1
|
Altinok FA, Dallali I, Boubekka A, Hasan A, Ozturk Y. Optimized primary dorsal root ganglion cell culture protocol for reliable K + current patch-clamp recordings. Neurosci Lett 2025; 844:138038. [PMID: 39536901 DOI: 10.1016/j.neulet.2024.138038] [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: 09/21/2024] [Revised: 10/25/2024] [Accepted: 11/09/2024] [Indexed: 11/16/2024]
Abstract
DRG primary neuron cultures, derived from rodents, closely mimic properties of sensory neurons in vivo and are highly useful for studying pain and neurological disorders. These cultures are pivotal in patch-clamp electrophysiology for sensory neuron properties analysis. A detailed, replicable protocol in scientific research ensures experiment accuracy and reproducibility. This paper provides comprehensive details for replicating the protocol and achieving consistent results in primary DRG cell culture as used for patch-clamp recordings. We outlined a comprehensive protocol for establishing primary DRG cell culture, optimized for improved gigaseal formation in whole-cell patch-clamp recordings. Additionally, we conducted a simulation study focused on recording macroscopic K+ channels. The findings established an optimized novel protocol that works reliably for whole-cell patch-clamp recordings and data analysis using primary DRG cells prepared as described in this publication. The details for the protocol in the literature are dispersed across various publications, making it challenging to find a comprehensive summary in one source. This study confirms, for the first time, the efficacy of using fewer protocol steps, which reduces stress and variability in obtaining suitable cells for patch-clamp recordings compared to existing methods in the literature. Given the challenges posed by the dissociation process of primary DRG cells and the importance of comprehensive method documentation in the literature, the protocol presented provides improved and consistent applications of primary DRG cell culture in patch-clamp recordings.
Collapse
Affiliation(s)
- Feyza Alyu Altinok
- Department of Pharmacology, Anadolu University Faculty of Pharmacy, 26470 Eskisehir, Turkey.
| | - Ilhem Dallali
- Graduate School of Education, Anadolu University, 26470 Eskisehir, Turkey
| | - Abderaouf Boubekka
- Graduate School of Education, Anadolu University, 26470 Eskisehir, Turkey
| | - Ahmed Hasan
- Graduate School of Education, Anadolu University, 26470 Eskisehir, Turkey
| | - Yusuf Ozturk
- Department of Pharmacology, Istanbul Aydin University Faculty of Pharmacy, 34295 Istanbul, Turkey
| |
Collapse
|
2
|
Zhao H, Guillaud L, Emily MF, Xu X, Moshniaha L, Hanayama H, Kabe R, Terenzio M, Narita A. Nanographene-Based Polymeric Nanoparticles as Near-Infrared Emissive Neuronal Tracers. ACS NANO 2024; 18:34730-34740. [PMID: 39668551 DOI: 10.1021/acsnano.4c10754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2024]
Abstract
Precise tracking of axonal transport is key to deciphering neuronal functions. To achieve long-term imaging at both ultrastructural and macroscopic resolutions, it is critical to develop fluorescent transport tracers with high photostability and biocompatibility. Herein, we report the investigation of nanographene (NG)-based polymeric nanoparticles (NPs) as near-infrared (NIR)-emissive neuronal tracers. Dibenzo[a,m]dinaphtho[3,2,1-ef:1',2',3'-hi]coronene (DBDNC) was employed as the NG, which exhibited a broad NIR emission with a maximum at 711 nm inside the NPs. DBDNC-NPs displayed high photostability and low cytotoxicity, enabling live tracing of retrograde axonal transport in mouse sensory neurons cultured in microfluidic chambers. We also elucidated how DBDNC-NPs undergo retrograde axonal transport following the endolysosomal pathway. This work provides a proof of concept for NIR-emissive, NG-based neuronal tracers with potential for applications in neurobiology.
Collapse
Affiliation(s)
- Hao Zhao
- Organic and Carbon Nanomaterials Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna-son, Kunigami-gun, Okinawa 904-0495, Japan
| | - Laurent Guillaud
- Molecular Neuroscience Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna-son, Kunigami-gun, Okinawa 904-0495, Japan
| | - Maria Fransiska Emily
- Molecular Neuroscience Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna-son, Kunigami-gun, Okinawa 904-0495, Japan
| | - Xiushang Xu
- Organic and Carbon Nanomaterials Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna-son, Kunigami-gun, Okinawa 904-0495, Japan
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Liliia Moshniaha
- Organic Optoelectronics Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna-son, Kunigami-gun, Okinawa 904-0495, Japan
| | - Hiroki Hanayama
- Organic and Carbon Nanomaterials Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna-son, Kunigami-gun, Okinawa 904-0495, Japan
| | - Ryota Kabe
- Organic Optoelectronics Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna-son, Kunigami-gun, Okinawa 904-0495, Japan
| | - Marco Terenzio
- Molecular Neuroscience Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna-son, Kunigami-gun, Okinawa 904-0495, Japan
| | - Akimitsu Narita
- Organic and Carbon Nanomaterials Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna-son, Kunigami-gun, Okinawa 904-0495, Japan
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| |
Collapse
|
3
|
Kheyrollah M, Brandt N, Bräuer AU, Schrader S, Mertsch S. The role of lysophosphatidic acid and its receptors in corneal nerve regeneration. Ocul Surf 2024; 36:10-18. [PMID: 39709127 DOI: 10.1016/j.jtos.2024.12.006] [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: 08/07/2024] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 12/23/2024]
Abstract
The integrity of corneal nerves is critical for ocular surface health, and damages can lead to Neurotrophic Keratopathy (NK). Despite the regenerative abilities of the peripheral nerve system (PNS), corneal nerve regeneration is often incomplete, and the underlying mechanisms are poorly understood. This study aims to identify potential factors that can enhance corneal nerve regeneration for NK treatment, with a focus on Lysophosphatidic acid (LPA). Thus, the effect of LPA and its underlying pathways in nerve regeneration is investigated in detail using in vitro mouse sensory neurons. To elucidate the impact of LPA as well as to reveal the responsible receptor, several functional assays as well as siRNA-based knock-down experiments were conducted. Additionally, possible changes in underlying pathways were investigated on mRNA levels. LPA-treated neurons significantly reduced fiber growth. However, LPAR2 knockdown neurons (Lpar2-KD) following LPA treatment showed a significant increase in fiber length. Additionally, LPA-treated neurons demonstrated enhanced levels of Lpar2 mRNA. On the other hand, nerve regeneration indicators such as Ngf, Gap-43, and Cdc42, along with LPA downstream signaling components like Pi3k and Ras, were elevated in Lpar2-KD neurons. In conclusion, this study elucidates the inhibitory effects of LPA on fiber outgrowth of sensory neurons. Furthermore, LPAR2 was identified as the responsible receptor for the LPA effect. Thus, Lpar2 knockdown might be a promising therapeutic approach to enhance neuronal regeneration in patients with NK.
