1
|
Ta D, Anthony A, Sliow A, Wan B, Zhang L, Higgins M, Lam L, Mahns D, Gargiulo G, Breen P, Mawad D, Ruprai H, Myers S, Laurenti D, Lauto A. Effect of Brief Electrical Stimulation on Cell Biomechanics in Hereditary Sensory Neuropathy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2408941. [PMID: 39901509 DOI: 10.1002/smll.202408941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 01/15/2025] [Indexed: 02/05/2025]
Abstract
SH-SY5Y neuroblastoma cells are widely used to model neurodegenerative disorders like Alzheimer's, Parkinson's, Huntington's, and Hereditary Sensory Neuropathy type 1A (HSN-1A), a peripheral nerve condition causing axon degeneration and sensory loss. A cell model of HSN-1A is developed by overexpressing wild-type and mutant SPTLC1 genes (C133W, C133Y, V144D). Cells are cultured on plastic and gold substrates, with brief electrical stimulation applied to the gold-grown cells. Atomic force microscopy (AFM) is used to measure Young's modulus, indentation, and energy dissipation. Finite Element Method and non-linear modeling validate the results. In the absence of stimulation, mutant cells show lower stiffness compared to non-transfected cells, indicating a direct biomechanical impact of the mutations. Brief electrical stimulation significantly increases the stiffness of mutant cells, particularly in C133W (99%), C133Y (100%), and V144D (111%) variants, despite the mutations. Energy dissipation of stimulated V144D cells decreases to levels comparable to untreated non-transfected cells. The simulations support the AFM measurements, demonstrating that brief electrical stimulation can partially reverse the biomechanical effects of gene mutations.
Collapse
Affiliation(s)
- Daniel Ta
- School of Science, Western Sydney University, Penrith, New South Wales, 2751, Australia
| | - Anu Anthony
- School of Science, Western Sydney University, Penrith, New South Wales, 2751, Australia
| | - Ashour Sliow
- School of Science, Western Sydney University, Penrith, New South Wales, 2751, Australia
| | - Boyang Wan
- School of Aerospace, Mechanical and Mechatronic Engineering, The University of Sydney, Sydney, New South Wales, 2006, Australia
| | - Leo Zhang
- Centre for Advanced Manufacturing Technology, School of Engineering, Design and Built Environment, Western Sydney University, Penrith, New South Wales, 2751, Australia
| | - Michael Higgins
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute AIIM Facility Innovation Campus, University of Wollongong, Wollongong, New South Wales, 2522, Australia
| | - Lisa Lam
- School of Science, Western Sydney University, Penrith, New South Wales, 2751, Australia
| | - David Mahns
- School of Medicine, Western Sydney University, Penrith, New South Wales, 2751, Australia
| | - Gaetano Gargiulo
- Biomedical Engineering & Neuroscience Research Group, The MARCS Institute, Western Sydney University, Penrith, NSW, 2751, Australia
| | - Paul Breen
- Biomedical Engineering & Neuroscience Research Group, The MARCS Institute, Western Sydney University, Penrith, NSW, 2751, Australia
| | - Damia Mawad
- The School of Materials Science and Engineering, UNSW Sydney, Sydney, New South Wales, 2052, Australia
| | - Herleen Ruprai
- School of Science, Western Sydney University, Penrith, New South Wales, 2751, Australia
| | - Simon Myers
- School of Science, Western Sydney University, Penrith, New South Wales, 2751, Australia
| | - Daunia Laurenti
- School of Medicine, Western Sydney University, Penrith, New South Wales, 2751, Australia
- NICM Health Research Institute, Western Sydney University, Penrith, New South Wales, 2751, Australia
| | - Antonio Lauto
- School of Science, Western Sydney University, Penrith, New South Wales, 2751, Australia
| |
Collapse
|
2
|
Maulana TI, Wevers NR, Kristoforus T, Chandler M, Lanz HL, Joore J, Vulto P, Villenave R, Kustermann S, Loskill P, Bircsak KM. Opportunities for Microphysiological Systems in Toxicity Testing of New Drug Modalities. Annu Rev Pharmacol Toxicol 2025; 65:47-69. [PMID: 39227343 DOI: 10.1146/annurev-pharmtox-061724-080621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
New drug modalities offer life-saving benefits for patients through access to previously undruggable targets. Yet these modalities pose a challenge for the pharmaceutical industry, as side effects are complex, unpredictable, and often uniquely human. With animal studies having limited predictive value due to translatability challenges, the pharmaceutical industry seeks out new approach methodologies. Microphysiological systems (MPS) offer important features that enable complex toxicological processes to be modeled in vitro such as (a) an adjustable complexity of cellular components, including immune components; (b) a modifiable tissue architecture; (c) integration and monitoring of dynamic mechanisms; and (d) a multiorgan connection. Here we review MPS studies in the context of four clinical adverse events triggered by new drug modalities: peripheral neuropathy, thrombocytopenia, immune-mediated hepatotoxicity, and cytokine release syndrome. We conclude that while the use of MPS for testing new drug modality-induced toxicities is still in its infancy, we see strong potential going forward.
Collapse
Affiliation(s)
- Tengku Ibrahim Maulana
- Department for Microphysiological Systems, Institute of Biomedical Engineering, Faculty of Medicine, Eberhard Karls University Tübingen, Tübingen, Germany
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | | | - Theodora Kristoforus
- Department for Microphysiological Systems, Institute of Biomedical Engineering, Faculty of Medicine, Eberhard Karls University Tübingen, Tübingen, Germany
| | | | | | - Jos Joore
- MIMETAS BV, Oegstgeest, The Netherlands
| | | | - Remi Villenave
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Stefan Kustermann
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Peter Loskill
- Department for Microphysiological Systems, Institute of Biomedical Engineering, Faculty of Medicine, Eberhard Karls University Tübingen, Tübingen, Germany
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | | |
Collapse
|
3
|
Crugeiras J, Calls A, Contreras E, Alemany M, Navarro X, Yuste VJ, Casanovas O, Udina E, Bruna J. Oxygen matters: Unraveling the role of oxygen in the neuronal response to cisplatin. J Peripher Nerv Syst 2024; 29:528-536. [PMID: 39329299 PMCID: PMC11625991 DOI: 10.1111/jns.12659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 08/31/2024] [Accepted: 09/11/2024] [Indexed: 09/28/2024]
Abstract
BACKGROUND AND AIMS Cell culture is a fundamental experimental tool for understanding cell physiology. However, translating these findings to in vivo settings has proven challenging. Replicating donor tissue conditions, including oxygen levels, is crucial for achieving meaningful results. Nevertheless, oxygen culture conditions are often overlooked, particularly in the context of chemotherapy-induced neurotoxicity. METHODS In this study, we investigated the role of oxygen levels in primary neuronal cultures by comparing neuronal performance under cisplatin exposure (1 μg/mL) in supraphysiological normoxia (representing atmospheric conditions in a standard incubator; 18.5% O2) and physioxia (representing physiologic oxygen conditions in nervous tissue; 5% O2). Experiments were also conducted to assess survival, neurite development, senescence marker expression, and proinflammatory cytokine secretion. RESULTS Under control conditions, both oxygen concentration conditions exhibited similar behaviors. However, after cisplatin administration, sensory neurons cultured under supraphysiological normoxic conditions show higher mortality, exhibit an evolutionarily proinflammatory cytokine profile over time, and activate apoptotic-regulated neuron death markers. In contrast, under physiological conditions, neurons treated with cisplatin exhibited senescence marker expression and an attenuated inflammatory secretome. INTERPRETATION These results underscore the critical role of oxygen in neuronal culture, particularly in studying compounds where neuronal damage is mechanistically linked to oxidative stress. Even at identical doses of evaluated neurotoxic drugs, distinct cellular phenotypic fates can emerge, impacting translatability to the in vivo setting.
Collapse
Affiliation(s)
- Jose Crugeiras
- Department of Cell Biology, Physiology, and ImmunologyInstitute of Neuroscience, Autonomous University of BarcelonaBellaterraSpain
- Biomedical Research Center Network on Neurodegenerative Diseases (CIBERNED)BellaterraSpain
| | - Aina Calls
- Department of Cell Biology, Physiology, and ImmunologyInstitute of Neuroscience, Autonomous University of BarcelonaBellaterraSpain
- Biomedical Research Center Network on Neurodegenerative Diseases (CIBERNED)BellaterraSpain
| | - Estefanía Contreras
- Department of Cell Biology, Physiology, and ImmunologyInstitute of Neuroscience, Autonomous University of BarcelonaBellaterraSpain
- Biomedical Research Center Network on Neurodegenerative Diseases (CIBERNED)BellaterraSpain
| | - Montse Alemany
- Unit of Neuro‐Oncology, Hospital Universitari de BellvitgeBellvitge Institute for Biomedical Research (IDIBELL)BarcelonaSpain
| | - Xavier Navarro
- Department of Cell Biology, Physiology, and ImmunologyInstitute of Neuroscience, Autonomous University of BarcelonaBellaterraSpain
- Biomedical Research Center Network on Neurodegenerative Diseases (CIBERNED)BellaterraSpain
| | - Victor J. Yuste
- Department of BiochemistryInstitute of Neuroscience, Autonomous University of BarcelonaBellaterraSpain
| | - Oriol Casanovas
- Tumor Angiogenesis Group, ProCURE ProgramCatalan Institute of Oncology, OncoBell Program, IDIBELLBarcelonaSpain
| | - Esther Udina
- Department of Cell Biology, Physiology, and ImmunologyInstitute of Neuroscience, Autonomous University of BarcelonaBellaterraSpain
- Biomedical Research Center Network on Neurodegenerative Diseases (CIBERNED)BellaterraSpain
| | - Jordi Bruna
- Biomedical Research Center Network on Neurodegenerative Diseases (CIBERNED)BellaterraSpain
- Unit of Neuro‐Oncology, Hospital Universitari de BellvitgeBellvitge Institute for Biomedical Research (IDIBELL)BarcelonaSpain
| |
Collapse
|
4
|
Tarasiuk O, Invernizzi C, Alberti P. In vitro neurotoxicity testing: lessons from chemotherapy-induced peripheral neurotoxicity. Expert Opin Drug Metab Toxicol 2024; 20:1037-1052. [PMID: 39246127 DOI: 10.1080/17425255.2024.2401584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 08/30/2024] [Accepted: 09/03/2024] [Indexed: 09/10/2024]
Abstract
INTRODUCTION Chemotherapy induced peripheral neurotoxicity (CIPN) is a long-lasting, or even permanent, late toxicity caused by largely used anticancer drugs. CIPN affects a growing population of cancer survivors and diminishes their quality of life since there is no curative/preventive treatment. Among several reasons for this unmet clinical need, there is an incomplete knowledge on mechanisms leading to CIPN. Therefore, bench side research is still greatly needed: in vitro studies are pivotal to both evaluate neurotoxicity mechanisms and potential neuroprotection strategies. AREAS COVERED Advantages and disadvantages of in vitro approaches are addressed with respect to their applicability to the CIPN field. Different cell cultures and techniques to assess neurotoxicity/neuroprotection are described. PubMed search-string: (chemotherapy-induced) AND (((neuropathy) OR neurotoxicity) OR neuropathic pain) AND (in vitro) AND (((((model) OR SH-SY5Y) OR PC12) OR iPSC) OR DRG neurons); (chemotherapy-induced) AND (((neuropathy) OR neurotoxicity) OR neuropathic pain) AND (model) AND (((neurite elongation) OR cell viability) OR morphology). No articles published before 1990 were selected. EXPERT OPINION CIPN is an ideal experimental setting to test axonal damage and, in general, peripheral nervous system mechanisms of disease and neuroprotection. Therefore, starting from robust preclinical data in this field, potentially, relevant biological rationale can be transferred to other human spontaneous diseases of the peripheral nervous system.
