuscular functional decay in ALS and provide possible "response biomarkers" in pre-clinical and clinical studies.
Collapse
|
12
|
Stazi M, Megighian A, D'Este G, Negro S, Ivanušec A, Lonati D, Pirazzini M, Križaj I, Montecucco C. An agonist of CXCR4 induces a rapid recovery from the neurotoxic effects of Vipera ammodytes and Vipera aspis venoms. J Neurochem 2024; 168:428-440. [PMID: 36912731 DOI: 10.1111/jnc.15803] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/30/2023] [Accepted: 02/06/2023] [Indexed: 03/14/2023]
Abstract
People bitten by Alpine vipers are usually treated with antivenom antisera to prevent the noxious consequences caused by the injected venom. However, this treatment suffers from a number of drawbacks and additional therapies are necessary. The venoms of Vipera ammodytes and of Vipera aspis are neurotoxic and cause muscle paralysis by inducing neurodegeneration of motor axon terminals because they contain a presynaptic acting sPLA2 neurotoxin. We have recently found that any type of damage to motor axons is followed by the expression and activation of the intercellular signaling axis consisting of the CXCR4 receptor present on the membrane of the axon stump and of its ligand, the chemokine CXCL12 released by activated terminal Schwann cells. We show here that also V. ammodytes and V. aspis venoms cause the expression of the CXCL12-CXCR4 axis. We also show that a small molecule agonist of CXCR4, dubbed NUCC-390, induces a rapid regeneration of the motor axon terminal with functional recovery of the neuromuscular junction. These findings qualify NUCC-390 as a promising novel therapeutics capable of improving the recovery from the paralysis caused by the snakebite of the two neurotoxic Alpine vipers.
Collapse
Affiliation(s)
- M Stazi
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - A Megighian
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - G D'Este
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - S Negro
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - A Ivanušec
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Ljubljana, Slovenia
- Doctoral School, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - D Lonati
- Pavia Poison Control Centre - National Toxicology Information Centre - Clinical and Experimental Lab, Toxicology Unit, Istituti Clinici Scientifici Maugeri SpA SB IRCCS, Pavia, Italy
| | - M Pirazzini
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - I Križaj
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Ljubljana, Slovenia
| | - C Montecucco
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- National Research Council, Institute of Neuroscience, Padova, Italy
| |
Collapse
|
13
|
D’Este G, Fabris F, Stazi M, Baggio C, Simonato M, Megighian A, Rigoni M, Negro S, Montecucco C. Agonists of melatonin receptors strongly promote the functional recovery from the neuroparalysis induced by neurotoxic snakes. PLoS Negl Trop Dis 2024; 18:e0011825. [PMID: 38190386 PMCID: PMC10798625 DOI: 10.1371/journal.pntd.0011825] [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: 11/28/2023] [Revised: 01/19/2024] [Accepted: 12/22/2023] [Indexed: 01/10/2024] Open
Abstract
Snake envenoming is a major, but neglected, tropical disease. Among venomous snakes, those inducing neurotoxicity such as kraits (Bungarus genus) cause a potentially lethal peripheral neuroparalysis with respiratory deficit in a large number of people each year. In order to prevent the development of a deadly respiratory paralysis, hospitalization with pulmonary ventilation and use of antivenoms are the primary therapies currently employed. However, hospitals are frequently out of reach for envenomated patients and there is a general consensus that additional, non-expensive treatments, deliverable even long after the snake bite, are needed. Traumatic or toxic degenerations of peripheral motor neurons cause a neuroparalysis that activates a pro-regenerative intercellular signaling program taking place at the neuromuscular junction (NMJ). We recently reported that the intercellular signaling axis melatonin-melatonin receptor 1 (MT1) plays a major role in the recovery of function of the NMJs after degeneration of motor axon terminals caused by massive Ca2+ influx. Here we show that the small chemical MT1 agonists: Ramelteon and Agomelatine, already licensed for the treatment of insomnia and depression, respectively, are strong promoters of the neuroregeneration after paralysis induced by krait venoms in mice, which is also Ca2+ mediated. The venom from a Bungarus species representative of the large class of neurotoxic snakes (including taipans, coral snakes, some Alpine vipers in addition to other kraits) was chosen. The functional recovery of the NMJ was demonstrated using electrophysiological, imaging and lung ventilation detection methods. According to the present results, we propose that Ramelteon and Agomelatine should be tested in human patients bitten by neurotoxic snakes acting presynaptically to promote their recovery of health. Noticeably, these drugs are commercially available, safe, non-expensive, have a long bench life and can be administered long after a snakebite even in places far away from health facilities.
Collapse
Affiliation(s)
- Giorgia D’Este
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Federico Fabris
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Marco Stazi
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Chiara Baggio
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | | | - Aram Megighian
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- Padua Neuroscience Center, University of Padova, Padova, Italy
| | - Michela Rigoni
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- Myology Center (CIR-Myo), University of Padova, Padova, Italy
| | - Samuele Negro
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- U.O.C. Clinica Neurologica, Azienda Ospedale-Università Padova, Padova, Italy
| | - Cesare Montecucco
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- CNR Institute of Neuroscience, Padova, Italy
| |
Collapse
|
14
|
Li W, Liang J, Li S, Jiang S, Song M, Xu S, Wang L, Meng H, Zhai D, Tang L, Yang Y, Zhang B. The CXCL12-CXCR4-NLRP3 axis promotes Schwann cell pyroptosis and sciatic nerve demyelination in rats. Clin Exp Immunol 2023; 214:219-234. [PMID: 37497691 PMCID: PMC10714193 DOI: 10.1093/cei/uxad081] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/06/2023] [Accepted: 07/26/2023] [Indexed: 07/28/2023] Open
Abstract
Studies have shown that the activation of the NOD-like receptor protein 3 (NLRP3) inflammasome is detrimental to the functional recovery of the sciatic nerve, but the regulatory mechanisms of the NLRP3 inflammasome in peripheral nerves are unclear. C-X-C motif chemokine 12 (CXCL12) can bind to C-X-C chemokine receptor type 4 (CXCR4) and participate in a wide range of nerve inflammation by regulating the NLRP3 inflammasome. Based on these, we explore whether CXCL12-CXCR4 axis regulates the NLRP3 inflammasome in the peripheral nerve. We found that CXCR4/CXCL12, NLRP3 inflammasome-related components, pyroptosis-related proteins and inflammatory factors in the sciatic nerve injured rats were markedly increased compared with the sham-operated group. AMD3100, a CXCR4 antagonist, reverses the activation of NLRP3 inflammasome, Schwann cell pyroptosis and sciatic nerve demyelination. We further treated rat Schwann cells with LPS (lipopolysaccharide) and adenosine triphosphate (ATP) to mimic the cellular inflammation model of sciatic nerve injury, and the results were consistent with those in vivo. In addition, both in vivo and in vitro experiments demonstrated that AMD3100 treatment reduced the phosphorylation of nuclear factor κB (NF-κB) and the expression of thioredoxin interacting protein (TXNIP), which contributes to activating NLRP3 inflammasome. Therefore, our findings suggest that, after sciatic nerve injury, CXCL12-CXCR4 axis may promote Schwann cell pyroptosis and sciatic nerve demyelination through activating NLRP3 inflammasome and slow the recovery process of the sciatic nerve.
Collapse
Affiliation(s)
- Wei Li
- Department of Immunology, Medical College of Qingdao University, Qingdao, Shandong Province, China
| | - Jie Liang
- Department of Immunology, Medical College of Qingdao University, Qingdao, Shandong Province, China
| | - Shaohua Li
- Department of Laboratory Medicine, The Third People’s Hospital of Qingdao, Qingdao, Shandong Province, China
| | - Suli Jiang
- Department of Immunology, Medical College of Qingdao University, Qingdao, Shandong Province, China
| | - Meiying Song
- Department of Immunology, Medical College of Qingdao University, Qingdao, Shandong Province, China
| | - Shuo Xu
- Department of Immunology, Medical College of Qingdao University, Qingdao, Shandong Province, China
| | - Luoyang Wang
- Department of Immunology, Medical College of Qingdao University, Qingdao, Shandong Province, China
| | - Haining Meng
- School of Emergency Medicine, Medical College of Qingdao University, Qingdao, Shandong Province, China
| | - Dongchang Zhai
- Department of Special Medicine, School of Basic Medical College, Qingdao University, Qingdao, Shandong Province, China
| | - Lei Tang
- Department of Special Medicine, School of Basic Medical College, Qingdao University, Qingdao, Shandong Province, China
| | - Yanyan Yang
- Department of Immunology, Medical College of Qingdao University, Qingdao, Shandong Province, China
| | - Bei Zhang
- Department of Immunology, Medical College of Qingdao University, Qingdao, Shandong Province, China
| |
Collapse
|
15
|
Theme 04 - In Vivo Experimetal Models. Amyotroph Lateral Scler Frontotemporal Degener 2023; 24:128-139. [PMID: 37966319 DOI: 10.1080/21678421.2023.2260194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2023]
|
16
|
Theme 07 - Pre-Clinical Therapeutic Strategies. Amyotroph Lateral Scler Frontotemporal Degener 2023; 24:173-191. [PMID: 37966323 DOI: 10.1080/21678421.2023.2260198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2023]
|
17
|
Schwann cell functions in peripheral nerve development and repair. Neurobiol Dis 2023; 176:105952. [PMID: 36493976 DOI: 10.1016/j.nbd.2022.105952] [Citation(s) in RCA: 83] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/23/2022] [Accepted: 12/05/2022] [Indexed: 12/12/2022] Open
Abstract
The glial cell of the peripheral nervous system (PNS), the Schwann cell (SC), counts among the most multifaceted cells of the body. During development, SCs secure neuronal survival and participate in axonal path finding. Simultaneously, they orchestrate the architectural set up of the developing nerves, including the blood vessels and the endo-, peri- and epineurial layers. Perinatally, in rodents, SCs radially sort and subsequently myelinate individual axons larger than 1 μm in diameter, while small calibre axons become organised in non-myelinating Remak bundles. SCs have a vital role in maintaining axonal health throughout life and several specialized SC types perform essential functions at specific locations, such as terminal SC at the neuromuscular junction (NMJ) or SC within cutaneous sensory end organs. In addition, neural crest derived satellite glia maintain a tight communication with the soma of sensory, sympathetic, and parasympathetic neurons and neural crest derivatives are furthermore an indispensable part of the enteric nervous system. The remarkable plasticity of SCs becomes evident in the context of a nerve injury, where SC transdifferentiate into intriguing repair cells, which orchestrate a regenerative response that promotes nerve repair. Indeed, the multiple adaptations of SCs are captivating, but remain often ill-resolved on the molecular level. Here, we summarize and discuss the knowns and unknowns of the vast array of functions that this single cell type can cover in peripheral nervous system development, maintenance, and repair.