Collapse
Affiliation(s)
- Maryam Kheyrollah
- Laboratory of Experimental Ophthalmology, Department of Ophthalmology, Pius-Hospital, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Germany; Research Group Anatomy, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Germany
| | - Nicola Brandt
- Research Group Anatomy, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Germany
| | - Anja U Bräuer
- Research Group Anatomy, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Germany; Research Center Neurosensory Science, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Stefan Schrader
- Laboratory of Experimental Ophthalmology, Department of Ophthalmology, Pius-Hospital, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Germany; Research Center Neurosensory Science, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Sonja Mertsch
- Laboratory of Experimental Ophthalmology, Department of Ophthalmology, Pius-Hospital, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Germany.
| |
Collapse
|
4
|
Tusnim J, Kutuzov P, Grasman JM. In Vitro Models for Peripheral Nerve Regeneration. Adv Healthc Mater 2024; 13:e2401605. [PMID: 39324286 DOI: 10.1002/adhm.202401605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 08/14/2024] [Indexed: 09/27/2024]
Abstract
Peripheral nerve injury (PNI) resulting in lesions is highly prevalent clinically, but current therapeutic approaches fail to provide satisfactory outcomes in many patients. While peripheral nerves have intrinsic regenerative capacity, the regenerative capabilities of peripheral nerves are often insufficient to restore full functionality. This highlights an unmet need for developing more effective strategies to repair damaged peripheral nerves and improve regenerative success. Consequently, researchers are actively exploring a variety of therapeutic strategies, encompassing the local delivery of trophic factors or bioactive molecules, the design of advanced biomaterials that interact with regenerating axons, and augmentation with nerve guidance conduits or complex prostheses. However, clinical translation of these technologies remains limited, emphasizing the need for continued research on peripheral nerve regeneration modalities that can enhance functional restoration. Experimental models that accurately recapitulate key aspects of peripheral nerve injury and repair biology can accelerate therapeutic development by enabling systematic testing of new techniques. Advancing regenerative therapies for PNI requires bridging the gap between basic science discoveries and clinical application. This review discusses different in vitro models of peripheral nerve injury and repair, including their advantages, limitations, and potential applications.
Collapse
Affiliation(s)
- Jarin Tusnim
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Peter Kutuzov
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Jonathan M Grasman
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| |
Collapse
|
5
|
McCulloch MK, Mehryab F, Rashnonejad A. Navigating the Landscape of CMT1B: Understanding Genetic Pathways, Disease Models, and Potential Therapeutic Approaches. Int J Mol Sci 2024; 25:9227. [PMID: 39273178 PMCID: PMC11395143 DOI: 10.3390/ijms25179227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 08/13/2024] [Accepted: 08/19/2024] [Indexed: 09/15/2024] Open
Abstract
Charcot-Marie-Tooth type 1B (CMT1B) is a peripheral neuropathy caused by mutations in the gene encoding myelin protein zero (MPZ), a key component of the myelin sheath in Schwann cells. Mutations in the MPZ gene can lead to protein misfolding, unfolded protein response (UPR), endoplasmic reticulum (ER) stress, or protein mistrafficking. Despite significant progress in understanding the disease mechanisms, there is currently no effective treatment for CMT1B, with therapeutic strategies primarily focused on supportive care. Gene therapy represents a promising therapeutic approach for treating CMT1B. To develop a treatment and better design preclinical studies, an in-depth understanding of the pathophysiological mechanisms and animal models is essential. In this review, we present a comprehensive overview of the disease mechanisms, preclinical models, and recent advancements in therapeutic research for CMT1B, while also addressing the existing challenges in the field. This review aims to deepen the understanding of CMT1B and to encourage further research towards the development of effective treatments for CMT1B patients.