Collapse
Affiliation(s)
- Olga Tarasiuk
- Experimental Neurology Unit, School of Medicine and Surgery, Monza, Italy
- NeuroMI (Milan Center for Neuroscience), Milan, Italy
| | - Chiara Invernizzi
- Experimental Neurology Unit, School of Medicine and Surgery, Monza, Italy
- Neuroscience, School of Medicine and Surgery, Monza, Italy
| | - Paola Alberti
- Experimental Neurology Unit, School of Medicine and Surgery, Monza, Italy
- NeuroMI (Milan Center for Neuroscience), Milan, Italy
- Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| |
Collapse
|
5
|
Smulders PS, Heikamp K, Hermanides J, Hollmann MW, ten Hoope W, Weber NC. Chemotherapy-induced peripheral neuropathy models constructed from human induced pluripotent stem cells and directly converted cells: a systematic review. Pain 2024; 165:1914-1925. [PMID: 38381959 PMCID: PMC11331829 DOI: 10.1097/j.pain.0000000000003193] [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: 08/14/2023] [Revised: 01/04/2024] [Accepted: 01/04/2024] [Indexed: 02/23/2024]
Abstract
ABSTRACT Developments in human cellular reprogramming now allow for the generation of human neurons for in vitro disease modelling. This technique has since been used for chemotherapy-induced peripheral neuropathy (CIPN) research, resulting in the description of numerous CIPN models constructed from human neurons. This systematic review provides a critical analysis of available models and their methodological considerations (ie, used cell type and source, CIPN induction strategy, and validation method) for prospective researchers aiming to incorporate human in vitro models of CIPN in their research. The search strategy was developed with assistance from a clinical librarian and conducted in MEDLINE (PubMed) and Embase (Ovid) on September 26, 2023. Twenty-six peer-reviewed experimental studies presenting original data about human reprogrammed nonmotor neuron cell culture systems and relevant market available chemotherapeutics drugs were included. Virtually, all recent reports modeled CIPN using nociceptive dorsal root ganglion neurons. Drugs known to cause the highest incidence of CIPN were most used. Furthermore, treatment effects were almost exclusively validated by the acute effects of chemotherapeutics on neurite dynamics and cytotoxicity parameters, enabling the extrapolation of the half-maximal inhibitory concentration for the 4 most used chemotherapeutics. Overall, substantial heterogeneity was observed in the way studies applied chemotherapy and reported their findings. We therefore propose 6 suggestions to improve the clinical relevance and appropriateness of human cellular reprogramming-derived CIPN models.
Collapse
Affiliation(s)
- Pascal S.H. Smulders
- Department of Anesthesiology, Amsterdam UMC location University of Amsterdam, Laboratory for Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), Amsterdam, the Netherlands
| | - Kim Heikamp
- Department of Anesthesiology, Amsterdam UMC location University of Amsterdam, Laboratory for Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), Amsterdam, the Netherlands
| | - Jeroen Hermanides
- Department of Anesthesiology, Amsterdam UMC location University of Amsterdam, Laboratory for Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), Amsterdam, the Netherlands
| | - Markus W. Hollmann
- Department of Anesthesiology, Amsterdam UMC location University of Amsterdam, Laboratory for Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), Amsterdam, the Netherlands
| | - Werner ten Hoope
- Department of Anesthesiology, Amsterdam UMC location University of Amsterdam, Laboratory for Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), Amsterdam, the Netherlands
- Department of Anesthesiology, Rijnstate Hospital, Arnhem, the Netherlands
| | - Nina C. Weber
- Department of Anesthesiology, Amsterdam UMC location University of Amsterdam, Laboratory for Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), Amsterdam, the Netherlands
| |
Collapse
|
6
|
Utkina-Sosunova I, Chiorazzi A, de Planell-Saguer M, Li H, Meregalli C, Pozzi E, Carozzi VA, Canta A, Monza L, Alberti P, Fumagalli G, Karan C, Moayedi Y, Przedborski S, Cavaletti G, Lotti F. Molsidomine provides neuroprotection against vincristine-induced peripheral neurotoxicity through soluble guanylyl cyclase activation. Sci Rep 2024; 14:19341. [PMID: 39164364 PMCID: PMC11336221 DOI: 10.1038/s41598-024-70294-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 08/14/2024] [Indexed: 08/22/2024] Open
Abstract
Peripheral neurotoxicity is a dose-limiting adverse reaction of primary frontline chemotherapeutic agents, including vincristine. Neuropathy can be so disabling that patients drop out of potentially curative therapy, negatively impacting cancer prognosis. The hallmark of vincristine neurotoxicity is axonopathy, yet its underpinning mechanisms remain uncertain. We developed a comprehensive drug discovery platform to identify neuroprotective agents against vincristine-induced neurotoxicity. Among the hits identified, SIN-1-an active metabolite of molsidomine-prevents vincristine-induced axonopathy in both motor and sensory neurons without compromising vincristine anticancer efficacy. Mechanistically, we found that SIN-1's neuroprotective effect is mediated by activating soluble guanylyl cyclase. We modeled vincristine-induced peripheral neurotoxicity in rats to determine molsidomine therapeutic potential in vivo. Vincristine administration induced severe nerve damage and mechanical hypersensitivity that were attenuated by concomitant treatment with molsidomine. This study provides evidence of the neuroprotective properties of molsidomine and warrants further investigations of this drug as a therapy for vincristine-induced peripheral neurotoxicity.
Collapse
Affiliation(s)
- Irina Utkina-Sosunova
- Center for Motor Neuron Biology and Disease, Columbia University, New York, NY, 10032, USA
- Department of Pathology & Cell Biology, Columbia University, New York, NY, 10032, USA
- Department of Neurology, Columbia University, New York, NY, 10032, USA
| | - Alessia Chiorazzi
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Mariangels de Planell-Saguer
- Center for Motor Neuron Biology and Disease, Columbia University, New York, NY, 10032, USA
- Department of Pathology & Cell Biology, Columbia University, New York, NY, 10032, USA
- Department of Neurology, Columbia University, New York, NY, 10032, USA
| | - Hai Li
- Department of Systems Biology, Columbia University, New York, USA
- Sulzberger Columbia Genome Center, High Throughput Screening Facility, Columbia University Medical Center, New York, USA
| | - Cristina Meregalli
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Eleonora Pozzi
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Valentina Alda Carozzi
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Annalisa Canta
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Laura Monza
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Paola Alberti
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- Fondazione IRCCS San Gerardo Dei Tintori, Monza, Italy
| | - Giulia Fumagalli
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Charles Karan
- Department of Systems Biology, Columbia University, New York, USA
- Sulzberger Columbia Genome Center, High Throughput Screening Facility, Columbia University Medical Center, New York, USA
| | - Yalda Moayedi
- Department of Neurology, Columbia University, New York, NY, 10032, USA
- Department of Otolaryngology-Head & Neck Surgery, Columbia University, New York, NY, USA
| | - Serge Przedborski
- Center for Motor Neuron Biology and Disease, Columbia University, New York, NY, 10032, USA
- Department of Pathology & Cell Biology, Columbia University, New York, NY, 10032, USA
- Department of Neurology, Columbia University, New York, NY, 10032, USA
- Department of Neuroscience, Columbia University Medical Center, New York, USA
| | - Guido Cavaletti
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- Fondazione IRCCS San Gerardo Dei Tintori, Monza, Italy
| | - Francesco Lotti
- Center for Motor Neuron Biology and Disease, Columbia University, New York, NY, 10032, USA.
- Department of Pathology & Cell Biology, Columbia University, New York, NY, 10032, USA.