Collapse
|
18
|
Negro S, Lauria F, Stazi M, Tebaldi T, D’Este G, Pirazzini M, Megighian A, Lessi F, Mazzanti CM, Sales G, Romualdi C, Fillo S, Lista F, Sleigh JN, Tosolini AP, Schiavo G, Viero G, Rigoni M. Hydrogen peroxide induced by nerve injury promotes axon regeneration via connective tissue growth factor. Acta Neuropathol Commun 2022; 10:189. [PMID: 36567321 PMCID: PMC9791753 DOI: 10.1186/s40478-022-01495-5] [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: 08/26/2022] [Accepted: 12/12/2022] [Indexed: 12/26/2022] Open
Abstract
Regeneration of the neuromuscular junction (NMJ) leverages on extensive exchange of factors released from motor axon terminals (MATs), muscle fibers and perisynaptic Schwann cells (PSCs), among which hydrogen peroxide (H2O2) is a major pro-regenerative signal. To identify critical determinants of NMJ remodeling in response to injury, we performed temporal transcriptional profiling of NMJs from 2 month-old mice during MAT degeneration/regeneration, and cross-referenced the differentially expressed genes with those elicited by H2O2 in SCs. We identified an enrichment in extracellular matrix (ECM) transcripts, including Connective Tissue Growth Factor (Ctgf), which is usually expressed during development. We discovered that Ctgf levels are increased in a Yes-associated protein (YAP)-dependent fashion in response to rapid, local H2O2 signaling generated by stressed mitochondria in the injured sciatic nerve, a finding highlighting the importance of signals triggered by mechanical force to motor nerve repair. Through sequestration of Ctgf or inactivation of H2O2, we delayed the recovery of neuromuscular function by impairing SC migration and, in turn, axon-oriented re-growth. These data indicate that H2O2 and its downstream effector Ctgf are pro-regenerative factors that enable axonal growth, and reveal a striking ECM remodeling process during nerve regeneration upon local H2O2 signaling. Our study identifies key transcriptomic changes at the regenerating NMJ, providing a rich source of pro-regenerative factors with potential for alleviating the consequences of peripheral nerve injuries.
Collapse
Affiliation(s)
- Samuele Negro
- grid.5608.b0000 0004 1757 3470Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy ,grid.5608.b0000 0004 1757 3470U.O.C. Clinica Neurologica, Azienda Ospedale, University of Padua, 35128 Padua, Italy
| | - Fabio Lauria
- grid.419463.d0000 0004 1756 3731Institute of Biophysics, CNR Unit at Trento, 38123 Povo, Italy
| | - Marco Stazi
- grid.5608.b0000 0004 1757 3470Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy
| | - Toma Tebaldi
- grid.11696.390000 0004 1937 0351Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123 Povo, Italy ,grid.47100.320000000419368710Section of Hematology, Department of Internal Medicine, Yale Comprehensive Cancer Center, Yale University School of Medicine, New Haven, CT 06520 USA
| | - Giorgia D’Este
- grid.5608.b0000 0004 1757 3470Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy
| | - Marco Pirazzini
- grid.5608.b0000 0004 1757 3470Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy ,grid.5608.b0000 0004 1757 3470Myology Center (CIR-Myo), University of Padua, 35129 Padua, Italy
| | - Aram Megighian
- grid.5608.b0000 0004 1757 3470Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy ,grid.5608.b0000 0004 1757 3470Padua Neuroscience Center, University of Padua, 35131 Padua, Italy
| | - Francesca Lessi
- Laboratory of Genomics, Pisa Science Foundation, 56017 San Giuliano Terme, Italy
| | - Chiara M. Mazzanti
- Laboratory of Genomics, Pisa Science Foundation, 56017 San Giuliano Terme, Italy
| | - Gabriele Sales
- grid.5608.b0000 0004 1757 3470Department of Biology, University of Padua, 35131 Padua, Italy
| | - Chiara Romualdi
- grid.5608.b0000 0004 1757 3470Department of Biology, University of Padua, 35131 Padua, Italy
| | - Silvia Fillo
- grid.470599.60000 0004 1760 920XCenter of Medical and Veterinary Research of the Ministry of Defence, 00184 Rome, Italy
| | - Florigio Lista
- grid.470599.60000 0004 1760 920XCenter of Medical and Veterinary Research of the Ministry of Defence, 00184 Rome, Italy
| | - James N. Sleigh
- grid.83440.3b0000000121901201Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, WC1N 3BG UK ,grid.83440.3b0000000121901201UCL Queen Square Motor Neuron Disease Centre, University College London, London, WC1N 3BG UK ,grid.83440.3b0000000121901201UK Dementia Research Institute, University College London, London, WC1E 6BT UK
| | - Andrew P. Tosolini
- grid.83440.3b0000000121901201Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, WC1N 3BG UK ,grid.83440.3b0000000121901201UCL Queen Square Motor Neuron Disease Centre, University College London, London, WC1N 3BG UK
| | - Giampietro Schiavo
- grid.83440.3b0000000121901201Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, WC1N 3BG UK ,grid.83440.3b0000000121901201UCL Queen Square Motor Neuron Disease Centre, University College London, London, WC1N 3BG UK ,grid.83440.3b0000000121901201UK Dementia Research Institute, University College London, London, WC1E 6BT UK
| | - Gabriella Viero
- grid.419463.d0000 0004 1756 3731Institute of Biophysics, CNR Unit at Trento, 38123 Povo, Italy
| | - Michela Rigoni
- Department of Biomedical Sciences, University of Padua, 35131, Padua, Italy. .,Myology Center (CIR-Myo), University of Padua, 35129, Padua, Italy.
| |
Collapse
|
19
|
Stewart AN, Jones LAT, Gensel JC. Improving translatability of spinal cord injury research by including age as a demographic variable. Front Cell Neurosci 2022; 16:1017153. [PMID: 36467608 PMCID: PMC9714671 DOI: 10.3389/fncel.2022.1017153] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 11/04/2022] [Indexed: 11/18/2022] Open
Abstract
Pre-clinical and clinical spinal cord injury (SCI) studies differ in study design, particularly in the demographic characteristics of the chosen population. In clinical study design, criteria such as such as motor scores, neurological level, and severity of injury are often key determinants for participant inclusion. Further, demographic variables in clinical trials often include individuals from a wide age range and typically include both sexes, albeit historically most cases of SCI occur in males. In contrast, pre-clinical SCI models predominately utilize young adult rodents and typically use only females. While it is often not feasible to power SCI clinical trials to test multi-variable designs such as contrasting different ages, recent pre-clinical findings in SCI animal models have emphasized the importance of considering age as a biological variable prior to human experiments. Emerging pre-clinical data have identified case examples of treatments that diverge in efficacy across different demographic variables and have elucidated several age-dependent effects in SCI. The extent to which these differing or diverging treatment responses manifest clinically can not only complicate statistical findings and trial interpretations but also may be predictive of worse outcomes in select clinical populations. This review highlights recent literature including age as a biological variable in pre-clinical studies and articulates the results with respect to implications for clinical trials. Based on emerging unpredictable treatment outcomes in older rodents, we argue for the importance of including age as a biological variable in pre-clinical animal models prior to clinical testing. We believe that careful analyses of how age interacts with SCI treatments and pathophysiology will help guide clinical trial design and may improve both the safety and outcomes of such important efforts.
Collapse
Affiliation(s)
- Andrew N. Stewart
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, United States,Spinal Cord and Brain Injury Research Center, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Linda A. T. Jones
- Center for Outcomes and Measurement, Jefferson College of Rehabilitation Sciences, Thomas Jefferson University, Philadelphia, PA, United States
| | - John C. Gensel
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, United States,Spinal Cord and Brain Injury Research Center, College of Medicine, University of Kentucky, Lexington, KY, United States,*Correspondence: John C. Gensel,
| |
Collapse
|
20
|
Negro S, Pirazzini M, Rigoni M. Models and methods to study Schwann cells. J Anat 2022; 241:1235-1258. [PMID: 34988978 PMCID: PMC9558160 DOI: 10.1111/joa.13606] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 11/26/2021] [Accepted: 11/29/2021] [Indexed: 12/22/2022] Open
Abstract
Schwann cells (SCs) are fundamental components of the peripheral nervous system (PNS) of all vertebrates and play essential roles in development, maintenance, function, and regeneration of peripheral nerves. There are distinct populations of SCs including: (1) myelinating SCs that ensheath axons by a specialized plasma membrane, called myelin, which enhances the conduction of electric impulses; (2) non-myelinating SCs, including Remak SCs, which wrap bundles of multiple axons of small caliber, and perysinaptic SCs (PSCs), associated with motor axon terminals at the neuromuscular junction (NMJ). All types of SCs contribute to PNS regeneration through striking morphological and functional changes in response to nerve injury, are affected in peripheral neuropathies and show abnormalities and a diminished plasticity during aging. Therefore, methodological approaches to study and manipulate SCs in physiological and pathophysiological conditions are crucial to expand the present knowledge on SC biology and to devise new therapeutic strategies to counteract neurodegenerative conditions and age-derived denervation. We present here an updated overview of traditional and emerging methodologies for the study of SCs for scientists approaching this research field.
Collapse
Affiliation(s)
- Samuele Negro
- Department of Biomedical SciencesUniversity of PaduaPaduaItaly
| | - Marco Pirazzini
- Department of Biomedical SciencesUniversity of PaduaPaduaItaly
- CIR‐MyoCentro Interdipartimentale di Ricerca di MiologiaUniversity of PaduaPadovaItaly
| | - Michela Rigoni
- Department of Biomedical SciencesUniversity of PaduaPaduaItaly
- CIR‐MyoCentro Interdipartimentale di Ricerca di MiologiaUniversity of PaduaPadovaItaly
| |
Collapse
|
21
|
Research progress of targeting NLRP3 inflammasome in peripheral nerve injury and pain. Int Immunopharmacol 2022; 110:109026. [DOI: 10.1016/j.intimp.2022.109026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 06/25/2022] [Accepted: 06/30/2022] [Indexed: 01/08/2023]
|
22
|
Recovery from the Neuroparalysis Caused by the Micrurus nigrocinctus Venom Is Accelerated by an Agonist of the CXCR4 Receptor. Toxins (Basel) 2022; 14:toxins14080531. [PMID: 36006193 PMCID: PMC9412299 DOI: 10.3390/toxins14080531] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/22/2022] [Accepted: 07/30/2022] [Indexed: 12/10/2022] Open
Abstract
Snake envenoming is a major but neglected human disease in tropical and subtropical regions. Among venomous snakes in the Americas, coral snakes of the genus Micrurus are particularly dangerous because they cause a peripheral neuroparalysis that can persist for many days or, in severe cases, progress to death. Ventilatory support and the use of snake species-specific antivenoms may prevent death from respiratory paralysis in most cases. However, there is a general consensus that additional and non-expensive treatments that can be delivered even long after the snake bite are needed. Neurotoxic degeneration of peripheral motor neurons activates pro-regenerative intercellular signaling programs, the greatest of which consist of the chemokine CXCL12α, produced by perisynaptic Schwann cells, which act on the CXCR4 receptor expressed on damaged neuronal axons. We recently found that the CXCR4 agonist NUCC-390 promotes axonal growth. Here, we show that the venom of the highly neurotoxic snake Micrurus nigrocinctus causes a complete degeneration of motor axon terminals of the soleus muscle, followed by functional regeneration whose time course is greatly accelerated by NUCC-390. These results suggest that NUCC-390 is a potential candidate for treating human patients envenomed by Micrurus nigrocinctus as well as other neurotoxic Micrurus spp. in order to improve the recovery of normal neuromuscular physiology, thus reducing the mortality and hospital costs of envenoming.