Collapse
Affiliation(s)
- Mary Kate McCulloch
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, 575 Children’s Crossroad, Columbus, OH 43215, USA
- Molecular, Cellular, and Developmental Biology Program, The Ohio State University, Columbus, OH 43210, USA
| | - Fatemeh Mehryab
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, 575 Children’s Crossroad, Columbus, OH 43215, USA
| | - Afrooz Rashnonejad
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, 575 Children’s Crossroad, Columbus, OH 43215, USA
- Molecular, Cellular, and Developmental Biology Program, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
6
|
El Chemali L, Boutary S, Liu S, Liu GJ, Middleton RJ, Banati RB, Bahrenberg G, Rupprecht R, Schumacher M, Massaad-Massade L. GRT-X Stimulates Dorsal Root Ganglia Axonal Growth in Culture via TSPO and Kv7.2/3 Potassium Channel Activation. Int J Mol Sci 2024; 25:7327. [PMID: 39000434 PMCID: PMC11242890 DOI: 10.3390/ijms25137327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 06/27/2024] [Accepted: 06/30/2024] [Indexed: 07/16/2024] Open
Abstract
GRT-X, which targets both the mitochondrial translocator protein (TSPO) and the Kv7.2/3 (KCNQ2/3) potassium channels, has been shown to efficiently promote recovery from cervical spine injury. In the present work, we investigate the role of GRT-X and its two targets in the axonal growth of dorsal root ganglion (DRG) neurons. Neurite outgrowth was quantified in DRG explant cultures prepared from wild-type C57BL6/J and TSPO-KO mice. TSPO was pharmacologically targeted with the agonist XBD173 and the Kv7 channels with the activator ICA-27243 and the inhibitor XE991. GRT-X efficiently stimulated DRG axonal growth at 4 and 8 days after its single administration. XBD173 also promoted axonal elongation, but only after 8 days and its repeated administration. In contrast, both ICA27243 and XE991 tended to decrease axonal elongation. In dissociated DRG neuron/Schwann cell co-cultures, GRT-X upregulated the expression of genes associated with axonal growth and myelination. In the TSPO-KO DRG cultures, the stimulatory effect of GRT-X on axonal growth was completely lost. However, GRT-X and XBD173 activated neuronal and Schwann cell gene expression after TSPO knockout, indicating the presence of additional targets warranting further investigation. These findings uncover a key role of the dual mode of action of GRT-X in the axonal elongation of DRG neurons.
Collapse
Affiliation(s)
- Léa El Chemali
- Maladies et Hormones du Système Nerveux, Inserm, Université Paris-Saclay, 94276 Le Kremlin-Bicêtre, France
| | - Suzan Boutary
- Maladies et Hormones du Système Nerveux, Inserm, Université Paris-Saclay, 94276 Le Kremlin-Bicêtre, France
| | - Song Liu
- Maladies et Hormones du Système Nerveux, Inserm, Université Paris-Saclay, 94276 Le Kremlin-Bicêtre, France
| | - Guo-Jun Liu
- Australian Nuclear Science and Technology Organisation (ANSTO), Kirrawee, NSW 2232, Australia
- Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2006, Australia
| | - Ryan J Middleton
- Australian Nuclear Science and Technology Organisation (ANSTO), Kirrawee, NSW 2232, Australia
| | - Richard B Banati
- Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2006, Australia
| | - Gregor Bahrenberg
- Global Preclinical R&D, Grünenthal Innovation, Grünenthal GmbH, Zieglerstraße 6, D-52078 Aachen, Germany
| | - Rainer Rupprecht
- Department of Psychiatry and Psychotherapy, University of Regensburg, D-93053 Regensburg, Germany
| | - Michael Schumacher
- Maladies et Hormones du Système Nerveux, Inserm, Université Paris-Saclay, 94276 Le Kremlin-Bicêtre, France
| | - Liliane Massaad-Massade
- Maladies et Hormones du Système Nerveux, Inserm, Université Paris-Saclay, 94276 Le Kremlin-Bicêtre, France
| |
Collapse
|
7
|
Lee FS, Nguyen UN, Munns EJ, Wachs RA. Identification of compounds that cause axonal dieback without cytotoxicity in dorsal root ganglia explants and intervertebral disc cells with potential to treat pain via denervation. PLoS One 2024; 19:e0300254. [PMID: 38696450 PMCID: PMC11065314 DOI: 10.1371/journal.pone.0300254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 02/23/2024] [Indexed: 05/04/2024] Open
Abstract
Low back pain, knee osteoarthritis, and cancer patients suffer from chronic pain. Aberrant nerve growth into intervertebral disc, knee, and tumors, are common pathologies that lead to these chronic pain conditions. Axonal dieback induced by capsaicin (Caps) denervation has been FDA-approved to treat painful neuropathies and knee osteoarthritis but with short-term efficacy and discomfort. Herein, we propose to evaluate pyridoxine (Pyr), vincristine sulfate (Vcr) and ionomycin (Imy) as axonal dieback compounds for denervation with potential to alleviate pain. Previous literature suggests Pyr, Vcr, and Imy can cause undesired axonal degeneration, but no previous work has evaluated axonal dieback and cytotoxicity on adult rat dorsal root ganglia (DRG) explants. Thus, we performed axonal dieback screening using adult rat DRG explants in vitro with Caps as a positive control and assessed cytotoxicity. Imy inhibited axonal outgrowth and slowed axonal dieback, while Pyr and Vcr at high concentrations produced significant reduction in axon length and robust axonal dieback within three days. DRGs treated with Caps, Vcr, or Imy had increased DRG cytotoxicity compared to matched controls, but overall cytotoxicity was minimal and at least 88% lower compared to lysed DRGs. Pyr did not lead to any DRG cytotoxicity. Further, neither Pyr nor Vcr triggered intervertebral disc cell death or affected cellular metabolic activity after three days of incubation in vitro. Overall, our findings suggest Pyr and Vcr are not toxic to DRGs and intervertebral disc cells, and there is potential for repurposing these compounds for axonal dieback compounds to cause local denervation and alleviate pain.