- Department of Neurology, Columbia University, New York, NY, 10032, USA.
| |
Collapse
|
7
|
Creanga-Murariu I, Filipiuc LE, Gogu MR, Ciorpac M, Cumpat CM, Tamba BI, Alexa-Stratulat T. The potential neuroprotective effects of cannabinoids against paclitaxel-induced peripheral neuropathy: in vitro study on neurite outgrowth. Front Pharmacol 2024; 15:1395951. [PMID: 38933665 PMCID: PMC11199736 DOI: 10.3389/fphar.2024.1395951] [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: 03/04/2024] [Accepted: 05/27/2024] [Indexed: 06/28/2024] Open
Abstract
Introduction: Chemotherapy-induced peripheral neuropathy (CIPN) is a shared burden for 68.1% of oncological patients undergoing chemotherapy with Paclitaxel (PTX). The symptoms are intense and troublesome, patients reporting paresthesia, loss of sensation, and dysesthetic pain. While current medications focus on decreasing the symptom intensity, often ineffective, no medication is yet recommended by the guidelines for the prevention of CIPN. Cannabinoids are an attractive option, as their neuroprotective features have already been demonstrated in neuropathies with other etiologies, by offering the peripheral neurons protection against toxic effects, which promotes analgesia. Methods: We aim to screen several new cannabinoids for their potential use as neuroprotective agents for CIPN by investigating the cellular toxicity profile and by assessing the potential neuroprotective features against PTX using a primary dorsal root ganglion neuronal culture. Results: Our study showed that synthetic cannabinoids JWH-007, AM-694 and MAB-CHMINACA and phytocannabinoids Cannabixir® Medium dried flowers (NC1) and Cannabixir® THC full extract (NC2) preserve the viability of fibroblasts and primary cultured neurons, in most of the tested dosages and time-points. The combination between the cannabinoids and PTX conducted to a cell viability of 70%-89% compared to 40% when PTX was administered alone for 48 h. When assessing the efficacy for neuroprotection, the combination between cannabinoids and PTX led to better preservation of neurite length at all tested time-points compared to controls, highly drug and exposure-time dependent. By comparison, the combination of the cannabinoids and PTX administered for 24 h conducted to axonal shortening between 23% and 44%, as opposed to PTX only, which shortened the axons by 63% compared to their baseline values. Discussion and Conclusion: Cannabinoids could be potential new candidates for the treatment of paclitaxel-induced peripheral neuropathy; however, our findings need to be followed by additional tests to understand the exact mechanism of action, which would support the translation of the cannabinoids in the oncological clinical practice.
Collapse
Affiliation(s)
- Ioana Creanga-Murariu
- Advanced Research and Development Center for Experimental Medicine (CEMEX), Iasi, Romania
- Grigore T. Popa University of Medicine and Pharmacy, Iasi, Romania
- Oncology Department, Regional Institute of Oncology, Iasi, Romania
| | - Leontina-Elena Filipiuc
- Advanced Research and Development Center for Experimental Medicine (CEMEX), Iasi, Romania
- Grigore T. Popa University of Medicine and Pharmacy, Iasi, Romania
| | - Maria-Raluca Gogu
- Advanced Research and Development Center for Experimental Medicine (CEMEX), Iasi, Romania
| | - Mitica Ciorpac
- Advanced Research and Development Center for Experimental Medicine (CEMEX), Iasi, Romania
| | - Carmen Marinela Cumpat
- Grigore T. Popa University of Medicine and Pharmacy, Iasi, Romania
- Clinical Rehabilitation Hospital, Cardiovascular and Respiratory Rehabilitation Clinic, Iasi, Romania
| | - Bogdan-Ionel Tamba
- Advanced Research and Development Center for Experimental Medicine (CEMEX), Iasi, Romania
- Grigore T. Popa University of Medicine and Pharmacy, Iasi, Romania
| | - Teodora Alexa-Stratulat
- Grigore T. Popa University of Medicine and Pharmacy, Iasi, Romania
- Oncology Department, Regional Institute of Oncology, Iasi, Romania
| |
Collapse
|
8
|
Kim JH, Cetinkaya-Fisgin A, Zahn N, Sari MC, Hoke A, Barman I. Label-Free Visualization and Morphological Profiling of Neuronal Differentiation and Axonal Degeneration through Quantitative Phase Imaging. Adv Biol (Weinh) 2024; 8:e2400020. [PMID: 38548657 PMCID: PMC11090721 DOI: 10.1002/adbi.202400020] [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: 03/13/2024] [Indexed: 05/15/2024]
Abstract
Understanding the intricate processes of neuronal growth, degeneration, and neurotoxicity is paramount for unraveling nervous system function and holds significant promise in improving patient outcomes, especially in the context of chemotherapy-induced peripheral neuropathy (CIPN). These processes are influenced by a broad range of entwined events facilitated by chemical, electrical, and mechanical signals. The progress of each process is inherently linked to phenotypic changes in cells. Currently, the primary means of demonstrating morphological changes rely on measurements of neurite outgrowth and axon length. However, conventional techniques for monitoring these processes often require extensive preparation to enable manual or semi-automated measurements. Here, a label-free and non-invasive approach is employed for monitoring neuronal differentiation and degeneration using quantitative phase imaging (QPI). Operating on unlabeled specimens and offering little to no phototoxicity and photobleaching, QPI delivers quantitative maps of optical path length delays that provide an objective measure of cellular morphology and dynamics. This approach enables the visualization and quantification of axon length and other physical properties of dorsal root ganglion (DRG) neuronal cells, allowing greater understanding of neuronal responses to stimuli simulating CIPN conditions. This research paves new avenues for the development of more effective strategies in the clinical management of neurotoxicity.
Collapse
Affiliation(s)
- Jeong Hee Kim
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Aysel Cetinkaya-Fisgin
- Department of Neurology, Neuromuscular Division, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Noah Zahn
- Department Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Mehmet Can Sari
- Department of Neurology, Neuromuscular Division, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Ahmet Hoke
- Department of Neurology, Neuromuscular Division, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Ishan Barman
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
- Department of Oncology, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| |
Collapse
|
9
|
Matsuda K, Han X, Matsuda N, Yamanaka M, Suzuki I. Development of an In Vitro Assessment Method for Chemotherapy-Induced Peripheral Neuropathy (CIPN) by Integrating a Microphysiological System (MPS) with Morphological Deep Learning of Soma and Axonal Images. TOXICS 2023; 11:848. [PMID: 37888698 PMCID: PMC10611258 DOI: 10.3390/toxics11100848] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 10/07/2023] [Accepted: 10/08/2023] [Indexed: 10/28/2023]
Abstract
Several anticancer drugs used in cancer therapy induce chemotherapy-induced peripheral neuropathy (CIPN), leading to dose reduction or therapy cessation. Consequently, there is a demand for an in vitro assessment method to predict CIPN and mechanisms of action (MoA) in drug candidate compounds. In this study, a method assessing the toxic effects of anticancer drugs on soma and axons using deep learning image analysis is developed, culturing primary rat dorsal root ganglion neurons with a microphysiological system (MPS) that separates soma from neural processes and training two artificial intelligence (AI) models on soma and axonal area images. Exposing the control compound DMSO, negative compound sucrose, and known CIPN-causing drugs (paclitaxel, vincristine, oxaliplatin, suramin, bortezomib) for 24 h, results show the somatic area-learning AI detected significant cytotoxicity for paclitaxel (* p < 0.05) and oxaliplatin (* p < 0.05). In addition, axonal area-learning AI detected significant axonopathy with paclitaxel (* p < 0.05) and vincristine (* p < 0.05). Combining these models, we detected significant toxicity in all CIPN-causing drugs (** p < 0.01) and could classify anticancer drugs based on their different MoA on neurons, suggesting that the combination of MPS-based culture segregating soma and axonal areas and AI image analysis of each area provides an effective evaluation method to predict CIPN from low concentrations and infer the MoA.
Collapse
Affiliation(s)
- Kazuki Matsuda
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-ku, Sendai 982-8577, Japan; (K.M.); (X.H.); (N.M.)
| | - Xiaobo Han
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-ku, Sendai 982-8577, Japan; (K.M.); (X.H.); (N.M.)
| | - Naoki Matsuda
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-ku, Sendai 982-8577, Japan; (K.M.); (X.H.); (N.M.)
| | - Makoto Yamanaka
- Business Creation Division Organs on Chip Project, Usio Inc., 1-6-5 Marunouchi, Chiyoda-ku, Tokyo 100-8150, Japan;
| | - Ikuro Suzuki
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-ku, Sendai 982-8577, Japan; (K.M.); (X.H.); (N.M.)
| |
Collapse
|
10
|
Yazar U, Guvercin AR, Rouhikia M, Aktoklu M, Demirci MA, Erbay I, Ayar A. Cerebrolysin provides effective protection on high glucose-induced neuropathy in cultured rat dorsal root ganglion neurons. J Recept Signal Transduct Res 2023; 43:109-114. [PMID: 38079610 DOI: 10.1080/10799893.2023.2291566] [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: 05/03/2023] [Accepted: 11/23/2023] [Indexed: 01/25/2024]
Abstract
Cerebrolysin, an endogenous peptide with neuroprotective and neurotrophic properties, indicated to be beneficial on diabetic neuropathy by preliminary clinical and experimental studies but without evidence on central or peripheral action. Dorsal root ganglion (DRG) neurons, based on involvement of pain sensation in both health and disease as first relay centers for transmission and processing of peripheral nociceptive sensory signals, was used to investigate possible effects of Cerebrolysin on high glucose-induced neuropathy, as model. DRG's were obtained from adult rats and the isolated neurons were seeded on E-Plate®'s equipped with gold microelectrodes, and incubated in culture media in a CO2 incubator at 37 C. DRGs were exposed to high glucose (50 mM) in the absence and presence of different concentrations of Cerebrolysin ® (2-40 mg/ml). Cell index (derived from cell viability and neurite outgrowth) was recorded with Real-Time Cell Analyzer and was used as primary outcome measure. High glucose-induced cellular neuropathy and neuroprotective effects of Cerebrolysin was evaluated from area under the curve (AUC) of cell index-time graphs. Exposure of DRG neurons to high glucose caused a rapid and persistent decrease in the mean AUC values compared to normoglycemic controls. Co-treatment with Cerebrolysin (40 mg/ml) attenuated this high glucose-induced effect in a concentration-dependent manner. In normoglycemic conditions, treatment with Cerebrolysin caused a dose-dependent increase in the mean AUC values. Cerebrolysin treatment resulted in maintenance of the functional integrity, survival, and promotion of neurite outgrowth of the cultured DRG neurons exposed to high glucose, indicating involvement of peripheral sensory neurons.