Collapse
|
23
|
Perez-Gonzalez AP, Provost F, Rousse I, Piovesana R, Benzina O, Darabid H, Lamoureux B, Wang YS, Arbour D, Robitaille R. Functional adaptation of glial cells at neuromuscular junctions in response to injury. Glia 2022; 70:1605-1629. [PMID: 35474470 PMCID: PMC9543218 DOI: 10.1002/glia.24184] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 04/14/2022] [Accepted: 04/18/2022] [Indexed: 12/04/2022]
Abstract
Synaptic elements from neuromuscular junctions (NMJs) undergo massive morphological and functional changes upon nerve injury. While morphological changes of NMJ‐associated glia in response to injury has been investigated, their functional properties remain elusive. Perisynaptic Schwann cells (PSCs), glial cells at the NMJ, are essential for NMJ maintenance and repair, and are involved in synaptic efficacy and plasticity. Importantly, these functions are regulated by PSCs ability to detect synaptic transmission through, notably, muscarinic (mAChRs) and purinergic receptors' activation. Using Ca2+ imaging and electrophysiological recordings of synaptic transmission at the mouse NMJ, we investigated PSC receptors activation following denervation and during reinnervation in adults and at denervated NMJs in an ALS mouse model (SOD1G37R). We observed reduced PSCs mAChR‐mediated Ca2+ responses at denervated and reinnervating NMJs. Importantly, PSC phenotypes during denervation and reinnervation were distinct than the one observed during NMJ maturation. At denervated NMJs, exogenous activation of mAChRs greatly diminished galectin‐3 expression, a glial marker of phagocytosis. PSCs Ca2+ responses at reinnervating NMJs did not correlate with the number of innervating axons or process extensions. Interestingly, we observed an extended period of reduced PSC mAChRs activation after the injury (up to 60 days), suggesting a glial memory of injury. PSCs associated with denervated NMJs in an ALS model (SOD1G37R mice) did not show any muscarinic adaptation, a phenotype incompatible with NMJ repair. Understanding functional mechanisms that underlie this glial response to injury may contribute to favor complete NMJ and motor recovery.
Collapse
Affiliation(s)
- Anna P Perez-Gonzalez
- Département de Neurosciences, Université de Montréal, Montréal, Québec, Canada.,Groupe de Recherche sur le Système Nerveux Central, Université de Montréal, Montréal, Québec, Canada
| | - Frédéric Provost
- Département de Neurosciences, Université de Montréal, Montréal, Québec, Canada.,Groupe de Recherche sur le Système Nerveux Central, Université de Montréal, Montréal, Québec, Canada
| | - Isabelle Rousse
- Département de Neurosciences, Université de Montréal, Montréal, Québec, Canada.,Groupe de Recherche sur le Système Nerveux Central, Université de Montréal, Montréal, Québec, Canada
| | - Roberta Piovesana
- Département de Neurosciences, Université de Montréal, Montréal, Québec, Canada.,Groupe de Recherche sur le Système Nerveux Central, Université de Montréal, Montréal, Québec, Canada
| | - Ouafa Benzina
- Département de Neurosciences, Université de Montréal, Montréal, Québec, Canada.,Groupe de Recherche sur le Système Nerveux Central, Université de Montréal, Montréal, Québec, Canada
| | - Houssam Darabid
- Département de Neurosciences, Université de Montréal, Montréal, Québec, Canada.,Groupe de Recherche sur le Système Nerveux Central, Université de Montréal, Montréal, Québec, Canada
| | - Benoit Lamoureux
- Département de Neurosciences, Université de Montréal, Montréal, Québec, Canada.,Groupe de Recherche sur le Système Nerveux Central, Université de Montréal, Montréal, Québec, Canada
| | - Yu Shi Wang
- Département de Neurosciences, Université de Montréal, Montréal, Québec, Canada.,Groupe de Recherche sur le Système Nerveux Central, Université de Montréal, Montréal, Québec, Canada
| | - Danielle Arbour
- Département de Neurosciences, Université de Montréal, Montréal, Québec, Canada.,Groupe de Recherche sur le Système Nerveux Central, Université de Montréal, Montréal, Québec, Canada
| | - Richard Robitaille
- Département de Neurosciences, Université de Montréal, Montréal, Québec, Canada.,Groupe de Recherche sur le Système Nerveux Central, Université de Montréal, Montréal, Québec, Canada.,Centre Interdisciplinaire de Recherche sur le Cerveau et l'apprentissage, Montréal, Québec, Canada
| |
Collapse
|
24
|
Fabris F, Šoštarić P, Matak I, Binz T, Toffan A, Simonato M, Montecucco C, Pirazzini M, Rossetto O. Detection of VAMP Proteolysis by Tetanus and Botulinum Neurotoxin Type B In Vivo with a Cleavage-Specific Antibody. Int J Mol Sci 2022; 23:ijms23084355. [PMID: 35457172 PMCID: PMC9024618 DOI: 10.3390/ijms23084355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/08/2022] [Accepted: 04/12/2022] [Indexed: 11/17/2022] Open
Abstract
Tetanus and Botulinum type B neurotoxins are bacterial metalloproteases that specifically cleave the vesicle-associated membrane protein VAMP at an identical peptide bond, resulting in inhibition of neuroexocytosis. The minute amounts of these neurotoxins commonly used in experimental animals are not detectable, nor is detection of their VAMP substrate sensitive enough. The immune detection of the cleaved substrate is much more sensitive, as we have previously shown for botulinum neurotoxin type A. Here, we describe the production in rabbit of a polyclonal antibody raised versus a peptide encompassing the 13 residues C-terminal with respect to the neurotoxin cleavage site. The antibody was affinity purified and found to recognize, with high specificity and selectivity, the novel N-terminus of VAMP that becomes exposed after cleavage by tetanus toxin and botulinum toxin type B. This antibody recognizes the neoepitope not only in native and denatured VAMP but also in cultured neurons and in neurons in vivo in neurotoxin-treated mice or rats, suggesting the great potential of this novel tool to elucidate tetanus and botulinum B toxin activity in vivo.
Collapse
Affiliation(s)
- Federico Fabris
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy; (F.F.); (C.M.)
| | - Petra Šoštarić
- Department of Pharmacology, School of Medicine, University of Zagreb, Šalata 11, 10000 Zagreb, Croatia; (P.Š.); (I.M.)
| | - Ivica Matak
- Department of Pharmacology, School of Medicine, University of Zagreb, Šalata 11, 10000 Zagreb, Croatia; (P.Š.); (I.M.)
| | - Thomas Binz
- Institute of Cellular Biochemistry, Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany;
| | - Anna Toffan
- Istituto Zooprofilattico Sperimentale delle Venezie, Viale dell’Università 10, 35020 Legnaro, Italy;
| | - Morena Simonato
- Institute of Neuroscience, Italian Research Council, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy;
| | - Cesare Montecucco
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy; (F.F.); (C.M.)
- Institute of Neuroscience, Italian Research Council, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy;
| | - Marco Pirazzini
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy; (F.F.); (C.M.)
- Interdepartmental Research Center of Myology CIR-Myo, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy
- Correspondence: (M.P.); (O.R.)
| | - Ornella Rossetto
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy; (F.F.); (C.M.)
- Institute of Neuroscience, Italian Research Council, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy;
- Interdepartmental Research Center of Myology CIR-Myo, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy
- Correspondence: (M.P.); (O.R.)
| |
Collapse
|
25
|
Stazi M, Fabris F, Tan KY, Megighian A, Rubini A, Mattarei A, Negro S, D'Este G, Lista F, Rossetto O, Tan CH, Montecucco C. An agonist of the CXCR4 receptor is therapeutic for the neuroparalysis induced by
Bungarus
snakes envenoming. Clin Transl Med 2022; 12:e651. [PMID: 35075797 PMCID: PMC8787102 DOI: 10.1002/ctm2.651] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/27/2021] [Accepted: 10/31/2021] [Indexed: 11/08/2022] Open
Affiliation(s)
- Marco Stazi
- Department of Biomedical Sciences University of Padova Padova Italy
| | - Federico Fabris
- Department of Biomedical Sciences University of Padova Padova Italy
| | - Kae Yi Tan
- Department of Molecular Medicine Faculty of Medicine University of Malaya Kuala Lumpur Malaysia
| | - Aram Megighian
- Department of Biomedical Sciences University of Padova Padova Italy
| | | | - Andrea Mattarei
- Department of Pharmaceutical and Pharmacological Sciences University of Padova Padova Italy
| | - Samuele Negro
- Department of Biomedical Sciences University of Padova Padova Italy
| | - Giorgia D'Este
- Department of Biomedical Sciences University of Padova Padova Italy
| | - Florigio Lista
- Center of Medical and Veterinary Research of the Ministry of Defense Policlinico Militare Rome Italy
| | - Ornella Rossetto
- Department of Biomedical Sciences University of Padova Padova Italy
| | - Choo Hock Tan
- Department of Pharmacology Faculty of Medicine University of Malaya Kuala Lumpur Malaysia
| | - Cesare Montecucco
- Department of Biomedical Sciences University of Padova Padova Italy
- CNR Institute of Neuroscience Padova Italy
| |
Collapse
|
26
|
Latrotoxin-Induced Neuromuscular Junction Degeneration Reveals Urocortin 2 as a Critical Contributor to Motor Axon Terminal Regeneration. Int J Mol Sci 2022; 23:ijms23031186. [PMID: 35163106 PMCID: PMC8835473 DOI: 10.3390/ijms23031186] [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: 12/23/2021] [Revised: 01/18/2022] [Accepted: 01/20/2022] [Indexed: 11/24/2022] Open
Abstract
We used α-Latrotoxin (α-LTx), the main neurotoxic component of the black widow spider venom, which causes degeneration of the neuromuscular junction (NMJ) followed by a rapid and complete regeneration, as a molecular tool to identify by RNA transcriptomics factors contributing to the structural and functional recovery of the NMJ. We found that Urocortin 2 (UCN2), a neuropeptide involved in the stress response, is rapidly expressed at the NMJ after acute damage and that inhibition of CRHR2, the specific receptor of UCN2, delays neuromuscular transmission rescue. Experiments in neuronal cultures show that CRHR2 localises at the axonal tips of growing spinal motor neurons and that its expression inversely correlates with synaptic maturation. Moreover, exogenous UCN2 enhances the growth of axonal sprouts in cultured neurons in a CRHR2-dependent manner, pointing to a role of the UCN2-CRHR2 axis in the regulation of axonal growth and synaptogenesis. Consistently, exogenous administration of UCN2 strongly accelerates the regrowth of motor axon terminals degenerated by α-LTx, thereby contributing to the functional recovery of neuromuscular transmission after damage. Taken together, our results posit a novel role for UCN2 and CRHR2 as a signalling axis involved in NMJ regeneration.