Collapse
Affiliation(s)
- Fei San Lee
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska United States of America
| | - Uyen N. Nguyen
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska United States of America
| | - Eliza J. Munns
- Department of Electrical, Computer, and Biomedical Engineering, Union College, Schenectady, New York, United States of America
| | - Rebecca A. Wachs
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, Nebraska United States of America
| |
Collapse
|
8
|
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
|
9
|
Elfarnawany A, Dehghani F. Time- and Concentration-Dependent Adverse Effects of Paclitaxel on Non-Neuronal Cells in Rat Primary Dorsal Root Ganglia. TOXICS 2023; 11:581. [PMID: 37505547 PMCID: PMC10385404 DOI: 10.3390/toxics11070581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/27/2023] [Accepted: 07/01/2023] [Indexed: 07/29/2023]
Abstract
Paclitaxel is a chemotherapeutic agent used to treat a wide range of malignant tumors. Although it has anti-tumoral properties, paclitaxel also shows significant adverse effects on the peripheral nervous system, causing peripheral neuropathy. Paclitaxel has previously been shown to exert direct neurotoxic effects on primary DRG neurons. However, little is known about paclitaxel's effects on non-neuronal DRG cells. They provide mechanical and metabolic support and influence neuronal signaling. In the present study, paclitaxel effects on primary DRG non-neuronal cells were analyzed and their concentration or/and time dependence investigated. DRGs of Wister rats (6-8 weeks old) were isolated, and non-neuronal cell populations were separated by the density gradient centrifugation method. Different concentrations of Paclitaxel (0.01 µM-10 µM) were tested on cell viability by MTT assay, cell death by lactate dehydrogenase (LDH) assay, and propidium iodide (PI) assay, as well as cell proliferation by Bromodeoxyuridine (BrdU) assay at 24 h, 48 h, and 72 h post-treatment. Furthermore, phenotypic effects have been investigated by using immunofluorescence techniques. Paclitaxel exhibited several toxicological effects on non-neuronal cells, including a reduction in cell viability, an increase in cell death, and an inhibition of cell proliferation. These effects were concentration- and time-dependent. Cellular and nuclear changes such as shrinkage, swelling of cell bodies, nuclear condensation, chromatin fragmentation, retraction, and a loss in processes were observed. Paclitaxel showed adverse effects on primary DRG non-neuronal cells, which might have adverse functional consequences on sensory neurons of the DRG, asking for consideration in the management of peripheral neuropathy.
Collapse
Affiliation(s)
- Amira Elfarnawany
- Department of Anatomy and Cell Biology, Medical Faculty, Martin Luther University Halle-Wittenberg, Grosse Steinstrasse 52, 06108 Halle (Saale), Germany;
- Zoology Department, Faculty of Science, Tanta University, Tanta 31527, Egypt
| | - Faramarz Dehghani
- Department of Anatomy and Cell Biology, Medical Faculty, Martin Luther University Halle-Wittenberg, Grosse Steinstrasse 52, 06108 Halle (Saale), Germany;
| |
Collapse
|
10
|
Gong GQ, Bilanges B, Allsop B, Masson GR, Roberton V, Askwith T, Oxenford S, Madsen RR, Conduit SE, Bellini D, Fitzek M, Collier M, Najam O, He Z, Wahab B, McLaughlin SH, Chan AWE, Feierberg I, Madin A, Morelli D, Bhamra A, Vinciauskaite V, Anderson KE, Surinova S, Pinotsis N, Lopez-Guadamillas E, Wilcox M, Hooper A, Patel C, Whitehead MA, Bunney TD, Stephens LR, Hawkins PT, Katan M, Yellon DM, Davidson SM, Smith DM, Phillips JB, Angell R, Williams RL, Vanhaesebroeck B. A small-molecule PI3Kα activator for cardioprotection and neuroregeneration. Nature 2023; 618:159-168. [PMID: 37225977 PMCID: PMC7614683 DOI: 10.1038/s41586-023-05972-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 03/17/2023] [Indexed: 05/26/2023]
Abstract
Harnessing the potential beneficial effects of kinase signalling through the generation of direct kinase activators remains an underexplored area of drug development1-5. This also applies to the PI3K signalling pathway, which has been extensively targeted by inhibitors for conditions with PI3K overactivation, such as cancer and immune dysregulation. Here we report the discovery of UCL-TRO-1938 (referred to as 1938 hereon), a small-molecule activator of the PI3Kα isoform, a crucial effector of growth factor signalling. 1938 allosterically activates PI3Kα through a distinct mechanism by enhancing multiple steps of the PI3Kα catalytic cycle and causes both local and global conformational changes in the PI3Kα structure. This compound is selective for PI3Kα over other PI3K isoforms and multiple protein and lipid kinases. It transiently activates PI3K signalling in all rodent and human cells tested, resulting in cellular responses such as proliferation and neurite outgrowth. In rodent models, acute treatment with 1938 provides cardioprotection from ischaemia-reperfusion injury and, after local administration, enhances nerve regeneration following nerve crush. This study identifies a chemical tool to directly probe the PI3Kα signalling pathway and a new approach to modulate PI3K activity, widening the therapeutic potential of targeting these enzymes through short-term activation for tissue protection and regeneration. Our findings illustrate the potential of activating kinases for therapeutic benefit, a currently largely untapped area of drug development.