Collapse
Affiliation(s)
- Ugur Yazar
- Departments of Neurosurgery, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | - Ali Rıza Guvercin
- Departments of Neurosurgery, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | - Mahindokht Rouhikia
- Departments of Physiology, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | - Mehmet Aktoklu
- Departments of Neurosurgery, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | - Mehmet Ali Demirci
- Departments of Neurosurgery, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | - Ibrahim Erbay
- Departments of Physiology, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | - Ahmet Ayar
- Departments of Physiology, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| |
Collapse
|
11
|
Vecchi JT, Mullan S, Lopez JA, Rhomberg M, Yamamoto A, Hallam A, Lee A, Sonka M, Hansen MR. Sensitivity of CNN image analysis to multifaceted measurements of neurite growth. BMC Bioinformatics 2023; 24:320. [PMID: 37620759 PMCID: PMC10464248 DOI: 10.1186/s12859-023-05444-4] [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: 12/14/2022] [Accepted: 08/11/2023] [Indexed: 08/26/2023] Open
Abstract
Quantitative analysis of neurite growth and morphology is essential for understanding the determinants of neural development and regeneration, however, it is complicated by the labor-intensive process of measuring diverse parameters of neurite outgrowth. Consequently, automated approaches have been developed to study neurite morphology in a high-throughput and comprehensive manner. These approaches include computer-automated algorithms known as 'convolutional neural networks' (CNNs)-powerful models capable of learning complex tasks without the biases of hand-crafted models. Nevertheless, their complexity often relegates them to functioning as 'black boxes.' Therefore, research in the field of explainable AI is imperative to comprehend the relationship between CNN image analysis output and predefined morphological parameters of neurite growth in order to assess the applicability of these machine learning approaches. In this study, drawing inspiration from the field of automated feature selection, we investigate the correlation between quantified metrics of neurite morphology and the image analysis results from NeuriteNet-a CNN developed to analyze neurite growth. NeuriteNet accurately distinguishes images of neurite growth based on different treatment groups within two separate experimental systems. These systems differentiate between neurons cultured on different substrate conditions and neurons subjected to drug treatment inhibiting neurite outgrowth. By examining the model's function and patterns of activation underlying its classification decisions, we discover that NeuriteNet focuses on aspects of neuron morphology that represent quantifiable metrics distinguishing these groups. Additionally, it incorporates factors that are not encompassed by neuron morphology tracing analyses. NeuriteNet presents a novel tool ideally suited for screening morphological differences in heterogeneous neuron groups while also providing impetus for targeted follow-up studies.
Collapse
Affiliation(s)
- Joseph T Vecchi
- Department of Molecular Physiology and Biophysics, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Department of Otolaryngology Head-Neck Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Sean Mullan
- Iowa Institute for Biomedical Imaging, Electrical and Computer Engineering, University of Iowa, Iowa City, IA, USA
| | - Josue A Lopez
- Department of Neuroscience, University of Texas-Austin, Austin, TX, USA
| | - Madeline Rhomberg
- Department of Otolaryngology Head-Neck Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | | | - Annabelle Hallam
- Department of Otolaryngology Head-Neck Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Amy Lee
- Department of Neuroscience, University of Texas-Austin, Austin, TX, USA
| | - Milan Sonka
- Iowa Institute for Biomedical Imaging, Electrical and Computer Engineering, University of Iowa, Iowa City, IA, USA
| | - Marlan R Hansen
- Department of Molecular Physiology and Biophysics, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
- Department of Otolaryngology Head-Neck Surgery, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
12
|
Chang YC, Lo YC, Chang HS, Lin HC, Chiu CC, Chen YF. An efficient cellular image-based platform for high-content screening of neuroprotective agents against chemotherapy-induced neuropathy. Neurotoxicology 2023; 96:118-128. [PMID: 37086979 DOI: 10.1016/j.neuro.2023.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/12/2023] [Accepted: 04/19/2023] [Indexed: 04/24/2023]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a major dose-limiting side effect, with no approved therapy for prevention or treatment. Here, we aimed to establish a high-content image platform based on the neurite outgrowth of dorsal root ganglia (DRG)-derived neuron cells for the discovery of neuroprotective agents against paclitaxel-induced CIPN. ND7/23 cells, an immortalized hybrid DRG cell line, were maturely differentiated by induction with nerve growth factor and upregulation of intracellular cAMP levels. High-content image analyses of the neurofilament-stained neurite network showed that paclitaxel disrupted the neurite outgrowth of well-differentiated ND7/23 DRG neuron cells, recapitulating characteristic effects of paclitaxel on primary cultured DRG neurons. This process coincided with the upregulated activity of store-operated Ca2+ entry, similar to those found in rodent models of paclitaxel-induced CIPN. The previously identified neuroprotective agents, minoxidil and 8-Br-cyclic adenosine monophosphate ribose (8-Br-cADPR), attenuated the reduction in total neurite outgrowth in paclitaxel-damaged ND7/23 cells. Additionally, the total neurite outgrowth of well-differentiated ND7/23 cells was concentration-dependently reduced by the neurotoxic chemotherapeutic agents, oxaliplatin and bortezomib, but not the less neurotoxic 5-fluorouracil. We demonstrated that high-content analyses of neurite morphology in well-differentiated DRG neuron-derived cells provide an effective, reproducible, and high-throughput strategy for developing therapeutics against CIPN.
Collapse
Affiliation(s)
- Yang-Chen Chang
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yi-Ching Lo
- Department of Pharmacology, School of Medicine, Kaohsiung Medical University, Taiwan
| | - Hsun-Shuo Chang
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan; School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Taiwan; Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Hui-Ching Lin
- Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chien-Chih Chiu
- Department of Biotechnology, School of Life Science, Kaohsiung Medical University, Taiwan
| | - Yih-Fung Chen
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan; School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Taiwan; Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.
| |
Collapse
|
13
|
Snavely AR, Heo K, Petrova V, Ho TSY, Huang X, Hermawan C, Kagan R, Deng T, Singeç I, Chen L, Barret LB, Woolf CJ. Bortezomib-induced neurotoxicity in human neurons is the consequence of nicotinamide adenine dinucleotide depletion. Dis Model Mech 2022; 15:dmm049358. [PMID: 36398590 PMCID: PMC9789399 DOI: 10.1242/dmm.049358] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 11/07/2022] [Indexed: 11/19/2022] Open
Abstract
The proteosome inhibitor bortezomib has revolutionized the treatment of multiple hematologic malignancies, but in many cases, its efficacy is limited by a dose-dependent peripheral neuropathy. We show that human induced pluripotent stem cell (hiPSC)-derived motor neurons and sensory neurons provide a model system for the study of bortezomib-induced peripheral neuropathy, with promising implications for furthering the mechanistic understanding of and developing treatments for preventing axonal damage. Human neurons in tissue culture displayed distal-to-proximal neurite degeneration when exposed to bortezomib. This process coincided with disruptions in mitochondrial function and energy homeostasis, similar to those described in rodent models of bortezomib-induced neuropathy. Moreover, although the degenerative process was unaffected by inhibition of caspases, it was completely blocked by exogenous nicotinamide adenine dinucleotide (NAD+), a mediator of the SARM1-dependent axon degeneration pathway. We demonstrate that bortezomib-induced neurotoxicity in relevant human neurons proceeds through mitochondrial dysfunction and NAD+ depletion-mediated axon degeneration, raising the possibility that targeting these changes might provide effective therapeutics for the prevention of bortezomib-induced neuropathy and that modeling chemotherapy-induced neuropathy in human neurons has utility.
Collapse
Affiliation(s)
- Andrew R. Snavely
- F.M. Kirby Neurobiology Center, Program in Neurobiology, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Keungjung Heo
- F.M. Kirby Neurobiology Center, Program in Neurobiology, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Veselina Petrova
- F.M. Kirby Neurobiology Center, Program in Neurobiology, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Tammy Szu-Yu Ho
- F.M. Kirby Neurobiology Center, Program in Neurobiology, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Xuan Huang
- F.M. Kirby Neurobiology Center, Program in Neurobiology, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Crystal Hermawan
- F.M. Kirby Neurobiology Center, Program in Neurobiology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Ruth Kagan
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Tao Deng
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Ilyas Singeç
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Long Chen
- F.M. Kirby Neurobiology Center, Program in Neurobiology, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Lee B. Barret
- F.M. Kirby Neurobiology Center, Program in Neurobiology, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Clifford J. Woolf
- F.M. Kirby Neurobiology Center, Program in Neurobiology, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
14
|
Tsai NW, Lin CC, Yeh TY, Chiu YA, Chiu HH, Huang HP, Hsieh ST. An induced pluripotent stem cell-based model identifies molecular targets of vincristine neurotoxicity. Dis Model Mech 2022; 15:dmm049471. [PMID: 36518084 PMCID: PMC10655812 DOI: 10.1242/dmm.049471] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 09/29/2022] [Indexed: 11/19/2023] Open
Abstract
To model peripheral nerve degeneration and investigate molecular mechanisms of neurodegeneration, we established a cell system of induced pluripotent stem cell (iPSC)-derived sensory neurons exposed to vincristine, a drug that frequently causes chemotherapy-induced peripheral neuropathy. Sensory neurons differentiated from iPSCs exhibit distinct neurochemical patterns according to the immunocytochemical phenotypes, and gene expression of peripherin (PRPH, hereafter referred to as Peri) and neurofilament heavy chain (NEFH, hereafter referred to as NF). The majority of iPSC-derived sensory neurons were PRPH positive/NEFH negative, i.e. Peri(+)/NF(-) neurons, whose somata were smaller than those of Peri(+)/NF(+) neurons. On exposure to vincristine, projections from the cell body of a neuron, i.e. neurites, were degenerated quicker than somata, the lethal concentration to kill 50% (LC50) of neurites being below the LC50 for somata, consistent with the clinical pattern of length-dependent neuropathy. We then examined the molecular expression in the MAP kinase signaling pathways of, extracellular signal-regulated kinases 1/2 (MAPK1/3, hereafter referred to as ERK), p38 mitogen-activated protein kinases (MAPK11/12/13/14, hereafter referred to as p38) and c-Jun N-terminal kinases (MAPK8/9/10, hereafter referred to as JNK). Regarding these three cascades, only phosphorylation of JNK was upregulated but not that of p38 or ERK1/2. Furthermore, vincristine-treatment resulted in impaired autophagy and reduced autophagic flux. Rapamycin-treatment reversed the effect of impaired autophagy and JNK activation. These results not only established a platform to study peripheral degeneration of human neurons but also provide molecular mechanisms for neurodegeneration with the potential for therapeutic targets.