Collapse
|
27
|
Terheyden-Keighley D, Hilla AM, Fischer D. CXCR4 signaling in central nervous system regeneration: friend or foe? Neural Regen Res 2021; 17:1481-1483. [PMID: 34916425 PMCID: PMC8771105 DOI: 10.4103/1673-5374.330605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Affiliation(s)
- Daniel Terheyden-Keighley
- Department of Cell Physiology, Ruhr University of Bochum, Universitätsstraβe 150, 44780 Bochum, Germany
| | - Alexander M Hilla
- Department of Cell Physiology, Ruhr University of Bochum, Universitätsstraβe 150, 44780 Bochum, Germany
| | - Dietmar Fischer
- Department of Cell Physiology, Ruhr University of Bochum, Universitätsstraβe 150, 44780 Bochum, Germany
| |
Collapse
|
28
|
Hörner SJ, Couturier N, Bruch R, Koch P, Hafner M, Rudolf R. hiPSC-Derived Schwann Cells Influence Myogenic Differentiation in Neuromuscular Cocultures. Cells 2021; 10:cells10123292. [PMID: 34943800 PMCID: PMC8699767 DOI: 10.3390/cells10123292] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/20/2021] [Accepted: 11/21/2021] [Indexed: 12/13/2022] Open
Abstract
Motoneurons, skeletal muscle fibers, and Schwann cells form synapses, termed neuromuscular junctions (NMJs). These control voluntary body movement and are affected in numerous neuromuscular diseases. Therefore, a variety of NMJ in vitro models have been explored to enable mechanistic and pharmacological studies. So far, selective integration of Schwann cells in these models has been hampered, due to technical limitations. Here we present robust protocols for derivation of Schwann cells from human induced pluripotent stem cells (hiPSC) and their coculture with hiPSC-derived motoneurons and C2C12 muscle cells. Upon differentiation with tuned BMP signaling, Schwann cells expressed marker proteins, S100b, Gap43, vimentin, and myelin protein zero. Furthermore, they displayed typical spindle-shaped morphologies with long processes, which often aligned with motoneuron axons. Inclusion of Schwann cells in coculture experiments with hiPSC-derived motoneurons and C2C12 myoblasts enhanced myotube growth and affected size and number of acetylcholine receptor plaques on myotubes. Altogether, these data argue for the availability of a consistent differentiation protocol for Schwann cells and their amenability for functional integration into neuromuscular in vitro models, fostering future studies of neuromuscular mechanisms and disease.
Collapse
Affiliation(s)
- Sarah Janice Hörner
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany; (S.J.H.); (N.C.); (R.B.); (M.H.)
- Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Nathalie Couturier
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany; (S.J.H.); (N.C.); (R.B.); (M.H.)
| | - Roman Bruch
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany; (S.J.H.); (N.C.); (R.B.); (M.H.)
| | - Philipp Koch
- Central Institute of Mental Health, Medical Faculty Mannheim of Heidelberg University, 68159 Mannheim, Germany;
- Hector Institute for Translational Brain Research (HITBR gGmbH), 68159 Mannheim, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Mathias Hafner
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany; (S.J.H.); (N.C.); (R.B.); (M.H.)
- Institute of Medical Technology, Mannheim University of Applied Sciences and Heidelberg University, 68163 Mannheim, Germany
| | - Rüdiger Rudolf
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany; (S.J.H.); (N.C.); (R.B.); (M.H.)
- Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany
- Institute of Medical Technology, Mannheim University of Applied Sciences and Heidelberg University, 68163 Mannheim, Germany
- Correspondence:
| |
Collapse
|
29
|
Huang G, Hu M, Lu D, Hong L. Protective effect and potential mechanism of Schwann cell-derived exosomes on mechanical damage of rat dorsal root ganglion cells. J Obstet Gynaecol Res 2021; 47:3691-3701. [PMID: 34365704 DOI: 10.1111/jog.14968] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 07/19/2021] [Accepted: 07/23/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND Pudendal nerve (PN) injury was one of the most important pathogenesis of stress urinary incontinence (SUI). Schwann cell (SC)-derived exosomes could promote axonal regeneration. Wnt protein could significantly promote axonal regeneration and participate in the regulation of proliferation and differentiation of neural stem cells. Therefore, we sought to determine whether SCs-derived exosomes might also protect against damaged dorsal root ganglion cells (DRGs) through the Wnt/β-catenin pathway. MATERIAL AND METHODS The DRGs injury model was fabricated using a four-point bending system. The exosomes were separated from the SCs supernatant. XAV939, which was a small molecule inhibitor, was used to inhibit the Wnt/β-catenin pathway. Next, Cell Counting Kit-8 (CCK8) kit was used to detect cell activity. We evaluated the proliferative activity of DRG cells using the cell cycle and apoptosis detection kit. We assessed the cell apoptotic rates through the Annexin V/PE double staining. Finally, we detect the expression of downstream proteins of Wnt/β-catenin pathway in DRG cells using western blotting. RESULTS SC-derived exosomes had protective effects on DRGs after mechanical damage, which could promote cell proliferation, transition of the cell cycle to the G2 phase, and inhibit cell apoptosis. Exogenous administration of XAV939 suppressed the promoting effect of SCs -derived exosomes on DRG cells and the expression of downstream proteins of Wnt/β-catenin pathway in DRG cells was also suppressed. CONCLUSION These results suggested that SC-derived exosomes have a repairing effect on DRG cells injury caused by cyclic mechanical stretching (CMS) and the Wnt/β-catenin pathway is potentially involved in the process.
Collapse
Affiliation(s)
- Guotao Huang
- Dept. of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ming Hu
- Dept. of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Danhua Lu
- Dept. of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Li Hong
- Dept. of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
30
|
CXCL12 promotes spinal nerve regeneration and functional recovery after spinal cord injury. Neuroreport 2021; 32:450-457. [PMID: 33657074 DOI: 10.1097/wnr.0000000000001613] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Spinal cord injury (SCI) leads to permanent loss of motor and sensory function due to the complex mechanisms of the external microenvironment and internal neurobiochemistry that restrict neuronal plasticity and axonal regeneration. Chemokine CXCL12 was verified in regulating the development of central nervous system (CNS) and repairing of CNS disease. In the present study, CXCL12 was downregulated in the spinal cord after SCI. SCI also induced gliosis and loss of synapse. Intrathecal treatment of CXCL12 promoted the functional recovery of SCI by inducing the formation of neuronal connections and suppressing glia scar. To confirm whether CXCL12 promoted synapse formation and functional neuronal connections, the primary cortical neurons were treated with CXCL12 peptide, the synapse was examined using Immunofluorescence staining and the function of synapse was tested using a whole-cell patch clamp. The results indicated that CXCL12 peptide promoted axonal elongation, branche formation, dendrite generation and synaptogenesis. The electrophysiological results showed that CXCL12 peptide increased functional connections among neurons. Taken together, the present study illustrated an underlying mechanism of the development of SCI and indicated a potential approach to facilitate functional recovery of spinal cord after SCI.
Collapse
|
31
|
Rios R, Jablonka-Shariff A, Broberg C, Snyder-Warwick AK. Macrophage roles in peripheral nervous system injury and pathology: Allies in neuromuscular junction recovery. Mol Cell Neurosci 2021; 111:103590. [PMID: 33422671 DOI: 10.1016/j.mcn.2021.103590] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 12/15/2020] [Accepted: 01/01/2021] [Indexed: 12/11/2022] Open
Abstract
Peripheral nerve injuries remain challenging to treat despite extensive research on reparative processes at the injury site. Recent studies have emphasized the importance of immune cells, particularly macrophages, in recovery from nerve injury. Macrophage plasticity enables numerous functions at the injury site. At early time points, macrophages perform inflammatory functions, but at later time points, they adopt pro-regenerative phenotypes to support nerve regeneration. Research has largely been limited, however, to the injury site. The neuromuscular junction (NMJ), the synapse between the nerve terminal and end target muscle, has received comparatively less attention, despite the importance of NMJ reinnervation for motor recovery. Macrophages are present at the NMJ following nerve injury. Moreover, in denervating diseases, such as amyotrophic lateral sclerosis (ALS), macrophages may also play beneficial roles at the NMJ. Evidence of positive macrophages roles at the injury site after peripheral nerve injury and at the NMJ in denervating pathologies suggest that macrophages may promote NMJ reinnervation. In this review, we discuss the intersection of nerve injury and immunity, with a focus on macrophages.
Collapse
Affiliation(s)
- Rachel Rios
- Washington University School of Medicine, St. Louis, MO, United States of America
| | - Albina Jablonka-Shariff
- Division of Plastic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Curtis Broberg
- Washington University School of Medicine, St. Louis, MO, United States of America
| | - Alison K Snyder-Warwick
- Division of Plastic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, United States of America.
| |
Collapse
|
32
|
Stazi M, Negro S, Megighian A, D'Este G, Solimena M, Jockers R, Lista F, Montecucco C, Rigoni M. Melatonin promotes regeneration of injured motor axons via MT 1 receptors. J Pineal Res 2021; 70:e12695. [PMID: 32939783 PMCID: PMC7757164 DOI: 10.1111/jpi.12695] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 09/07/2020] [Accepted: 09/08/2020] [Indexed: 01/03/2023]
Abstract
Melatonin is an ancient multi-tasking molecule produced by the pineal gland and by several extrapineal tissues. A variety of activities has been ascribed to this hormone in different physiological and pathological contexts, but little is known about its role in peripheral neuroregeneration. Here, we have exploited two different types of injury to test the capability of melatonin to stimulate regeneration of motor axons: (a) the acute and reversible presynaptic degeneration induced by the spider neurotoxin α-Latrotoxin and (b) the compression/transection of the sciatic nerve. We found that in both cases melatonin administration accelerates the process of nerve repair. This pro-regenerative action is MT1 -mediated, and at least in part due to a sustained activation of the ERK1/2 pathway. These findings reveal a receptor-mediated, pro-regenerative action of melatonin in vivo that holds important clinical implications, as it posits melatonin as a safe candidate molecule for the treatment of a number of peripheral neurodegenerative conditions.