Collapse
Affiliation(s)
- Grace Q Gong
- Cell Signalling, Cancer Institute, University College London, London, UK
| | - Benoit Bilanges
- Cell Signalling, Cancer Institute, University College London, London, UK
| | - Ben Allsop
- Drug Discovery Group, Translational Research Office, University College London, London, UK
| | - Glenn R Masson
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
- Division of Cellular Medicine, School of Medicine, University of Dundee, Dundee, UK
| | - Victoria Roberton
- UCL Centre for Nerve Engineering, UCL School of Pharmacy, University College London, London, UK
| | - Trevor Askwith
- Drug Discovery Group, Translational Research Office, University College London, London, UK
| | - Sally Oxenford
- Drug Discovery Group, Translational Research Office, University College London, London, UK
| | - Ralitsa R Madsen
- Cell Signalling, Cancer Institute, University College London, London, UK
| | - Sarah E Conduit
- Cell Signalling, Cancer Institute, University College London, London, UK
| | - Dom Bellini
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | - Martina Fitzek
- Hit Discovery, Discovery Sciences, R&D, AstraZeneca, Alderley Park, Macclesfield, UK
| | - Matt Collier
- Hit Discovery, Discovery Sciences, R&D, AstraZeneca, Alderley Park, Macclesfield, UK
| | - Osman Najam
- The Hatter Cardiovascular Institute, University College London, London, UK
| | - Zhenhe He
- The Hatter Cardiovascular Institute, University College London, London, UK
| | - Ben Wahab
- Medicines Discovery Institute, School of Biosciences, Cardiff University, Cardiff, UK
| | | | - A W Edith Chan
- Wolfson Institute for Biomedical Research, University College London, London, UK
| | | | - Andrew Madin
- Hit Discovery, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Daniele Morelli
- Cell Signalling, Cancer Institute, University College London, London, UK
| | - Amandeep Bhamra
- Proteomics Research Translational Technology Platform, Cancer Institute, University College London, London, UK
| | - Vanesa Vinciauskaite
- Division of Cellular Medicine, School of Medicine, University of Dundee, Dundee, UK
| | | | - Silvia Surinova
- Proteomics Research Translational Technology Platform, Cancer Institute, University College London, London, UK
| | - Nikos Pinotsis
- Institute of Structural and Molecular Biology, Birkbeck College, London, UK
| | | | - Matthew Wilcox
- UCL Centre for Nerve Engineering, UCL School of Pharmacy, University College London, London, UK
| | - Alice Hooper
- Drug Discovery Group, Translational Research Office, University College London, London, UK
| | - Chandni Patel
- Drug Discovery Group, Translational Research Office, University College London, London, UK
| | - Maria A Whitehead
- Cell Signalling, Cancer Institute, University College London, London, UK
| | - Tom D Bunney
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London, UK
| | | | | | - Matilda Katan
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London, UK
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, University College London, London, UK
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, London, UK
| | - David M Smith
- Emerging Innovations, Discovery Sciences, R&D, AstraZeneca, Cambridge, UK
| | - James B Phillips
- UCL Centre for Nerve Engineering, UCL School of Pharmacy, University College London, London, UK
| | - Richard Angell
- Drug Discovery Group, Translational Research Office, University College London, London, UK
- Medicines Discovery Institute, School of Biosciences, Cardiff University, Cardiff, UK
| | - Roger L Williams
- Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | | |
Collapse
|
11
|
3D culture of the spinal cord with roots as an ex vivo model for comparative studies of motor and sensory nerve regeneration. Exp Neurol 2023; 362:114322. [PMID: 36652972 DOI: 10.1016/j.expneurol.2023.114322] [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: 09/28/2022] [Revised: 12/21/2022] [Accepted: 01/13/2023] [Indexed: 01/18/2023]
Abstract
Motor and sensory nerves exhibit tissue-specific structural and functional features. However, in vitro models designed to reflect tissue-specific differences between motor and sensory nerve regeneration have rarely been reported. Here, by embedding the spinal cord with roots (SCWR) in a 3D hydrogel environment, we compared the nerve regeneration processes between the ventral and dorsal roots. The 3D hydrogel environment induced an outward migration of neurons in the gray matter of the spinal cord, which allowed the long-term survival of motor neurons. Tuj1 immunofluorescence labeling confirmed the regeneration of neurites from both the ventral and dorsal roots. Next, we detected asymmetric ventral and dorsal root regeneration in response to nerve growth factor (NGF) and glial cell line-derived neurotrophic factor (GDNF), and we observed motor and sensory Schwann cell phenotypes in the regenerated ventral and dorsal roots, respectively. Moreover, based on the SCWR model, we identified a targeted effect of collagen VI on sensory nerve fasciculation and characterized the protein expression profiles correlating to motor/sensory-specific nerve regeneration. These results suggest that the SCWR model can serve as a valuable ex vivo model for comparative study of motor and sensory nerve regeneration and for pharmacodynamic evaluations.
Collapse
|
12
|
Kalkan ÖF, Aktaş O, Sürmeneli YE, Alver A, Özcan M, Şahin Z. Does irisin has neuroprotective effect against diabetes induced neuropathy in male rats? Arch Physiol Biochem 2023; 129:439-448. [PMID: 33141621 DOI: 10.1080/13813455.2020.1835985] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
We aimed to investigate the contribution of irisin in the neuroprotective process of exercise training in diabetic rats. Serum irisin levels, thermal and mechanical pain thresholds and intracellular calcium ([Ca2+]i) levels in sensory neurons were measured at different time intervals during the eight weeks of exercise sessions for the control, non-exercise diabetics (3 groups) and exercise performing (low and high intensity groups) diabetic rats (n = 7-10 for all groups). Non-exercise diabetic groups were treated with irisin in different doses (1, 10 and 20 µg/kg respectively). Recovered pain thresholds at the end of the exercise sessions (p < .05), higher serum irisin levels that compared to control and diabetics (p < .05) and insignificant mean [Ca2+]i peak amplitudes in sensory neurons (p > .05) obtained from experiments. Furthermore, irisin injection decreased the thermal pain threshold of diabetics only at 60th minutes (p < .05). Irisin may have a role in the neuroprotective effect of exercise training.