Collapse
Affiliation(s)
- Neng-Wei Tsai
- Department of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei 100, Taiwan
| | - Cheng-Chen Lin
- Department of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei 100, Taiwan
| | - Ti-Yen Yeh
- Department of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei 100, Taiwan
| | - Yu-An Chiu
- Department of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei 100, Taiwan
| | - Hsin-Hui Chiu
- Department of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei 100, Taiwan
| | - Hsiang-Po Huang
- Department of Medical Genomics and Proteomics, National Taiwan University College of Medicine, Taipei 100, Taiwan
- Department of Pediatrics, National Taiwan University Children's Hospital, Taipei 100, Taiwan
| | - Sung-Tsang Hsieh
- Department of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei 100, Taiwan
- Department of Brain and Mind Sciences, National Taiwan University College of Medicine, Taipei 100, Taiwan
- Department of Neurology, National Taiwan University Hospital, Taipei 100, Taiwan
| |
Collapse
|
15
|
Klein I, Boenert J, Lange F, Christensen B, Wassermann MK, Wiesen MHJ, Olschewski DN, Rabenstein M, Müller C, Lehmann HC, Fink GR, Schroeter M, Rueger MA, Vay SU. Glia from the central and peripheral nervous system are differentially affected by paclitaxel chemotherapy via modulating their neuroinflammatory and neuroregenerative properties. Front Pharmacol 2022; 13:1038285. [PMID: 36408236 PMCID: PMC9666700 DOI: 10.3389/fphar.2022.1038285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 10/06/2022] [Indexed: 11/07/2022] Open
Abstract
Glia are critical players in defining synaptic contacts and maintaining neuronal homeostasis. Both astrocytes as glia of the central nervous system (CNS), as well as satellite glial cells (SGC) as glia of the peripheral nervous system (PNS), intimately interact with microglia, especially under pathological conditions when glia regulate degenerative as well as regenerative processes. The chemotherapeutic agent paclitaxel evokes peripheral neuropathy and cognitive deficits; however, the mechanisms underlying these diverse clinical side effects are unclear. We aimed to elucidate the direct effects of paclitaxel on the function of astrocytes, microglia, and SGCs, and their glia-glia and neuronal-glia interactions. After intravenous application, paclitaxel was present in the dorsal root ganglia of the PNS and the CNS of rodents. In vitro, SGC enhanced the expression of pro-inflammatory factors and reduced the expression of neurotrophic factor NT-3 upon exposure to paclitaxel, resulting in predominantly neurotoxic effects. Likewise, paclitaxel induced a switch towards a pro-inflammatory phenotype in microglia, exerting neurotoxicity. In contrast, astrocytes expressed neuroprotective markers and increasingly expressed S100A10 after paclitaxel exposure. Astrocytes, and to a lesser extent SGCs, had regulatory effects on microglia independent of paclitaxel exposure. Data suggest that paclitaxel differentially modulates glia cells regarding their (neuro-) inflammatory and (neuro-) regenerative properties and also affects their interaction. By elucidating those processes, our data contribute to the understanding of the mechanistic pathways of paclitaxel-induced side effects in CNS and PNS.
Collapse
Affiliation(s)
- Ines Klein
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Neurology, Cologne, Germany
| | - Janne Boenert
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Neurology, Cologne, Germany
| | - Felix Lange
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Neurology, Cologne, Germany
| | - Britt Christensen
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Neurology, Cologne, Germany
| | - Meike K. Wassermann
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Neurology, Cologne, Germany
| | - Martin H. J. Wiesen
- Center of Pharmacology, Therapeutic Drug Monitoring, University Hospital of Cologne, Cologne, Germany
| | - Daniel Navin Olschewski
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Neurology, Cologne, Germany
| | - Monika Rabenstein
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Neurology, Cologne, Germany
| | - Carsten Müller
- Center of Pharmacology, Therapeutic Drug Monitoring, University Hospital of Cologne, Cologne, Germany
| | - Helmar C. Lehmann
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Neurology, Cologne, Germany
| | - Gereon Rudolf Fink
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Neurology, Cologne, Germany
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Centre Juelich, Juelich, Germany
| | - Michael Schroeter
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Neurology, Cologne, Germany
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Centre Juelich, Juelich, Germany
| | - Maria Adele Rueger
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Neurology, Cologne, Germany
- Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Centre Juelich, Juelich, Germany
| | - Sabine Ulrike Vay
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Neurology, Cologne, Germany
- *Correspondence: Sabine Ulrike Vay,
| |
Collapse
|
16
|
Alotaibi M, Al-Aqil F, Alqahtani F, Alanazi M, Nadeem A, Ahmad SF, Lapresa R, Alharbi M, Alshammari A, Alotaibi M, Saleh T, Alrowis R. Alleviation of cisplatin-induced neuropathic pain, neuronal apoptosis, and systemic inflammation in mice by rapamycin. Front Aging Neurosci 2022; 14:891593. [PMID: 36248001 PMCID: PMC9554141 DOI: 10.3389/fnagi.2022.891593] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 08/15/2022] [Indexed: 11/30/2022] Open
Abstract
Platinum-based chemotherapeutic treatment of cancer patients is associated with debilitating adverse effects. Several adverse effects have been well investigated, and can be managed satisfactorily, but chemotherapy-induced peripheral neuropathy (CIPN) remains poorly treated. Our primary aim in this study was to investigate the neuroprotective effect of the immunomodulatory drug rapamycin in the mitigation of cisplatin-induced neurotoxicity. Pain assays were performed in vivo to determine whether rapamycin would prevent or significantly decrease cisplatin-induced neurotoxicity in adult male Balb/c mice. Neuropathic pain induced by both chronic and acute exposure to cisplatin was measured by hot plate assay, cold plate assay, tail-flick test, and plantar test. Rapamycin co-treatment resulted in significant reduction in cisplatin-induced nociceptive-like symptoms. To understand the underlying mechanisms behind rapamycin-mediated neuroprotection, we investigated its effect on certain inflammatory mediators implicated in the propagation of chemotherapy-induced neurotoxicity. Interestingly, cisplatin was found to significantly increase peripheral IL-17A expression and CD8- T cells, which were remarkably reversed by the pre-treatment of mice with rapamycin. In addition, rapamycin reduced the cisplatin-induced neuronal apoptosis marked by decreased neuronal caspase-3 activity. The rapamycin neuroprotective effect was also associated with reversal of the changes in protein expression of p21Cip1, p53, and PUMA. Collectively, rapamycin alleviated some features of cisplatin-induced neurotoxicity in mice and can be further investigated for the treatment of cisplatin-induced peripheral neuropathy.
Collapse
Affiliation(s)
- Moureq Alotaibi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
- *Correspondence: Moureq Alotaibi,
| | - Faten Al-Aqil
- Deanship of Scientific Research, King Saud University, Riyadh, Saudi Arabia
| | - Faleh Alqahtani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Miteb Alanazi
- Pharmacy Services, King Saud University Medical City, Riyadh, Saudi Arabia
| | - Ahmed Nadeem
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Sheikh F. Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Rebeca Lapresa
- Institute of Functional Biology and Genomics, Consejo Superior de Investigaciones Científicas (CSIC), University of Salamanca, Salamanca, Spain
- Institute of Biomedical Research of Salamanca, University Hospital of Salamanca, Consejo Superior de Investigaciones Científicas (CSIC), University of Salamanca, Salamanca, Spain
| | - Metab Alharbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Abdulrahman Alshammari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Muteb Alotaibi
- Department of Neurology, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Tareq Saleh
- Department of Basic Medical Sciences, Faculty of Medicine, The Hashemite University, Zarqa, Jordan
| | - Raed Alrowis
- Department of Periodotics and Community Dentistry, College of Dentistry, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
17
|
de Clauser L, Kappert C, Sondermann JR, Gomez-Varela D, Flatters SJL, Schmidt M. Proteome and Network Analysis Provides Novel Insights Into Developing and Established Chemotherapy-Induced Peripheral Neuropathy. Front Pharmacol 2022; 13:818690. [PMID: 35250568 PMCID: PMC8895144 DOI: 10.3389/fphar.2022.818690] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 01/12/2022] [Indexed: 01/09/2023] Open
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a debilitating side-effect of cancer therapies. So far, the development of CIPN cannot be prevented, neither can established CIPN be reverted, often leading to the cessation of necessary chemotherapy. Thus, there is an urgent need to explore the mechanistic basis of CIPN to facilitate its treatment. Here we used an integrated approach of quantitative proteome profiling and network analysis in a clinically relevant rat model of paclitaxel-induced peripheral neuropathy. We analysed lumbar rat DRG at two critical time points: (1) day 7, right after cessation of paclitaxel treatment, but prior to neuropathy development (pre-CIPN); (2) 4 weeks after paclitaxel initiation, when neuropathy has developed (peak-CIPN). In this way we identified a differential protein signature, which shows how changes in the proteome correlate with the development and maintenance of CIPN, respectively. Extensive biological pathway and network analysis reveals that, at pre-CIPN, regulated proteins are prominently implicated in mitochondrial (dys)function, immune signalling, neuronal damage/regeneration, and neuronal transcription. Orthogonal validation in an independent rat cohort confirmed the increase of β-catenin (CTNNB1) at pre-CIPN. More importantly, detailed analysis of protein networks associated with β-catenin highlights translationally relevant and potentially druggable targets. Overall, this study demonstrates the enormous value of combining animal behaviour with proteome and network analysis to provide unprecedented insights into the molecular basis of CIPN. In line with emerging approaches of network medicine our results highlight new avenues for developing improved therapeutic options aimed at preventing and treating CIPN.