Collapse
Affiliation(s)
- Marco Stazi
- Department of Biomedical SciencesUniversity of PaduaPaduaItaly
| | - Samuele Negro
- Department of Biomedical SciencesUniversity of PaduaPaduaItaly
| | - Aram Megighian
- Department of Biomedical SciencesUniversity of PaduaPaduaItaly
- Padua Neuroscience InstitutePaduaItaly
| | - Giorgia D'Este
- Department of Biomedical SciencesUniversity of PaduaPaduaItaly
| | - Michele Solimena
- Molecular DiabetologyFaculty of MedicineUniversity Hospital, TU DresdenDresdenGermany
- Faculty of MedicinePaul Langerhans Institute Dresden (PLID) of the Helmholtz Center Munich at University Hospital Carl Gustav Carus, TU DresdenDresdenGermany
- German Center for Diabetes Research (DZD)Munich NeuherbergGermany
- Max Planck Institute of Molecular Cell Biology and Genetics (MPI‐CBG)DresdenGermany
| | - Ralf Jockers
- Institut CochinCNRSINSERMUniversité de ParisParisFrance
| | - Florigio Lista
- Center of Medical and Veterinary Research of the Ministry of DefenceRomeItaly
| | - Cesare Montecucco
- Department of Biomedical SciencesUniversity of PaduaPaduaItaly
- CNR Institute of NeurosciencePaduaItaly
| | - Michela Rigoni
- Department of Biomedical SciencesUniversity of PaduaPaduaItaly
| |
Collapse
|
33
|
Hou J, Wang C, Ma D, Chen Y, Jin H, An Y, Jia J, Huang L, Zhao H. The cardioprotective and anxiolytic effects of Chaihujialonggumuli granule on rats with anxiety after acute myocardial infarction is partly mediated by suppression of CXCR4/NF-κB/GSDMD pathway. Biomed Pharmacother 2021; 133:111015. [PMID: 33232924 DOI: 10.1016/j.biopha.2020.111015] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 11/04/2020] [Accepted: 11/11/2020] [Indexed: 12/17/2022] Open
Abstract
AIMS Over-expression of CXCR4 activates nuclear translocation of NF-κB, induces high expression of NLRP3, GSDMD, IL-1β and IL-18, which promotes severe inflammatory response following myocardial infarction. Previous studies revealed inflammation induces anxiety after myocardial infarction. The Chaihujialonggumuli granule has anti-inflammatory properties and could tranquillize mind. But the mechanism of its efficacy remains unknown. This study was to investigate the possible mechanism of BFG on cardioprotective and anxiolytic. METHODS The expression of CXCR4, NF-κB, NLRP3and GSDMD was measured with western-blot, QRT-PCR. The expression location of CXCR4, NLRP3, GSDMD were determined by immunohistochemistry. IL-1β、IL-18 in the peripheral blood were measured by ELISA. HE staining, Masson staining and transmission electron microscopy were used to observe morphological changes of cardiomyocytes. Echocardiography was used to assess cardiac function after cardiac surgery. Elevated cross maze test and open field test were used to evaluate behaviours. Western blot was used to detect the protein expressions of 5-HT, DA, IL-1β, IL-18 and neuron damage was investigated by Nissl staining in the hippocampus. RESULTS The up-regulation of CXCR4, NF-κB, NLRP3 and GSDMD were found in the infarcted area after left coronary artery ligation. Pathological staining and analysis showed that more severe inflammatory cytokines infiltration, myocardial fibrosis, were found in myocardial tissue of the complex group rats. And when compared to the sham group, the levels of IL-1β, IL-18 was increased of the complex group in both peripheral blood and brain. Behavioural test and echocardiography indicated that the rats in complex group exploration behaviours was significantly reduced, and with poor cardiac functional recovery. The AMD3100 had an inhibitory impact of CXCR4 on the activition of its downstream effectors, alleviating inflammatory reaction. Furthermore, the BFG decreased the expression level of CXCR4, NF-κB, GSDMD, NLRP3 in the infarcted area after myocardial infarction, when compared to the complex group. The assays in the brain indicated the BFG suppressed expression and activity of IL-1β, IL-18, and improved 5-HT and DA synthesis. CONCLUSIONS In sum, our study indicated that BFG may reduce inflammation, treat co-existing anxiety after myocardial infarction through inhibition of CXCR4/NF-κB/GSDMD signalling.
Collapse
Affiliation(s)
- Jiqiu Hou
- The Third Affiliate Hospital of Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Chao Wang
- The DongFang Hospital of Beijing University of Chinese Medicine, Beijing, 100078, China
| | - Di Ma
- The Third Affiliate Hospital of Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yali Chen
- The Third Affiliate Hospital of Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Huihui Jin
- The Third Affiliate Hospital of Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Ying An
- The Third Affiliate Hospital of Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Jingyun Jia
- The Third Affiliate Hospital of Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Lexi Huang
- The Third Affiliate Hospital of Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Haibin Zhao
- The DongFang Hospital of Beijing University of Chinese Medicine, Beijing, 100078, China.
| |
Collapse
|
34
|
Zelada D, Bermedo-García F, Collao N, Henríquez JP. Motor function recovery: deciphering a regenerative niche at the neuromuscular synapse. Biol Rev Camb Philos Soc 2020; 96:752-766. [PMID: 33336525 PMCID: PMC7986695 DOI: 10.1111/brv.12675] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 11/20/2020] [Accepted: 11/25/2020] [Indexed: 12/13/2022]
Abstract
The coordinated movement of many organisms relies on efficient nerve–muscle communication at the neuromuscular junction (NMJ), a peripheral synapse composed of a presynaptic motor axon terminal, a postsynaptic muscle specialization, and non‐myelinating terminal Schwann cells. NMJ dysfunctions are caused by traumatic spinal cord or peripheral nerve injuries as well as by severe motor pathologies. Compared to the central nervous system, the peripheral nervous system displays remarkable regenerating abilities; however, this capacity is limited by the denervation time frame and depends on the establishment of permissive regenerative niches. At the injury site, detailed information is available regarding the cells, molecules, and mechanisms involved in nerve regeneration and repair. However, a regenerative niche at the final functional step of peripheral motor innervation, i.e. at the mature neuromuscular synapse, has not been deciphered. In this review, we integrate classic and recent evidence describing the cells and molecules that could orchestrate a dynamic ecosystem to accomplish successful NMJ regeneration. We propose that such a regenerative niche must ensure at least two fundamental steps for successful NMJ regeneration: the proper arrival of incoming regenerating axons to denervated postsynaptic muscle domains, and the resilience of those postsynaptic domains, in morphological and functional terms. We here describe and combine the main cellular and molecular responses involved in each of these steps as potential targets to help successful NMJ regeneration.
Collapse
Affiliation(s)
- Diego Zelada
- Neuromuscular Studies Laboratory (NeSt Lab), Department of Cell Biology, Faculty of Biological Sciences, Center for Advanced Microscopy (CMA Bio-Bio), Universidad de Concepción, Casilla 160-C, Concepción, Chile
| | - Francisca Bermedo-García
- Neuromuscular Studies Laboratory (NeSt Lab), Department of Cell Biology, Faculty of Biological Sciences, Center for Advanced Microscopy (CMA Bio-Bio), Universidad de Concepción, Casilla 160-C, Concepción, Chile
| | - Nicolás Collao
- Neuromuscular Studies Laboratory (NeSt Lab), Department of Cell Biology, Faculty of Biological Sciences, Center for Advanced Microscopy (CMA Bio-Bio), Universidad de Concepción, Casilla 160-C, Concepción, Chile
| | - Juan P Henríquez
- Neuromuscular Studies Laboratory (NeSt Lab), Department of Cell Biology, Faculty of Biological Sciences, Center for Advanced Microscopy (CMA Bio-Bio), Universidad de Concepción, Casilla 160-C, Concepción, Chile
| |
Collapse
|
35
|
Trolese MC, Mariani A, Terao M, de Paola M, Fabbrizio P, Sironi F, Kurosaki M, Bonanno S, Marcuzzo S, Bernasconi P, Trojsi F, Aronica E, Bendotti C, Nardo G. CXCL13/CXCR5 signalling is pivotal to preserve motor neurons in amyotrophic lateral sclerosis. EBioMedicine 2020; 62:103097. [PMID: 33161233 PMCID: PMC7670099 DOI: 10.1016/j.ebiom.2020.103097] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 10/13/2020] [Accepted: 10/13/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND CXCL13 is a B and T lymphocyte chemokine that mediates neuroinflammation through its receptor CXCR5. This chemokine is highly expressed by motoneurons (MNs) in Amyotrophic Lateral Sclerosis (ALS) SOD1G93A (mSOD1) mice during the disease, particularly in fast-progressing mice. Accordingly, in this study, we investigated the role of this chemokine in ALS. METHODS We used in vitro and in vivo experimental paradigms derived from ALS mice and patients to investigate the expression level and distribution of CXCL13/CXCR5 axis and its role in MN death and disease progression. Moreover, we compared the levels of CXCL13 in the CSF and serum of ALS patients and controls. FINDINGS CXCL13 and CXCR5 are overexpressed in the spinal MNs and peripheral axons in mSOD1 mice. CXCL13 inhibition in the CNS of ALS mice resulted in the exacerbation of motor impairment (n = 4/group;Mean_Diff.=27.81) and decrease survival (n = 14_Treated:19.2 ± 1.05wks, n = 17_Controls:20.2 ± 0.6wks; 95% CI: 0.4687-1.929). This was corroborated by evidence from primary spinal cultures where the inhibition or activation of CXCL13 exacerbated or prevented the MN loss. Besides, we found that CXCL13/CXCR5 axis is overexpressed in the spinal cord MNs of ALS patients, and CXCL13 levels in the CSF discriminate ALS (n = 30) from Multiple Sclerosis (n = 16) patients with a sensitivity of 97.56%. INTERPRETATION We hypothesise that MNs activate CXCL13 signalling to attenuate CNS inflammation and prevent the neuromuscular denervation. The low levels of CXCL13 in the CSF of ALS patients might reflect the MN dysfunction, suggesting this chemokine as a potential clinical adjunct to discriminate ALS from other neurological diseases. FUNDING Vaccinex, Inc.; Regione Lombardia (TRANS-ALS).
Collapse
Affiliation(s)
- Maria Chiara Trolese
- Laboratory of Molecular Neurobiology, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, Milan 20156, Italy
| | - Alessandro Mariani
- Laboratory of Biology of Neurodegenerative Disorders, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, Milan 20156, Italy
| | - Mineko Terao
- Laboratory of Molecular Biology, Department of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche IRCCS, Via Mario Negri 2, Milan 20156, Italy
| | - Massimiliano de Paola
- Laboratory of Biology of Neurodegenerative Disorders, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, Milan 20156, Italy
| | - Paola Fabbrizio
- Laboratory of Molecular Neurobiology, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, Milan 20156, Italy
| | - Francesca Sironi
- Laboratory of Molecular Neurobiology, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, Milan 20156, Italy
| | - Mami Kurosaki
- Laboratory of Molecular Biology, Department of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche IRCCS, Via Mario Negri 2, Milan 20156, Italy
| | - Silvia Bonanno
- Neurology IV-Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, Milan 20133, Italy
| | - Stefania Marcuzzo
- Neurology IV-Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, Milan 20133, Italy
| | - Pia Bernasconi
- Neurology IV-Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, Milan 20133, Italy
| | - Francesca Trojsi
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", P.zza Miraglia 2, Naples 80138, Italy
| | - Eleonora Aronica
- Department of Pathology, Academic Medic\\\al Centre, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, Netherlands
| | - Caterina Bendotti
- Laboratory of Molecular Neurobiology, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, Milan 20156, Italy.
| | - Giovanni Nardo
- Laboratory of Molecular Neurobiology, Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, Milan 20156, Italy.