Collapse
Affiliation(s)
- Ömer Faruk Kalkan
- Faculty of Medicine, Department of Physiology, Karadeniz Technical University, Trabzon, Turkey
| | - Osman Aktaş
- Faculty of Medicine, Department of Physiology, Karadeniz Technical University, Trabzon, Turkey
| | - Yunus Emre Sürmeneli
- Faculty of Medicine, Department of Physiology, University of Health Sciences, Istanbul, Turkey
| | - Ahmet Alver
- Faculty of Medicine, Department of Biochemistry, Karadeniz Technical University, Trabzon, Turkey
| | - Mete Özcan
- Faculty of Medicine, Department of Biophysics, Firat University, Elazig, Turkey
| | - Zafer Şahin
- Faculty of Medicine, Department of Physiology, Karadeniz Technical University, Trabzon, Turkey
| |
Collapse
|
13
|
Enamorado M, Kulalert W, Han SJ, Rao I, Delaleu J, Link VM, Yong D, Smelkinson M, Gil L, Nakajima S, Linehan JL, Bouladoux N, Wlaschin J, Kabat J, Kamenyeva O, Deng L, Gribonika I, Chesler AT, Chiu IM, Le Pichon CE, Belkaid Y. Immunity to the microbiota promotes sensory neuron regeneration. Cell 2023; 186:607-620.e17. [PMID: 36640762 PMCID: PMC11512587 DOI: 10.1016/j.cell.2022.12.037] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 11/11/2022] [Accepted: 12/20/2022] [Indexed: 01/15/2023]
Abstract
Tissue immunity and responses to injury depend on the coordinated action and communication among physiological systems. Here, we show that, upon injury, adaptive responses to the microbiota directly promote sensory neuron regeneration. At homeostasis, tissue-resident commensal-specific T cells colocalize with sensory nerve fibers within the dermis, express a transcriptional program associated with neuronal interaction and repair, and promote axon growth and local nerve regeneration following injury. Mechanistically, our data reveal that the cytokine interleukin-17A (IL-17A) released by commensal-specific Th17 cells upon injury directly signals to sensory neurons via IL-17 receptor A, the transcription of which is specifically upregulated in injured neurons. Collectively, our work reveals that in the context of tissue damage, preemptive immunity to the microbiota can rapidly bridge biological systems by directly promoting neuronal repair, while also identifying IL-17A as a major determinant of this fundamental process.
Collapse
Affiliation(s)
- Michel Enamorado
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Warakorn Kulalert
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Seong-Ji Han
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Indira Rao
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; Immunology Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jérémie Delaleu
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Verena M Link
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Daniel Yong
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Margery Smelkinson
- Biological Imaging, Research Technology Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Louis Gil
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Saeko Nakajima
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jonathan L Linehan
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nicolas Bouladoux
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Josette Wlaschin
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Juraj Kabat
- Biological Imaging, Research Technology Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Olena Kamenyeva
- Biological Imaging, Research Technology Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Liwen Deng
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Inta Gribonika
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Alexander T Chesler
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Isaac M Chiu
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Claire E Le Pichon
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yasmine Belkaid
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; NIAID Microbiome Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
14
|
Agrawal L, Vimal SK, Barzaghi P, Shiga T, Terenzio M. Biodegradable and Electrically Conductive Melanin-Poly (3-Hydroxybutyrate) 3D Fibrous Scaffolds for Neural Tissue Engineering Applications. Macromol Biosci 2022; 22:e2200315. [PMID: 36114714 DOI: 10.1002/mabi.202200315] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Indexed: 01/15/2023]
Abstract
Due to the severity of peripheral nerve injuries (PNI) and spinal cord injuries (SCI), treatment options for patients are limited. In this context, biomaterials designed to promote regeneration and reinstate the lost function are being explored. Such biomaterials should be able to mimic the biological, chemical, and physical cues of the extracellular matrix for maximum effectiveness as therapeutic agents. Development of biomaterials with desirable physical, chemical, and electrical properties, however, has proven challenging. Here a novel biomaterial formulation achieved by blending the pigment melanin and the natural polymer Poly-3-hydroxybutyrate (PHB) is proposed. Physio-chemical measurements of electrospun fibers reveal a feature rich surface nano-topography, a semiconducting-nature, and brain-tissue-like poroviscoelastic properties. Resulting fibers improve cell adhesion and growth of mouse sensory and motor neurons, without any observable toxicity. Further, the presence of polar functional groups positively affect the kinetics of fibers degradation at a pH (≈7.4) comparable to that of body fluids. Thus, melanin-PHB blended scaffolds are found to be physio-chemically, electrically, and biologically compatible with neural tissues and could be used as a regenerative modality for neural tissue injuries. A biomaterial for scaffolds intended to promote regeneration of nerve tissue after injury is developed. This biomaterial, obtained by mixing the pigment melanin and the natural polymer PHB, is biodegradable, electrically conductive, and beneficial to the growth of motor and sensory neurons. Thus, it is believed that this biomaterial can be used in the context of healthcare applications.