Collapse
Affiliation(s)
- Larissa de Clauser
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Bolzano, Italy
- *Correspondence: Larissa de Clauser, ; Manuela Schmidt,
| | - Christin Kappert
- Max Planck Institute of Experimental Medicine, Goettingen, Germany
| | - Julia R. Sondermann
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
| | - David Gomez-Varela
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
| | - Sarah J. L. Flatters
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Manuela Schmidt
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
- *Correspondence: Larissa de Clauser, ; Manuela Schmidt,
| |
Collapse
|
18
|
Holzer AK, Karreman C, Suciu I, Furmanowsky LS, Wohlfarth H, Loser D, Dirks WG, Pardo González E, Leist M. OUP accepted manuscript. Stem Cells Transl Med 2022; 11:727-741. [PMID: 35689659 PMCID: PMC9299516 DOI: 10.1093/stcltm/szac031] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 04/09/2022] [Indexed: 11/12/2022] Open
Abstract
In vitro models of the peripheral nervous system would benefit from further refinements to better support studies on neuropathies. In particular, the assessment of pain-related signals is still difficult in human cell cultures. Here, we harnessed induced pluripotent stem cells (iPSCs) to generate peripheral sensory neurons enriched in nociceptors. The objective was to generate a culture system with signaling endpoints suitable for pharmacological and toxicological studies. Neurons generated by conventional differentiation protocols expressed moderate levels of P2X3 purinergic receptors and only low levels of TRPV1 capsaicin receptors, when maturation time was kept to the upper practically useful limit of 6 weeks. As alternative approach, we generated cells with an inducible NGN1 transgene. Ectopic expression of this transcription factor during a defined time window of differentiation resulted in highly enriched nociceptor cultures, as determined by functional (P2X3 and TRPV1 receptors) and immunocytochemical phenotyping, complemented by extensive transcriptome profiling. Single cell recordings of Ca2+-indicator fluorescence from >9000 cells were used to establish the “fraction of reactive cells” in a stimulated population as experimental endpoint, that appeared robust, transparent and quantifiable. To provide an example of application to biomedical studies, functional consequences of prolonged exposure to the chemotherapeutic drug oxaliplatin were examined at non-cytotoxic concentrations. We found (i) neuronal (allodynia-like) hypersensitivity to otherwise non-activating mechanical stimulation that could be blocked by modulators of voltage-gated sodium channels; (ii) hyper-responsiveness to TRPV1 receptor stimulation. These findings and several other measured functional alterations indicate that the model is suitable for pharmacological and toxicological studies related to peripheral neuropathies.
Collapse
Affiliation(s)
- Anna-Katharina Holzer
- In vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Konstanz, Germany
- Graduate School Biological Sciences (GBS), University of Konstanz, Konstanz, Germany
| | - Christiaan Karreman
- In vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Konstanz, Germany
| | - Ilinca Suciu
- In vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Konstanz, Germany
| | - Lara-Seline Furmanowsky
- In vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Konstanz, Germany
| | - Harald Wohlfarth
- In vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Konstanz, Germany
| | - Dominik Loser
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Wilhelm G Dirks
- Department of Human and Animal Cell Lines, DSMZ, German Collection of Microorganisms and Cell Cultures and German Biological Resource Center, Braunschweig, Germany
| | - Emilio Pardo González
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Marcel Leist
- Corresponding author: Marcel Leist, PhD, In Vitro Toxicology and Biomedicine, Dept Inaugurated by the Doerenkamp-Zbinden Foundation at the University of Konstanz, Universitaetsstr. 10, Konstanz 78457, Germany.
| |
Collapse
|
19
|
Considerations for a Reliable In Vitro Model of Chemotherapy-Induced Peripheral Neuropathy. TOXICS 2021; 9:toxics9110300. [PMID: 34822690 PMCID: PMC8620674 DOI: 10.3390/toxics9110300] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 12/13/2022]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is widely recognized as a potentially severe toxicity that often leads to dose reduction or discontinuation of cancer treatment. Symptoms may persist despite discontinuation of chemotherapy and quality of life can be severely compromised. The clinical symptoms of CIPN, and the cellular and molecular targets involved in CIPN, are just as diverse as the wide variety of anticancer agents that cause peripheral neurotoxicity. There is an urgent need for extensive molecular and functional investigations aimed at understanding the mechanisms of CIPN. Furthermore, a reliable human cell culture system that recapitulates the diversity of neuronal modalities found in vivo and the pathophysiological changes that underlie CIPN would serve to advance the understanding of the pathogenesis of CIPN. The demonstration of experimental reproducibility in a human peripheral neuronal cell system will increase confidence that such an in vitro model is clinically useful, ultimately resulting in deeper exploration for the prevention and treatment of CIPN. Herein, we review current in vitro models with a focus on key characteristics and attributes desirable for an ideal human cell culture model relevant for CIPN investigations.
Collapse
|
20
|
Wu S, Bai X, Guo C, Huang Z, Ouyang H, Huang J, Zeng W. Ganglioside-monosialic acid (GM1) for prevention of chemotherapy-induced peripheral neuropathy: a meta-analysis with trial sequential analysis. BMC Cancer 2021; 21:1173. [PMID: 34727879 PMCID: PMC8564974 DOI: 10.1186/s12885-021-08884-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 10/14/2021] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Chemotherapy-induced peripheral neuropathy (CIPN) is a dose-limiting side effect that largely remains an unresolved clinical issue, leading to long-term morbidity. This meta-analysis aimed to evaluate the efficacy and safety of Ganglioside-monosialic acid (GM1) in preventing CIPN. METHODS Systematic literature searches of PubMed, Web of Science, Embase, the Cochrane Central Register of Controlled Trials, and ClinicalTrials.gov were performed to identify randomized controlled trials and cohort studies that evaluated the efficacy of GM1 for preventing CIPN. Conventional meta-analysis with a random-effects model and trial sequential analysis (TSA) were performed. RESULTS A total of five studies involving 868 participants were included. The results showed that GM1 did not reduce the overall incidence of grade ≥ 2 CIPN when the common terminology criteria for adverse events (CTCAE) was used (OR 0.34, 95% CI 0.34-1.11). Subgroup analyses showed that GM1 could not reduce the risk of CTCAE grade ≥ 2 CIPN (OR 0.63, 95% CI 0.35-1.13) and neurotoxicity criteria of Debiopharm (DEB-NTC) grade ≥ 2 CIPN (OR 0.25, 95% CI 0.01-7.10) in oxaliplatin-treated patients, despite that GM1 was associated with a reduced risk of CTCAE grade ≥ 2 CIPN in the taxane subgroup of one study (OR 0.003, 95% CI 0.00-0.05). These results were confirmed by the sub-analysis of randomized controlled trials (RCTs). In TSA, the z-curve for the taxane subgroup crossed the upper trial sequential monitoring boundary (TSMB) but do not reach the required information size (RIS). The z-curves for the oxaliplatin subgroup remained in the nonsignificant area and did not reach the RIS. Further, GM1 did not influence the rate of response to chemotherapy and CTCAE grade ≥ 2 adverse events such as fatigue, nausea, diarrhea, and rash. CONCLUSIONS GM1 seemed to be well-tolerated and did not influence the anti-cancer effects of chemotherapeutic agents. Although the data did not confirm the effectiveness of GM1 in preventing oxaliplatin-induced peripheral neuropathy, GM1 might be able to prevent taxane-induced peripheral neuropathy. More studies are required in different ethnic populations receiving taxane-based chemotherapy to confirm these findings.
Collapse
Affiliation(s)
- Shaoyong Wu
- Department of Anesthesiology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, 510060, P. R. China
| | - Xiaohui Bai
- Department of Anesthesiology, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510289, P. R. China
| | - Caixia Guo
- Department of Obstetrics, The Fourth Affiliated Hospital of Guangzhou Medical University (Zengcheng District People's Hospital), Guangzhou, Guangdong, 511300, P. R. China
| | - Zhimei Huang
- Department of Minimal Invasive Intervention, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, 510060, P. R. China
| | - Handong Ouyang
- Department of Anesthesiology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, 510060, P. R. China
| | - Jingxiu Huang
- Department of Anesthesiology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, 510060, P. R. China
| | - Weian Zeng
- Department of Anesthesiology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, 510060, P. R. China.