| |
Collapse
|
36
|
Laiva AL, O'Brien FJ, Keogh MB. SDF-1α gene-activated collagen scaffold drives functional differentiation of human Schwann cells for wound healing applications. Biotechnol Bioeng 2020; 118:725-736. [PMID: 33064302 DOI: 10.1002/bit.27601] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 10/08/2020] [Accepted: 10/11/2020] [Indexed: 01/03/2023]
Abstract
Enhancing angiogenesis is the prime target of current biomaterial-based wound healing strategies. However, these approaches largely overlook the angiogenic role of the cells of the nervous system. Therefore, we explored the role of a collagen-chondroitin sulfate scaffold functionalized with a proangiogenic gene stromal-derived factor-1α (SDF-1α)-an SDF-1α gene-activated scaffold on the functional regulation of human Schwann cells (SCs). A preliminary 2D study was conducted by delivering plasmids encoding for the SDF-1α gene into a monolayer of SCs using polyethyleneimine-based nanoparticles. The delivery of the SDF-1α gene into the SCs enhanced the production of proangiogenic vascular endothelial growth factor (VEGF). Subsequently, we investigated the impact of SDF-1α gene-activated scaffold (3D) on the SCs for 2 weeks, using a gene-free scaffold as control. The transfection of the SCs within the gene-activated scaffold resulted in transient overexpression of SDF-1α transcripts and triggered the production of bioactive VEGF that enhanced endothelial angiogenesis. The overexpression of SDF-1α also caused transient activation of the transcription factor c-Jun and supported the differentiation of SCs towards a repair phenotype. This was characterized by elevated expression of neurotrophin receptor p75NGFR. During this developmental stage, the SCs also performed an extensive remodelling of the basement matrix (fibronectin, collagen IV, and laminin) to enrich their environment with the pro-neurogenic matrix protein laminin, revealing an enhanced pro-neurogenic behavior. Together, this study shows that SDF-1α gene-activated scaffold is a highly bioinstructive scaffold capable of enhancing proangiogenic regenerative response in human SCs for improved wound healing.
Collapse
Affiliation(s)
- Ashang L Laiva
- Department of Anatomy and Regenerative Medicine, Tissue Engineering Research Group, Royal College of Surgeons in Ireland, Dublin, Ireland.,Department of Biomedical Science, Royal College of Surgeons in Ireland, Bahrain, Adliya, Kingdom of Bahrain
| | - Fergal J O'Brien
- Department of Anatomy and Regenerative Medicine, Tissue Engineering Research Group, Royal College of Surgeons in Ireland, Dublin, Ireland.,Advanced Materials and Bioengineering Research Centre, Royal College of Surgeons in Ireland, Dublin, Ireland.,Advanced Materials and Bioengineering Research Centre, Trinity College Dublin, Dublin, Ireland.,Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Michael B Keogh
- Department of Anatomy and Regenerative Medicine, Tissue Engineering Research Group, Royal College of Surgeons in Ireland, Dublin, Ireland.,Department of Biomedical Science, Royal College of Surgeons in Ireland, Bahrain, Adliya, Kingdom of Bahrain
| |
Collapse
|
37
|
Stazi M, D’Este G, Mattarei A, Negro S, Lista F, Rigoni M, Megighian A, Montecucco C. An agonist of the CXCR4 receptor accelerates the recovery from the peripheral neuroparalysis induced by Taipan snake envenomation. PLoS Negl Trop Dis 2020; 14:e0008547. [PMID: 32898186 PMCID: PMC7537909 DOI: 10.1371/journal.pntd.0008547] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 10/06/2020] [Accepted: 06/19/2020] [Indexed: 01/15/2023] Open
Abstract
Envenomation by snakes is a major neglected human disease. Hospitalization and use of animal-derived antivenom are the primary therapeutic supports currently available. There is consensus that additional, not expensive, treatments that can be delivered even long after the snake bite are needed. We recently showed that the drug dubbed NUCC-390 shortens the time of recovery from the neuroparalysis caused by traumatic or toxic degeneration of peripheral motor neurons. These syndromes are characterized by the activation of a pro-regenerative molecular axis, consisting of the CXCR4 receptor expressed at the damaged site in neuronal axons and by the release of its ligand CXCL12α, produced by surrounding Schwann cells. This intercellular signaling axis promotes axonal growth and functional recovery from paralysis. NUCC-390 is an agonist of CXCR4 acting similarly to CXCL12α. Here, we have tested its efficacy in a murine model of neuroparalytic envenoming by a Papuan Taipan (Oxyuranus scutellatus) where a degeneration of the motor axon terminals caused by the presynaptic PLA2 toxin Taipoxin, contained in the venom, occurs. Using imaging of the neuromuscular junction and electrophysiological analysis, we found that NUCC-390 administration after injection of either the purified neuroparalytic Taipoxin or the whole Taipan venom, significantly accelerates the recovery from paralysis. These results indicate that NUCC-390, which is non-toxic in mice, should be considered for trials in humans to test its efficacy in accelerating the recovery from the peripheral neuroparalysis induced by Taipans. NUCC-390 should be tested as well in the envenomation by other snakes that cause neuroparalytic syndromes in humans. NUCC-390 could become an additional treatment, common to many snake envenomings, that can be delivered after the bite to reduce death by respiratory deficits and to shorten and improve functional recovery.
Collapse
Affiliation(s)
- Marco Stazi
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Giorgia D’Este
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Andrea Mattarei
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Samuele Negro
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Florigio Lista
- Department of Medical and Veterinary Research, the Ministry of Defense, Rome, Italy
| | - Michela Rigoni
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Aram Megighian
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Cesare Montecucco
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- CNR Institute of Neuroscience, Department of Biomedical Sciences, Padua, Italy
| |
Collapse
|
38
|
Bianchi ME, Mezzapelle R. The Chemokine Receptor CXCR4 in Cell Proliferation and Tissue Regeneration. Front Immunol 2020; 11:2109. [PMID: 32983169 PMCID: PMC7484992 DOI: 10.3389/fimmu.2020.02109] [Citation(s) in RCA: 190] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 08/04/2020] [Indexed: 12/13/2022] Open
Abstract
The CXCR4 receptor upon binding its ligands triggers multiple signaling pathways that orchestrate cell migration, hematopoiesis and cell homing, and retention in the bone marrow. However, CXCR4 also directly controls cell proliferation of non-hematopoietic cells. This review focuses on recent reports pointing to its pivotal role in tissue regeneration and stem cell activation, and discusses the connection to the known role of CXCR4 in promoting tumor growth. The mechanisms may be similar in all cases, since regeneration often recapitulates developmental processes, and cancer often exploits developmental pathways. Moreover, cell migration and cell proliferation appear to be downstream of the same signaling pathways. A deeper understanding of the complex signaling originating from CXCR4 is needed to exploit the opportunities to repair damaged organs safely and effectively.
Collapse
Affiliation(s)
- Marco E Bianchi
- Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | - Rosanna Mezzapelle
- Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
39
|
Ojeda J, Bermedo-García F, Pérez V, Mella J, Hanna P, Herzberg D, Tejero R, López-Manzaneda M, Tabares L, Henríquez JP. The Mouse Levator Auris Longus Muscle: An Amenable Model System to Study the Role of Postsynaptic Proteins to the Maintenance and Regeneration of the Neuromuscular Synapse. Front Cell Neurosci 2020; 14:225. [PMID: 32848618 PMCID: PMC7405910 DOI: 10.3389/fncel.2020.00225] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 06/26/2020] [Indexed: 12/16/2022] Open
Abstract
The neuromuscular junction (NMJ) is the peripheral synapse that controls the coordinated movement of many organisms. The NMJ is also an archetypical model to study synaptic morphology and function. As the NMJ is the primary target of neuromuscular diseases and traumatic injuries, the establishment of suitable models to study the contribution of specific postsynaptic muscle-derived proteins on NMJ maintenance and regeneration is a permanent need. Considering the unique experimental advantages of the levator auris longus (LAL) muscle, here we present a method allowing for efficient electroporation-mediated gene transfer and subsequent detailed studies of the morphology and function of the NMJ and muscle fibers. Also, we have standardized efficient facial nerve injury protocols to analyze LAL muscle NMJ degeneration and regeneration. Our results show that the expression of a control fluorescent protein does not alter either the muscle structural organization, the apposition of the pre- and post-synaptic domains, or the functional neurotransmission parameters of the LAL muscle NMJs; in turn, the overexpression of MuSK, a major regulator of postsynaptic assembly, induces the formation of ectopic acetylcholine receptor clusters. Our NMJ denervation experiments showed complete reinnervation of LAL muscle NMJs four weeks after facial nerve injury. Together, these experimental strategies in the LAL muscle constitute effective methods to combine protein expression with accurate analyses at the levels of structure, function, and regeneration of the NMJ.
Collapse
Affiliation(s)
- Jorge Ojeda
- Neuromuscular Studies Laboratory (NeSt Lab), Department of Cell Biology, Faculty of Biological Sciences, Center for Advanced Microscopy (CMA BioBio), Universidad de Concepción, Concepción, Chile.,Department of Medical Physiology and Biophysics, School of Medicine, Universidad de Sevilla, Sevilla, Spain.,Developmental Neurobiology Unit, Biomedical Sciences Research Laboratory, Basic Sciences Department, Faculty of Medicine, Universidad Católica de la Santísima Concepción, Concepción, Chile
| | - Francisca Bermedo-García
- Neuromuscular Studies Laboratory (NeSt Lab), Department of Cell Biology, Faculty of Biological Sciences, Center for Advanced Microscopy (CMA BioBio), Universidad de Concepción, Concepción, Chile
| | - Viviana Pérez
- Neuromuscular Studies Laboratory (NeSt Lab), Department of Cell Biology, Faculty of Biological Sciences, Center for Advanced Microscopy (CMA BioBio), Universidad de Concepción, Concepción, Chile
| | - Jessica Mella
- Neuromuscular Studies Laboratory (NeSt Lab), Department of Cell Biology, Faculty of Biological Sciences, Center for Advanced Microscopy (CMA BioBio), Universidad de Concepción, Concepción, Chile
| | - Patricia Hanna
- Neuromuscular Studies Laboratory (NeSt Lab), Department of Cell Biology, Faculty of Biological Sciences, Center for Advanced Microscopy (CMA BioBio), Universidad de Concepción, Concepción, Chile
| | - Daniel Herzberg
- Veterinary Sciences Clinic, Universidad de Concepción, Concepción, Chile
| | - Rocío Tejero
- Department of Medical Physiology and Biophysics, School of Medicine, Universidad de Sevilla, Sevilla, Spain
| | - Mario López-Manzaneda
- Department of Medical Physiology and Biophysics, School of Medicine, Universidad de Sevilla, Sevilla, Spain
| | - Lucia Tabares
- Department of Medical Physiology and Biophysics, School of Medicine, Universidad de Sevilla, Sevilla, Spain
| | - Juan Pablo Henríquez
- Neuromuscular Studies Laboratory (NeSt Lab), Department of Cell Biology, Faculty of Biological Sciences, Center for Advanced Microscopy (CMA BioBio), Universidad de Concepción, Concepción, Chile
| |
Collapse
|
40
|
Rigoni M, Negro S. Signals Orchestrating Peripheral Nerve Repair. Cells 2020; 9:E1768. [PMID: 32722089 PMCID: PMC7464993 DOI: 10.3390/cells9081768] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/19/2020] [Accepted: 07/20/2020] [Indexed: 12/22/2022] Open
Abstract
The peripheral nervous system has retained through evolution the capacity to repair and regenerate after assault from a variety of physical, chemical, or biological pathogens. Regeneration relies on the intrinsic abilities of peripheral neurons and on a permissive environment, and it is driven by an intense interplay among neurons, the glia, muscles, the basal lamina, and the immune system. Indeed, extrinsic signals from the milieu of the injury site superimpose on genetic and epigenetic mechanisms to modulate cell intrinsic programs. Here, we will review the main intrinsic and extrinsic mechanisms allowing severed peripheral axons to re-grow, and discuss some alarm mediators and pro-regenerative molecules and pathways involved in the process, highlighting the role of Schwann cells as central hubs coordinating multiple signals. A particular focus will be provided on regeneration at the neuromuscular junction, an ideal model system whose manipulation can contribute to the identification of crucial mediators of nerve re-growth. A brief overview on regeneration at sensory terminals is also included.