Collapse
Affiliation(s)
- Lokesh Agrawal
- Molecular Neuroscience Unit, Okinawa Institute of Science and Technology Graduate University, Kunigami-gun, Okinawa, 904-0412, Japan.,Graduate School of Comprehensive Human Sciences Kansei, Behavioral and Brain Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, 305-8577, Japan
| | - Sunil Kumar Vimal
- Department of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, P. R. China.,Universidad Integral del Caribe y América Latina, Kaminda Cas Grandi #79, Willemstad, Curacao
| | - Paolo Barzaghi
- Scientific Imaging Section, Okinawa Institute of Science and Technology Graduate University, Kunigami-gun, Okinawa, 904-0412, Japan
| | - Takashi Shiga
- Graduate School of Comprehensive Human Sciences Kansei, Behavioral and Brain Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, 305-8577, Japan.,Department of Neurobiology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, 305-8577, Japan
| | - Marco Terenzio
- Molecular Neuroscience Unit, Okinawa Institute of Science and Technology Graduate University, Kunigami-gun, Okinawa, 904-0412, Japan
| |
Collapse
|
15
|
Cisplatin-induced changes in calcitonin gene-related peptide or TNF-α release in rat dorsal root ganglia in vitro model of neurotoxicity are not reverted by rosiglitazone. Neurotoxicology 2022; 93:211-221. [DOI: 10.1016/j.neuro.2022.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 09/24/2022] [Accepted: 10/05/2022] [Indexed: 11/05/2022]
|
16
|
Xian F, Sondermann JR, Gomez Varela D, Schmidt M. Deep proteome profiling reveals signatures of age and sex differences in paw skin and sciatic nerve of naïve mice. eLife 2022; 11:e81431. [PMID: 36448997 PMCID: PMC9711526 DOI: 10.7554/elife.81431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 11/16/2022] [Indexed: 12/03/2022] Open
Abstract
The age and sex of studied animals profoundly impact experimental outcomes in biomedical research. However, most preclinical studies in mice use a wide-spanning age range from 4 to 20 weeks and do not assess male and female mice in parallel. This raises concerns regarding reproducibility and neglects potentially relevant age and sex differences, which are largely unknown at the molecular level in naïve mice. Here, we employed an optimized quantitative proteomics workflow in order to deeply profile mouse paw skin and sciatic nerves (SCN) - two tissues implicated in nociception and pain as well as diseases linked to inflammation, injury, and demyelination. Remarkably, we uncovered significant differences when comparing male and female mice at adolescent (4 weeks) and adult (14 weeks) age. Our analysis deciphered protein subsets and networks that were correlated with the age and/or sex of mice. Notably, among these were proteins/biological pathways with known (patho)physiological relevance, e.g., homeostasis and epidermal signaling in skin, and, in SCN, multiple myelin proteins and regulators of neuronal development. Extensive comparisons with available databases revealed that various proteins associated with distinct skin diseases and pain exhibited significant abundance changes in dependence on age and/or sex. Taken together, our study uncovers hitherto unknown sex and age differences at the level of proteins and protein networks. Overall, we provide a unique proteome resource that facilitates mechanistic insights into somatosensory and skin biology, and integrates age and sex as biological variables - a prerequisite for successful preclinical studies in mouse disease models.
Collapse
Affiliation(s)
- Feng Xian
- Systems Biology of Pain, Division of Pharmacology & Toxicology, Department of Pharmaceutical Sciences, Faculty of Life Sciences, University of ViennaViennaAustria
| | - Julia Regina Sondermann
- Systems Biology of Pain, Division of Pharmacology & Toxicology, Department of Pharmaceutical Sciences, Faculty of Life Sciences, University of ViennaViennaAustria
| | - David Gomez Varela
- Systems Biology of Pain, Division of Pharmacology & Toxicology, Department of Pharmaceutical Sciences, Faculty of Life Sciences, University of ViennaViennaAustria
| | - Manuela Schmidt
- Systems Biology of Pain, Division of Pharmacology & Toxicology, Department of Pharmaceutical Sciences, Faculty of Life Sciences, University of ViennaViennaAustria
| |
Collapse
|
17
|
Bigsby S, Neapetung J, Campanucci VA. Voltage-gated sodium channels in diabetic sensory neuropathy: Function, modulation, and therapeutic potential. Front Cell Neurosci 2022; 16:994585. [PMID: 36467605 PMCID: PMC9713017 DOI: 10.3389/fncel.2022.994585] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 10/11/2022] [Indexed: 10/29/2023] Open
Abstract
Voltage-gated sodium channels (Na V ) are the main contributors to action potential generation and essential players in establishing neuronal excitability. Na V channels have been widely studied in pain pathologies, including those that develop during diabetes. Diabetic sensory neuropathy (DSN) is one of the most common complications of the disease. DSN is the result of sensory nerve damage by the hyperglycemic state, resulting in a number of debilitating symptoms that have a significant negative impact in the quality of life of diabetic patients. Among those symptoms are tingling and numbness of hands and feet, as well as exacerbated pain responses to noxious and non-noxious stimuli. DSN is also a major contributor to the development of diabetic foot, which may lead to lower limb amputations in long-term diabetic patients. Unfortunately, current treatments fail to reverse or successfully manage DSN. In the current review we provide an updated report on Na V channels including structure/function and contribution to DSN. Furthermore, we summarize current research on the therapeutic potential of targeting Na V channels in pain pathologies, including DSN.
Collapse
Affiliation(s)
| | | | - Verónica A. Campanucci
- Department of Anatomy, Physiology and Pharmacology (APP), College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
18
|
Intisar A, Shin HY, Kim W, Kang HG, Kim MY, Kim YS, Cho Y, Mo YJ, Lim H, Lee S, Lu QR, Lee Y, Kim MS. Implantable Electroceutical Approach Improves Myelination by Restoring Membrane Integrity in a Mouse Model of Peripheral Demyelinating Neuropathy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2201358. [PMID: 35975427 PMCID: PMC9661852 DOI: 10.1002/advs.202201358] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 07/22/2022] [Indexed: 06/15/2023]
Abstract
Although many efforts are undertaken to treat peripheral demyelinating neuropathies based on biochemical interventions, unfortunately, there is no approved treatment yet. Furthermore, previous studies have not shown improvement of the myelin membrane at the biomolecular level. Here, an electroceutical treatment is introduced as a biophysical intervention to treat Charcot-Marie-Tooth (CMT) disease-the most prevalent peripheral demyelinating neuropathy worldwide-using a mouse model. The specific electrical stimulation (ES) condition (50 mV mm-1 , 20 Hz, 1 h) for optimal myelination is found via an in vitro ES screening system, and its promyelinating effect is validated with ex vivo dorsal root ganglion model. Biomolecular investigation via time-of-flight secondary ion mass spectrometry shows that ES ameliorates distribution abnormalities of peripheral myelin protein 22 and cholesterol in the myelin membrane, revealing the restoration of myelin membrane integrity. ES intervention in vivo via flexible implantable electrodes shows not only gradual rehabilitation of mouse behavioral phenotypes (balance and endurance), but also restored myelin thickness, compactness, and membrane integrity. This study demonstrates, for the first time, that an electroceutical approach with the optimal ES condition has the potential to treat CMT disease and restore impaired myelin membrane integrity, shifting the paradigm toward practical interventions for peripheral demyelinating neuropathies.