| |
Collapse
|
21
|
Klimas R, Sgodzai M, Motte J, Mohamad N, Renk P, Blusch A, Grüter T, Pedreiturria X, Gobrecht P, Fischer D, Schneider-Gold C, Reinacher-Schick A, Tannapfel A, Yoon MS, Gold R, Pitarokoili K. Dose-dependent immunomodulatory effects of bortezomib in experimental autoimmune neuritis. Brain Commun 2021; 3:fcab238. [PMID: 34708206 PMCID: PMC8545613 DOI: 10.1093/braincomms/fcab238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 05/02/2021] [Accepted: 06/09/2021] [Indexed: 11/13/2022] Open
Abstract
Proteasome inhibition with bortezomib has been reported to exert an immunomodulatory action in chronic autoimmune neuropathies. However, bortezomib used for the treatment of multiple myeloma induces a painful toxic polyneuropathy at a higher concentration. Therefore, we addressed this controversial effect and evaluated the neurotoxic and immunomodulatory mode of action of bortezomib in experimental autoimmune neuritis. Bortezomib-induced neuropathy was investigated in Lewis rats using the von Frey hair test, electrophysiological, qPCR and histological analyses of the sciatic nerve as well as dorsal root ganglia outgrowth studies. The immunomodulatory potential of bortezomib was characterized in Lewis rats after experimental autoimmune neuritis induction with P253-78 peptide. Clinical, electrophysiological, histological evaluation, von Frey hair test, flow cytometric and mRNA analyses were used to unravel the underlying mechanisms. We defined the toxic concentration of 0.2 mg/kg bortezomib applied intraperitoneally at Days 0, 4, 8 and 12. This dosage induces a painful toxic neuropathy but preserves axonal regeneration in vitro. Bortezomib at a concentration of 0.05 mg/kg significantly ameliorated experimental autoimmune neuritis symptoms, improved experimental autoimmune neuritis-induced hyperalgesia and nerve conduction studies, and reduced immune cell infiltration. Furthermore, proteasome inhibition induced a transcriptional downregulation of Nfkb in the sciatic nerve, while its inhibitor Ikba (also known as Nfkbia) was upregulated. Histological analyses of bone marrow tissue revealed a compensatory increase of CD138+ plasma cells. Our data suggest that low dose bortezomib (0.05 mg/kg intraperitoneally) has an immunomodulatory effect in the context of experimental autoimmune neuritis through proteasome inhibition and downregulation of nuclear factor 'kappa-light-chain-enhancer' of activated B-cells (NFKB). Higher bortezomib concentrations (0.2 mg/kg intraperitoneally) induce sensory neuropathy; however, the regeneration potential remains unaffected. Our data empathizes that bortezomib may serve as an attractive treatment option for inflammatory neuropathies in lower concentrations.
Collapse
Affiliation(s)
- Rafael Klimas
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Melissa Sgodzai
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Jeremias Motte
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Nuwin Mohamad
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Pia Renk
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Alina Blusch
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Thomas Grüter
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Xiomara Pedreiturria
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Philipp Gobrecht
- Department of Cell Physiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, 44801 Bochum, Germany
| | - Dietmar Fischer
- Department of Cell Physiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, 44801 Bochum, Germany
| | | | - Anke Reinacher-Schick
- Department of Oncology, St. Josef-Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Andrea Tannapfel
- Institute of Pathology, Ruhr-University Bochum, 44801 Bochum, Germany
| | - Min-Suk Yoon
- Department of Neurology, Evangelisches Krankenhaus Hattingen, 45525 Hattingen, Germany
| | - Ralf Gold
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Kalliopi Pitarokoili
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
| |
Collapse
|
22
|
Pathomechanisms of Paclitaxel-Induced Peripheral Neuropathy. TOXICS 2021; 9:toxics9100229. [PMID: 34678925 PMCID: PMC8540213 DOI: 10.3390/toxics9100229] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/09/2021] [Accepted: 09/16/2021] [Indexed: 12/18/2022]
Abstract
Peripheral neuropathy is one of the most common side effects of chemotherapy, affecting up to 60% of all cancer patients receiving chemotherapy. Moreover, paclitaxel induces neuropathy in up to 97% of all gynecological and urological cancer patients. In cancer cells, paclitaxel induces cell death via microtubule stabilization interrupting cell mitosis. However, paclitaxel also affects cells of the central and peripheral nervous system. The main symptoms are pain and numbness in hands and feet due to paclitaxel accumulation in the dorsal root ganglia. This review describes in detail the pathomechanisms of paclitaxel in the peripheral nervous system. Symptoms occur due to a length-dependent axonal sensory neuropathy, where axons are symmetrically damaged and die back. Due to microtubule stabilization, axonal transport is disrupted, leading to ATP undersupply and oxidative stress. Moreover, mitochondria morphology is altered during paclitaxel treatment. A key player in pain sensation and axonal damage is the paclitaxel-induced inflammation in the spinal cord as well as the dorsal root ganglia. An increased expression of chemokines and cytokines such as IL-1β, IL-8, and TNF-α, but also CXCR4, RAGE, CXCL1, CXCL12, CX3CL1, and C3 promote glial activation and accumulation, and pain sensation. These findings are further elucidated in this review.
Collapse
|
23
|
Wang M, Wang J, Tsui AYP, Li Z, Zhang Y, Zhao Q, Xing H, Wang X. Mechanisms of peripheral neurotoxicity associated with four chemotherapy drugs using human induced pluripotent stem cell-derived peripheral neurons. Toxicol In Vitro 2021; 77:105233. [PMID: 34390763 DOI: 10.1016/j.tiv.2021.105233] [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: 03/21/2021] [Revised: 07/22/2021] [Accepted: 08/09/2021] [Indexed: 01/22/2023]
Abstract
The awareness of the long-term toxicities of cancer survivors after chemotherapy treatment has been gradually strengthened as the population of cancer survivors grows. Generally, chemotherapy-induced peripheral neurotoxicity (CIPN) is studied by animal models which are not only expensive and time-consuming, but also species-specific differences. The generation of human induced pluripotent stem cells (hiPSCs) and differentiation of peripheral neurons have provided an in vitro model to elucidate the risk of CIPN. Here, we developed a drug-induced peripheral neurotoxicity model using hiPSC-derived peripheral neurons (hiPSC-PNs) to study the mechanisms of different chemotherapeutic agents on neuronal viability using LDH assay, a cell apoptosis assay determined by caspase 3/7 activation, neurite outgrowth, ion channel expression and neurotransmitter release following treatment of cisplatin, bortezomib, ixabepilone, or pomalidomide. Our data showed that the multiple endpoints of the hiPSC-PNs model had different sensitivity to various chemotherapeutic agents. Furthermore, the chemotherapeutics separated cell viability from the decrease in neurite lengthand changed levels of ion channels and neurotransmitters to a certain extent. Thus, we study the mechanisms of peripheral neurotoxicity induced by chemotherapeutic agents through changes in these indicators.
Collapse
Affiliation(s)
- Meiting Wang
- China State Institute of Pharmaceutical Industry, Shanghai InnoStar Bio-Tech Co., Ltd., Shanghai 201203, China
| | - Jiaxian Wang
- Nanjing HELP Stem Cell Innovations Co., Ltd., Nanjing 211100, China
| | - Alex Y P Tsui
- Nanjing HELP Stem Cell Innovations Co., Ltd., Nanjing 211100, China
| | - Zhaomin Li
- Nanjing HELP Stem Cell Innovations Co., Ltd., Nanjing 211100, China
| | - Yizhe Zhang
- China State Institute of Pharmaceutical Industry, Shanghai InnoStar Bio-Tech Co., Ltd., Shanghai 201203, China
| | - Qi Zhao
- China State Institute of Pharmaceutical Industry, Shanghai InnoStar Bio-Tech Co., Ltd., Shanghai 201203, China
| | - Hongyan Xing
- China State Institute of Pharmaceutical Industry, Shanghai InnoStar Bio-Tech Co., Ltd., Shanghai 201203, China
| | - Xijie Wang
- China State Institute of Pharmaceutical Industry, Shanghai InnoStar Bio-Tech Co., Ltd., Shanghai 201203, China.
| |
Collapse
|
24
|
Kankowski S, Grothe C, Haastert-Talini K. Neuropathic pain: Spotlighting anatomy, experimental models, mechanisms, and therapeutic aspects. Eur J Neurosci 2021; 54:4475-4496. [PMID: 33942412 DOI: 10.1111/ejn.15266] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 04/19/2021] [Accepted: 04/28/2021] [Indexed: 12/30/2022]
Abstract
The International Association for the Study of Pain defines neuropathic pain as "pain arising as a direct consequence of a lesion or disease affecting the somatosensory system". The associated changes can be observed in the peripheral as well as the central nervous system. The available literature discusses a wide variety of causes as predisposing for the development and amplification of neuropathic pain. Further, key interactions within sensory pathways have been discovered, but no common molecular mechanism leading to neuropathic pain has been identified until now. In the first part of this review, the pain mediating lateral spinothalamic tract is described. Different in vivo models are presented that allow studying trauma-, chemotherapy-, virus-, and diabetes-induced neuropathic pain in rodents. We furthermore discuss approaches to assess neuropathic pain in these models. Second, the current knowledge about cellular and molecular mechanisms suggested to underlie the development of neuropathic pain is presented and discussed. A summary of established therapies that are already applied in the clinic and novel, promising approaches closes the paper. In conclusion, the established animal models are able to emulate the diversity of neuropathic pain observed in the clinics. However, the assessment of neuropathic pain in the presented in vivo models should be improved. The determination of common molecular markers with suitable in vitro models would simplify the assessment of neuropathic pain in vivo. This would furthermore provide insights into common molecular mechanisms of the disease and establish a basis to search for satisfying therapeutic approaches.