Collapse
Affiliation(s)
- Michela Rigoni
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy;
- Myology Center (Cir-Myo), University of Padua, 35129 Padua, Italy
| | - Samuele Negro
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy;
| |
Collapse
|
41
|
Panganiban AT, Blair RV, Hattler JB, Bohannon DG, Bonaldo MC, Schouest B, Maness NJ, Kim WK. A Zika virus primary isolate induces neuroinflammation, compromises the blood-brain barrier and upregulates CXCL12 in adult macaques. Brain Pathol 2020; 30:1017-1027. [PMID: 32585067 DOI: 10.1111/bpa.12873] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 03/29/2020] [Accepted: 07/21/2020] [Indexed: 12/23/2022] Open
Abstract
Zika virus (ZIKV) is a flavivirus that can cause neuropathogenesis in adults and fetal neurologic malformation following the infection of pregnant women. We used a nonhuman primate model, the Indian-origin Rhesus macaque (IRM), to gain insight into virus-associated hallmarks of ZIKV-induced adult neuropathology. We find that the virus causes prevalent acute and chronic neuroinflammation and chronic disruption of the blood-brain barrier (BBB) in adult animals. ZIKV infection resulted in specific short- and long-term augmented expression of the chemokine CXCL12 in the central nervous system (CNS)of adult IRMs. Moreover, CXCL12 expression persists long after the initial viral infection is apparently cleared. CXCL12 plays a key role both in regulating lymphocyte trafficking through the BBB to the CNS and in mediating repair of damaged neural tissue including remyelination. Understanding how CXCL12 expression is controlled will likely be of central importance in the definition of ZIKV-associated neuropathology in adults.
Collapse
Affiliation(s)
- Antonito T Panganiban
- Division of Microbiology, Tulane National Primate Research Center, 18703 Three Rivers Road, Covington, LA, 70433, USA
| | - Robert V Blair
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, 70433, USA
| | - Julian B Hattler
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, 23507, USA
| | - Diana G Bohannon
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, 23507, USA
| | - Myrna C Bonaldo
- Laboratory of Flavivirus Molecular Biology, Fiocruz, Avenida Brasil, 4365, Manguinhos, Rio de Janeiro, RJ, 21040-360, Brazil
| | - Blake Schouest
- Division of Microbiology, Tulane National Primate Research Center, 18703 Three Rivers Road, Covington, LA, 70433, USA
| | - Nicholas J Maness
- Division of Microbiology, Tulane National Primate Research Center, 18703 Three Rivers Road, Covington, LA, 70433, USA
| | - Woong-Ki Kim
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, 23507, USA
| |
Collapse
|
42
|
Singh T, Robles D, Vazquez M. Neuronal substrates alter the migratory responses of nonmyelinating Schwann cells to controlled brain‐derived neurotrophic factor gradients. J Tissue Eng Regen Med 2020; 14:609-621. [DOI: 10.1002/term.3025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 01/16/2020] [Accepted: 02/02/2020] [Indexed: 12/19/2022]
Affiliation(s)
- Tanya Singh
- Department of Biomedical EngineeringCity College of New York New York NY USA
| | - Denise Robles
- Department of Biomedical EngineeringRutgers University, The State University of New Jersey New Brunswick NJ USA
| | - Maribel Vazquez
- Department of Biomedical EngineeringRutgers University, The State University of New Jersey New Brunswick NJ USA
| |
Collapse
|
43
|
Li L, Yokoyama H, Kaburagi H, Hirai T, Tsuji K, Enomoto M, Wakabayashi Y, Okawa A. Remnant neuromuscular junctions in denervated muscles contribute to functional recovery in delayed peripheral nerve repair. Neural Regen Res 2020; 15:731-738. [PMID: 31638098 PMCID: PMC6975147 DOI: 10.4103/1673-5374.266925] [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: 12/18/2022] Open
Abstract
Schwann cell proliferation in peripheral nerve injury (PNI) enhances axonal regeneration compared to central nerve injury. However, even in PNI, long-term nerve damage without repair induces degeneration of neuromuscular junctions (NMJs), and muscle atrophy results in irreversible dysfunction. The peripheral regeneration of motor axons depends on the duration of skeletal muscle denervation. To overcome this difficulty in nerve regeneration, detailed mechanisms should be determined for not only Schwann cells but also NMJ degeneration after PNI and regeneration after nerve repair. Here, we examined motor axon denervation in the tibialis anterior muscle after peroneal nerve transection in thy1-YFP mice and regeneration with nerve reconstruction using allografts. The number of NMJs in the tibialis anterior muscle was maintained up to 4 weeks and then decreased at 6 weeks after injury. In contrast, the number of Schwann cells showed a stepwise decline and then reached a plateau at 6 weeks after injury. For regeneration, we reconstructed the degenerated nerve with an allograft at 4 and 6 weeks after injury, and evaluated functional and histological outcomes for 10 to 12 weeks after grafting. A higher number of pretzel-shaped NMJs in the tibialis anterior muscle and better functional recovery were observed in mice with a 4-week delay in surgery than in those with a 6-week delay. Nerve repair within 4 weeks after PNI is necessary for successful recovery in mice. Prevention of synaptic acetylcholine receptor degeneration may play a key role in peripheral nerve regeneration. All animal experiments were approved by the Institutional Animal Care and Use Committee of Tokyo Medical and Dental University on 5 July 2017, 30 March 2018, and 15 May 2019 (A2017-311C, A2018-297A, and A2019-248A), respectively.
Collapse
Affiliation(s)
- Leyang Li
- Department of Orthopedic and Spinal Surgery, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hiroyuki Yokoyama
- Department of Orthopedic and Spinal Surgery, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hidetoshi Kaburagi
- Department of Orthopedic and Spinal Surgery, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takashi Hirai
- Department of Orthopedic and Spinal Surgery, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kunikazu Tsuji
- Department of Cartilage Regeneration, Tokyo Medical and Dental University, Tokyo, Japan
| | - Mitsuhiro Enomoto
- Department of Orthopedic and Spinal Surgery, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yoshiaki Wakabayashi
- Department of Orthopedic and Spinal Surgery, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| | - Atsushi Okawa
- Department of Orthopedic and Spinal Surgery, Graduate School, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
44
|
Zanetti G, Negro S, Megighian A, Mattarei A, Lista F, Fillo S, Rigoni M, Pirazzini M, Montecucco C. A CXCR4 receptor agonist strongly stimulates axonal regeneration after damage. Ann Clin Transl Neurol 2019; 6:2395-2402. [PMID: 31725979 PMCID: PMC6917312 DOI: 10.1002/acn3.50926] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 09/30/2019] [Accepted: 10/02/2019] [Indexed: 12/21/2022] Open
Abstract
Objective To test whether the signaling axis CXCL12α‐CXCR4 is activated upon crush/cut of the sciatic nerve and to test the activity of NUCC‐390, a new CXCR4 agonist, in promoting nerve recovery from damage. Methods The sciatic nerve was either crushed or cut. Expression and localization of CXCL12α and CXCR4 were evaluated by imaging with specific antibodies. Their functional involvement in nerve regeneration was determined by antibody‐neutralization of CXCL12α, and by the CXCR4 specific antagonist AMD3100, using as quantitative read‐out the compound muscle action potential (CMAP). NUCC‐390 activity on nerve regeneration was determined by imaging and CMAP recordings. Results CXCR4 is expressed at the injury site within the axonal compartment, whilst its ligand CXCL12α is expressed in Schwann cells. The CXCL12α‐CXCR4 axis is involved in the recovery of neurotransmission of the injured nerve. More importantly, the small molecule NUCC‐390 is a strong promoter of the functional and anatomical recovery of the nerve, by acting very similarly to CXCL12α. This pharmacological action is due to the capability of NUCC‐390 to foster elongation of motor neuron axons both in vitro and in vivo. Interpretation Imaging and electrophysiological data provide novel and compelling evidence that the CXCL12α‐CXCR4 axis is involved in sciatic nerve repair after crush/cut. This makes NUCC‐390 a strong candidate molecule to stimulate nerve repair by promoting axonal elongation. We propose this molecule to be tested in other models of neuronal damage, to lay the basis for clinical trials on the efficacy of NUCC‐390 in peripheral nerve repair in humans.
Collapse
Affiliation(s)
- Giulia Zanetti
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Samuele Negro
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Aram Megighian
- Department of Biomedical Sciences, University of Padua, Padua, Italy.,Padua Neuroscience Center, University of Padua, Padua, Italy
| | - Andrea Mattarei
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | | | - Silvia Fillo
- Scientific Department, Army Medical Center, Roma, Italy
| | - Michela Rigoni
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Marco Pirazzini
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Cesare Montecucco
- Department of Biomedical Sciences, University of Padua, Padua, Italy.,CNR Institute of Neuroscience, Padua, Italy
| |
Collapse
|
45
|
Negro S, Zanetti G, Mattarei A, Valentini A, Megighian A, Tombesi G, Zugno A, Dianin V, Pirazzini M, Fillo S, Lista F, Rigoni M, Montecucco C. An Agonist of the CXCR4 Receptor Strongly Promotes Regeneration of Degenerated Motor Axon Terminals. Cells 2019; 8:E1183. [PMID: 31575088 PMCID: PMC6829515 DOI: 10.3390/cells8101183] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 09/24/2019] [Accepted: 09/27/2019] [Indexed: 12/19/2022] Open
Abstract
The activation of the G-protein coupled receptor CXCR4 by its ligand CXCL12α is involved in a large variety of physiological and pathological processes, including the growth of B cells precursors and of motor axons, autoimmune diseases, stem cell migration, inflammation, and several neurodegenerative conditions. Recently, we demonstrated that CXCL12α potently stimulates the functional recovery of damaged neuromuscular junctions via interaction with CXCR4. This result prompted us to test the neuroregeneration activity of small molecules acting as CXCR4 agonists, endowed with better pharmacokinetics with respect to the natural ligand. We focused on NUCC-390, recently shown to activate CXCR4 in a cellular system. We designed a novel and convenient chemical synthesis of NUCC-390, which is reported here. NUCC-390 was tested for its capability to induce the regeneration of motor axon terminals completely degenerated by the presynaptic neurotoxin α-Latrotoxin. NUCC-390 was found to strongly promote the functional recovery of the neuromuscular junction, as assayed by electrophysiology and imaging. This action is CXCR4 dependent, as it is completely prevented by AMD3100, a well-characterized CXCR4 antagonist. These data make NUCC-390 a strong candidate to be tested in human therapy to promote nerve recovery of function after different forms of neurodegeneration.
Collapse
Affiliation(s)
- Samuele Negro
- Department of Biomedical Sciences, University of Padua, Padua 35131, Italy.
| | - Giulia Zanetti
- Department of Biomedical Sciences, University of Padua, Padua 35131, Italy.
| | - Andrea Mattarei
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua 35131, Italy.
| | - Alice Valentini
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua 35131, Italy.
| | - Aram Megighian
- Department of Biomedical Sciences, University of Padua, Padua 35131, Italy.
- Padua Neuroscience Institute, Padua 35131, Italy.
| | - Giulia Tombesi
- Department of Biology, University of Padua, Padua 35131, Italy.
| | - Alessandro Zugno
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua 35131, Italy.
| | - Valentina Dianin
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua 35131, Italy.
| | - Marco Pirazzini
- Department of Biomedical Sciences, University of Padua, Padua 35131, Italy.
| | - Silvia Fillo
- Center of Medical and Veterinary Research of the Ministry of Defence, Rome 00184, Italy.
| | - Florigio Lista
- Center of Medical and Veterinary Research of the Ministry of Defence, Rome 00184, Italy.
| | - Michela Rigoni
- Department of Biomedical Sciences, University of Padua, Padua 35131, Italy.
| | - Cesare Montecucco
- Department of Biomedical Sciences, University of Padua, Padua 35131, Italy.
- CNR Institute of Neuroscience, Padua 35131, Italy.
| |
Collapse
|
46
|
Piovesana R, Faroni A, Magnaghi V, Reid AJ, Tata AM. M2 receptors activation modulates cell growth, migration and differentiation of rat Schwann-like adipose-derived stem cells. Cell Death Discov 2019; 5:92. [PMID: 31069117 PMCID: PMC6499790 DOI: 10.1038/s41420-019-0174-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 02/13/2019] [Accepted: 03/04/2019] [Indexed: 12/12/2022] Open
Abstract
Schwann cells (SCs) play a central role in peripheral nervous system physiology and in the response to axon injury. The ability of SCs to proliferate, secrete growth factors, modulate immune response, migrate and re-myelinate regenerating axons has been largely documented. However, there are several restrictions hindering their clinical application, such as the difficulty in collection and a slow in vitro expansion. Adipose-derived stem cells (ASCs) present good properties for peripheral nerve regenerative medicine. When exposed to specific growth factors in vitro, they can acquire a SC-like phenotype (dASCs) expressing key SCs markers and assuming spindle-shaped morphology. Nevertheless, the differentiated phenotype is unstable and several strategies, including pharmacological stimulation, are being studied to improve differentiation outcomes. Cholinergic receptors are potential pharmacological targets expressed in glial cells. Our previous work demonstrated that muscarinic cholinergic receptors, in particular M2 subtype, are present in SCs and are able to modulate several physiological processes. In the present work, muscarinic receptors expression was characterised and the effects mediated by M2 muscarinic receptor were evaluated in rat dASCs. M2 receptor activation, by the preferred agonist arecaidine propargyl ester (APE), caused a reversible arrest of dASCs cell growth, supported by the downregulation of proteins involved in the maintenance of cell proliferation and upregulation of proteins involved in the differentiation (i.e., c-Jun and Egr-2), without affecting cell survival. Moreover, M2 receptor activation in dASCs enhances a pronounced spindle-shaped morphology, supported by Egr2 upregulation, and inhibits cell migration. Our data clearly demonstrate that rat dASCs express functional muscarinic receptors, in particular M2 subtype, which is able to modulate their physiological and morphological processes, as well as SCs differentiation. These novel findings could open new opportunities for the development of combined cell and pharmacological therapies for peripheral nerve regeneration, harnessing the potential of dASCs and M2 receptors.
Collapse
Affiliation(s)
- Roberta Piovesana
- 1Department of Biology and Biotechnologies "Charles Darwin", "Sapienza" University of Rome, Rome, 00185 Italy.,2Blond McIndoe Laboratories, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT UK
| | - Alessandro Faroni
- 2Blond McIndoe Laboratories, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT UK
| | - Valerio Magnaghi
- 3Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, 20133 Italy
| | - Adam J Reid
- 2Blond McIndoe Laboratories, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, M13 9PT UK.,4Department of Plastic Surgery & Burns, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Ada Maria Tata
- 1Department of Biology and Biotechnologies "Charles Darwin", "Sapienza" University of Rome, Rome, 00185 Italy.,5Research Center of Neurobiology "Daniel Bovet", "Sapienza" University of Rome, Rome, 00185 Italy
| |
Collapse
|
47
|
Neurobiology and therapeutic applications of neurotoxins targeting transmitter release. Pharmacol Ther 2019; 193:135-155. [DOI: 10.1016/j.pharmthera.2018.08.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
48
|
Santosa KB, Keane AM, Jablonka-Shariff A, Vannucci B, Snyder-Warwick AK. Clinical relevance of terminal Schwann cells: An overlooked component of the neuromuscular junction. J Neurosci Res 2018; 96:1125-1135. [PMID: 29536564 PMCID: PMC6292684 DOI: 10.1002/jnr.24231] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 12/30/2017] [Accepted: 02/09/2018] [Indexed: 12/13/2022]
Abstract
The terminal Schwann cell (tSC), a type of nonmyelinating Schwann cell, is a significant yet relatively understudied component of the neuromuscular junction. In addition to reviewing the role tSCs play on formation, maintenance, and remodeling of the synapse, we review studies that implicate tSCs in neuromuscular diseases including spinal muscular atrophy, Miller-Fisher syndrome, and amyotrophic lateral sclerosis, among others. We also discuss the importance of these cells on degeneration and regeneration after nerve injury. Knowledge of tSC biology may improve our understanding of disease pathogenesis and help us identify new and innovative therapeutic strategies for the many patients who suffer from neuromuscular disorders and nerve injuries.
Collapse
Affiliation(s)
- Katherine B. Santosa
- Postdoctoral Research Fellow, Division of Plastic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO
| | - Alexandra M. Keane
- Medical Student, Washington University School of Medicine, St. Louis, MO
| | - Albina Jablonka-Shariff
- Research Scientist, Division of Plastic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO
| | - Bianca Vannucci
- Medical Student, Washington University School of Medicine, St. Louis, MO
| | - Alison K. Snyder-Warwick
- Assistant Professor, Division of Plastic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
49
|
Zanetti G, Duregotti E, Locatelli CA, Giampreti A, Lonati D, Rossetto O, Pirazzini M. Variability in venom composition of European viper subspecies limits the cross-effectiveness of antivenoms. Sci Rep 2018; 8:9818. [PMID: 29959358 PMCID: PMC6026201 DOI: 10.1038/s41598-018-28135-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 06/14/2018] [Indexed: 12/17/2022] Open
Abstract
Medically relevant cases of snakebite in Europe are predominately caused by European vipers of the genus Vipera. Systemic envenoming by European vipers can cause severe pathology in humans and different clinical manifestations are associated with different members of this genus. The most representative vipers in Europe are V. aspis and V. berus and neurological symptoms have been reported in humans envenomed by the former but not by the latter species. In this study we determined the toxicological profile of V. aspis and V. berus venoms in vivo in mice and we tested the effectiveness of two antivenoms, commonly used as antidotes, in counteracting the specific activities of the two venoms. We found that V. aspis, but not V. berus, is neurotoxic and that this effect is due to the degeneration of peripheral nerve terminals at the NMJ and is not neutralized by the two tested antisera. Differently, V. berus causes a haemorrhagic effect, which is efficiently contrasted by the same antivenoms. These results indicate that the effectiveness of different antisera is strongly influenced by the variable composition of the venoms and reinforce the arguments supporting the use polyvalent antivenoms.
Collapse
Affiliation(s)
- Giulia Zanetti
- University of Padova, Department of Biomedical Sciences, Padova, 35131, Italy
| | - Elisa Duregotti
- University of Padova, Department of Biomedical Sciences, Padova, 35131, Italy
- King's College London, Department of Cardiology, James Black Centre, London, SE5 9NU, United Kingdom
| | - Carlo Alessandro Locatelli
- Istituti Clinici Scientifici Maugeri, IRCCS Maugeri Hospital and University of Pavia, Poison Control Centre and National Toxicology Information Centre - Toxicology Unit, Pavia, 27100, Italy
| | - Andrea Giampreti
- Istituti Clinici Scientifici Maugeri, IRCCS Maugeri Hospital and University of Pavia, Poison Control Centre and National Toxicology Information Centre - Toxicology Unit, Pavia, 27100, Italy
| | - Davide Lonati
- Istituti Clinici Scientifici Maugeri, IRCCS Maugeri Hospital and University of Pavia, Poison Control Centre and National Toxicology Information Centre - Toxicology Unit, Pavia, 27100, Italy
| | - Ornella Rossetto
- University of Padova, Department of Biomedical Sciences, Padova, 35131, Italy
| | - Marco Pirazzini
- University of Padova, Department of Biomedical Sciences, Padova, 35131, Italy.
| |
Collapse
|
50
|
Gao D, Sun H, Zhu J, Tang Y, Li S. CXCL12 induces migration of Schwann cells via p38 MAPK and autocrine of CXCL12 by the CXCR4 receptor. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2018; 11:3119-3125. [PMID: 31938440 PMCID: PMC6958085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 03/28/2018] [Indexed: 06/10/2023]
Abstract
Schwann cells (SCs) play a crucially supportive role in repair of injured peripheral nerve system (PNS). CXCL12 plays a significant role in migration of stem cells and embryonic developmental cells and CXCL12 is strongly chemotactic for a variety of cells. Our study was designed to determine the role of CXCL12 in Schwann cell proliferation and migration. Our study demonstrated that CXCL12 had no effect on Schwann cell proliferation while significantly promoting Schwann cell migration. CXCL12-induced Schwann cell migration was significantly attenuated by inhibition of its receptor CXCR4 and p38 MAPK through co-treatment with AMD3100 and SB203580, separately. Besides, Western blot, QRT-PCR, and ELISA indicated that treatment with CXCL12 enhanced expression of CXCL12 by Schwann cells. In conclusion, CXCL12-enhanced SCs migration is mediated by secreting CXCL12 and p38 MAPK via receptor CXCR4, suggesting that CXCL12 has potential application value for PNS regeneration and could serve as a new therapeutic strategy in peripheral nerve diseases.
Collapse
Affiliation(s)
- Dekun Gao
- Department of Neurosurgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai, China
| | - Hui Sun
- Department of Neurosurgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai, China
| | - Jin Zhu
- Department of Neurosurgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai, China
| | - Yinda Tang
- Department of Neurosurgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai, China
| | - Shiting Li
- Department of Neurosurgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai, China
| |
Collapse
|