Collapse
Affiliation(s)
- Svenja Kankowski
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School (MHH), Hannover, Germany
| | - Claudia Grothe
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School (MHH), Hannover, Germany.,Center for Systems Neuroscience (ZNS) Hannover, Hannover, Germany
| | - Kirsten Haastert-Talini
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School (MHH), Hannover, Germany.,Center for Systems Neuroscience (ZNS) Hannover, Hannover, Germany
| |
Collapse
|
25
|
Akin EJ, Alsaloum M, Higerd GP, Liu S, Zhao P, Dib-Hajj FB, Waxman SG, Dib-Hajj SD. Paclitaxel increases axonal localization and vesicular trafficking of Nav1.7. Brain 2021; 144:1727-1737. [PMID: 33734317 PMCID: PMC8320304 DOI: 10.1093/brain/awab113] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 01/09/2021] [Accepted: 03/04/2021] [Indexed: 01/15/2023] Open
Abstract
The microtubule-stabilizing chemotherapy drug paclitaxel (PTX) causes dose-limiting chemotherapy-induced peripheral neuropathy (CIPN), which is often accompanied by pain. Among the multifaceted effects of PTX is an increased expression of sodium channel Nav1.7 in rat and human sensory neurons, enhancing their excitability. However, the mechanisms underlying this increased Nav1.7 expression have not been explored, and the effects of PTX treatment on the dynamics of trafficking and localization of Nav1.7 channels in sensory axons have not been possible to investigate to date. In this study we used a recently developed live imaging approach that allows visualization of Nav1.7 surface channels and long-distance axonal vesicular transport in sensory neurons to fill this basic knowledge gap. We demonstrate concentration and time-dependent effects of PTX on vesicular trafficking and membrane localization of Nav1.7 in real-time in sensory axons. Low concentrations of PTX increase surface channel expression and vesicular flux (number of vesicles per axon). By contrast, treatment with a higher concentration of PTX decreases vesicular flux. Interestingly, vesicular velocity is increased for both concentrations of PTX. Treatment with PTX increased levels of endogenous Nav1.7 mRNA and current density in dorsal root ganglion neurons. However, the current produced by transfection of dorsal root ganglion neurons with Halo-tag Nav1.7 was not increased after exposure to PTX. Taken together, this suggests that the increased trafficking and surface localization of Halo-Nav1.7 that we observed by live imaging in transfected dorsal root ganglion neurons after treatment with PTX might be independent of an increased pool of Nav1.7 channels. After exposure to inflammatory mediators to mimic the inflammatory condition seen during chemotherapy, both Nav1.7 surface levels and vesicular transport are increased for both low and high concentrations of PTX. Overall, our results show that PTX treatment increases levels of functional endogenous Nav1.7 channels in dorsal root ganglion neurons and enhances trafficking and surface distribution of Nav1.7 in sensory axons, with outcomes that depend on the presence of an inflammatory milieu, providing a mechanistic explanation for increased excitability of primary afferents and pain in CIPN.
Collapse
Affiliation(s)
- Elizabeth J Akin
- Department of Neurology, Yale University, New Haven, CT 06510, USA.,Center for Neuroscience and Regeneration Research, Yale University, New Haven, CT 06510, USA.,Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Matthew Alsaloum
- Department of Neurology, Yale University, New Haven, CT 06510, USA.,Center for Neuroscience and Regeneration Research, Yale University, New Haven, CT 06510, USA.,Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA.,MD/PhD Program, Yale University, New Haven, CT 06510, USA
| | - Grant P Higerd
- Department of Neurology, Yale University, New Haven, CT 06510, USA.,Center for Neuroscience and Regeneration Research, Yale University, New Haven, CT 06510, USA.,Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA.,MD/PhD Program, Yale University, New Haven, CT 06510, USA
| | - Shujun Liu
- Department of Neurology, Yale University, New Haven, CT 06510, USA.,Center for Neuroscience and Regeneration Research, Yale University, New Haven, CT 06510, USA.,Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Peng Zhao
- Department of Neurology, Yale University, New Haven, CT 06510, USA.,Center for Neuroscience and Regeneration Research, Yale University, New Haven, CT 06510, USA.,Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Fadia B Dib-Hajj
- Department of Neurology, Yale University, New Haven, CT 06510, USA.,Center for Neuroscience and Regeneration Research, Yale University, New Haven, CT 06510, USA.,Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Stephen G Waxman
- Department of Neurology, Yale University, New Haven, CT 06510, USA.,Center for Neuroscience and Regeneration Research, Yale University, New Haven, CT 06510, USA.,Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Sulayman D Dib-Hajj
- Department of Neurology, Yale University, New Haven, CT 06510, USA.,Center for Neuroscience and Regeneration Research, Yale University, New Haven, CT 06510, USA.,Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
| |
Collapse
|
26
|
Pollard KJ, Bolon B, Moore MJ. Comparative Analysis of Chemotherapy-Induced Peripheral Neuropathy in Bioengineered Sensory Nerve Tissue Distinguishes Mechanistic Differences in Early-Stage Vincristine-, Cisplatin-, and Paclitaxel-Induced Nerve Damage. Toxicol Sci 2021; 180:76-88. [PMID: 33410881 PMCID: PMC7916732 DOI: 10.1093/toxsci/kfaa186] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a well-known, potentially permanent side effect of widely used antineoplastic agents. The mechanisms of neuropathic progression are poorly understood, and the need to test efficacy of novel interventions to treat CIPN continues to grow. Bioengineered microphysiological nerve tissue ("nerve on a chip") has been suggested as an in vitro platform for modeling the structure and physiology of in situ peripheral nerve tissue. Here, we find that length-dependent nerve conduction and histopathologic changes induced by cisplatin, paclitaxel, or vincristine in rat dorsal root ganglion-derived microphysiological sensory nerve tissue recapitulate published descriptions of clinical electrophysiological changes and neuropathologic biopsy findings in test animals and human patients with CIPN. We additionally confirm the postulated link between vincristine-induced axoplasmic transport failure and functional impairment of nerve conduction, the postulated paclitaxel-induced somal toxicity, and identify a potential central role of gliotoxicity in cisplatin-induced sensory neuropathy. Microphysiological CIPN combines the tight experimental control afforded by in vitro experimentation with clinically relevant functional and structural outputs that conventionally require in vivo models. Microphysiological nerve tissue provides a low-cost, high-throughput alternative to conventional nonclinical models for efficiently and effectively investigating lesions, mechanisms, and treatments of CIPN. Neural microphysiological systems are capable of modeling complex neurological disease at the tissue level offering unique advantages over conventional methodology for both testing and generating hypotheses in neurological disease modeling. Impact Statement Recapitulation of distinct hallmarks of clinical CIPN in microphysiological sensory nerve validates a novel peripheral neurotoxicity model with unique advantages over conventional model systems.
Collapse
Affiliation(s)
- Kevin J Pollard
- Department of Biomedical Engineering, Tulane University, New Orleans, Louisiana 70118, USA
| | - Brad Bolon
- GEMpath, Inc, Longmont, Colorado 80504-3711, USA
| | - Michael J Moore
- Department of Biomedical Engineering, Tulane University, New Orleans, Louisiana 70118, USA
- AxoSim, Inc, New Orleans, Louisiana 70112, USA
| |
Collapse
|
27
|
Geisler S. Vincristine- and bortezomib-induced neuropathies - from bedside to bench and back. Exp Neurol 2021; 336:113519. [PMID: 33129841 PMCID: PMC11160556 DOI: 10.1016/j.expneurol.2020.113519] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 10/21/2020] [Accepted: 10/25/2020] [Indexed: 12/11/2022]
Abstract
Vincristine and bortezomib are effective chemotherapeutics widely used to treat hematological cancers. Vincristine blocks tubulin polymerization, whereas bortezomib is a proteasome inhibitor. Despite different mechanisms of action, the main non-hematological side effect of both is peripheral neuropathy that can last long after treatment has ended and cause permanent disability. Many different cellular and animal models of various aspects of vincristine and bortezomib-induced neuropathies have been generated to investigate underlying molecular mechanisms and serve as platforms to develop new therapeutics. These models revealed that bortezomib induces several transcriptional programs in dorsal root ganglia that result in the activation of different neuroinflammatory pathways and secondary central sensitization. In contrast, vincristine has direct toxic effects on the axon, which are accompanied by changes similar to those observed after nerve cut. Axon degeneration following both vincristine and bortezomib is mediated by a phylogenetically ancient, genetically encoded axon destruction program that leads to the activation of the Toll-like receptor adaptor SARM1 (sterile alpha and TIR motif containing protein 1) and local decrease of nicotinamide dinucleotide (NAD+). Here, I describe current in vitro and in vivo models of vincristine- and bortezomib induced neuropathies, present discoveries resulting from these models in the context of clinical findings and discuss how increased understanding of molecular mechanisms underlying different aspects of neuropathies can be translated to effective treatments to prevent, attenuate or reverse vincristine- and bortezomib-induced neuropathies. Such treatments could improve the quality of life of patients both during and after cancer therapy and, accordingly, have enormous societal impact.
Collapse
Affiliation(s)
- Stefanie Geisler
- Department of Neurology, Washington University School of Medicine in St. Louis, MO, USA.
| |
Collapse
|
28
|
Alberti P, Lehmann HC. Chemotherapy induced peripheral neurotoxicity: Six essential articles for effective future research. Exp Neurol 2020; 337:113555. [PMID: 33340795 DOI: 10.1016/j.expneurol.2020.113555] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Paola Alberti
- Experimental Neurology Unit, School of Medicine and Surgery, Monza, Italy; NeuroMI (Milan Center for Neuroscience), Milan, Italy
| | - Helmar C Lehmann
- Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.
| |
Collapse
|
29
|
Antonuzzo A, Bossi P, Cortinovis D. Elapsed time for an unresolved adverse event: systemic anticancer therapy-induced neurotoxicity calls for action. Ann Oncol 2020; 31:1289-1290. [DOI: 10.1016/j.annonc.2020.08.2103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 08/18/2020] [Indexed: 10/23/2022] Open
|
30
|
Schwann Cell Cultures: Biology, Technology and Therapeutics. Cells 2020; 9:cells9081848. [PMID: 32781699 PMCID: PMC7465416 DOI: 10.3390/cells9081848] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 08/01/2020] [Accepted: 08/05/2020] [Indexed: 12/14/2022] Open
Abstract
Schwann cell (SC) cultures from experimental animals and human donors can be prepared using nearly any type of nerve at any stage of maturation to render stage- and patient-specific populations. Methods to isolate, purify, expand in number, and differentiate SCs from adult, postnatal and embryonic sources are efficient and reproducible as these have resulted from accumulated refinements introduced over many decades of work. Albeit some exceptions, SCs can be passaged extensively while maintaining their normal proliferation and differentiation controls. Due to their lineage commitment and strong resistance to tumorigenic transformation, SCs are safe for use in therapeutic approaches in the peripheral and central nervous systems. This review summarizes the evolution of work that led to the robust technologies used today in SC culturing along with the main features of the primary and expanded SCs that make them irreplaceable models to understand SC biology in health and disease. Traditional and emerging approaches in SC culture are discussed in light of their prospective applications. Lastly, some basic assumptions in vitro SC models are identified in an attempt to uncover the combined value of old and new trends in culture protocols and the cellular products that are derived.
Collapse
|