Collapse
Affiliation(s)
- Aseer Intisar
- Department of New BiologyDGISTDaegu42988Republic of Korea
| | - Hyun Young Shin
- CTCELLS Corp.Daegu42988Republic of Korea
- SBCure Corp.Daegu43017Republic of Korea
| | | | - Hyun Gyu Kang
- Department of New BiologyDGISTDaegu42988Republic of Korea
| | - Min Young Kim
- Department of New BiologyDGISTDaegu42988Republic of Korea
| | - Yu Seon Kim
- Well Aging Research CenterDGISTDaegu42988Republic of Korea
| | - Youngjun Cho
- Department of Robotics and Mechatronics EngineeringDGISTDaegu42988Republic of Korea
| | - Yun Jeoung Mo
- Well Aging Research CenterDGISTDaegu42988Republic of Korea
| | - Heejin Lim
- Department of New BiologyDGISTDaegu42988Republic of Korea
| | - Sanghoon Lee
- Department of Robotics and Mechatronics EngineeringDGISTDaegu42988Republic of Korea
| | - Q. Richard Lu
- Department of PediatricsCincinnati Children's Hospital Medical CenterCincinnatiOH45229USA
| | - Yun‐Il Lee
- Well Aging Research CenterDGISTDaegu42988Republic of Korea
| | - Minseok S. Kim
- Department of New BiologyDGISTDaegu42988Republic of Korea
- CTCELLS Corp.Daegu42988Republic of Korea
- Translational Responsive Medicine Center (TRMC)DGISTDaegu42988Republic of Korea
- New Biology Research Center (NBRC)DGISTDaegu42988Republic of Korea
| |
Collapse
|
19
|
Generation of an in vitro model for peripheral neuropathy in Fabry disease using CRISPR-Cas9 in the nociceptive dorsal root ganglion cell line 50B11. Mol Genet Metab Rep 2022; 31:100871. [PMID: 35782611 PMCID: PMC9248215 DOI: 10.1016/j.ymgmr.2022.100871] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 04/14/2022] [Indexed: 11/20/2022] Open
Abstract
Fabry disease is a glycosphingolipid storage disorder that is caused by a genetic deficiency of the lysosomal enzyme alpha-galactosidase A (AGA, EC 3.2.1.22). As a result, the glycolipid substrate, globotriaosylceramide (Gb3) accumulates in various cell types throughout the body producing a multisystem disease that affects the vascular, cardiac, renal, and nervous systems. A hallmark of this disorder is neuropathic pain that occurs in up to 80% of Fabry patients and has been characterized as a small fiber neuropathy. The molecular mechanism by which changes in AGA activity produce neuropathic pain is not clear, in part due to a lack of relevant model systems. Using 50B11 cells, an immortalized dorsal root ganglion neuron with nociceptive characteristics derived from rat, we used CRISPR-Cas9 gene editing of the galactosidase alpha (GLA) gene for AGA to create two stable knock-out clones that have the phenotypic characteristics of Fabry cells. The cell lines show severely reduced lysosomal AGA activity in homogenates as well as impaired degradation of Gb3 in cultured cells. This phenotype is stable over long-term culture. Similar to the unedited 50B11 cell line, the clones differentiate in response to forskolin and extend neurites. Flow cytometry experiments demonstrate that the gene-edited cells express TRPV1 pain receptor at increased levels compared to control, suggesting a possible mechanism for increased pain sensitization in Fabry patients. Our 50B11 cell lines show phenotypic characteristics of Fabry disease and grow well under standard cell culture conditions. These cell lines can provide a convenient model system to help elucidate the molecular mechanism of pain in Fabry patients.
Collapse
|
20
|
Emily MF, Agrawal L, Barzaghi P, Otsuki M, Terenzio M. Use of Microfluidics Chambers to Image Axonal transport in Adult Sensory Neurons. Methods Mol Biol 2022; 2431:271-288. [PMID: 35412282 DOI: 10.1007/978-1-0716-1990-2_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Transport of cargoes along axons is crucial for ensuring effective neuronal function and survival. Lysosomes, which are membrane-bound organelles responsible for the degradation of macromolecules, are among the many cargoes being transported. Compartmentalized systems that allow for the separation of the somatic compartment from the axonal network, are widely used in the field of neurobiology and in the study of axonal transport in particular. Among the various solutions available, microfluidics chambers that take advantage of fluidic separation between different compartments, have seen widespread adoption. Said chambers are made of polydimethylsiloxane (PDMS), a transparent, gas permeable compound, which is compatible with fluorescence microscopy, and have significantly positively impacted cellular neuroscience, drastically increasing our understanding of axonal peripheral signaling. Here we describe a two-layered microfluidics chamber, engineered to allow for the culture of adult sensory neurons. This device was designed to promote the proper placement of adult sensory neurons in the somatic chamber in proximity of the microgrooves. We detail the production of the master mold, how to fabricate and assemble the device and how to disaggregate and load the cells in it. In addition, we provide details on how to conduct and analyze an axonal transport experiment using a custom made script in MATLAB designed by our laboratory.
Collapse
Affiliation(s)
- Maria Fransiska Emily
- Molecular Neuroscience Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Lokesh Agrawal
- Molecular Neuroscience Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Paolo Barzaghi
- Imaging Section, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Miki Otsuki
- Molecular Neuroscience Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Marco Terenzio
- Molecular Neuroscience Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan.
| |
Collapse
|
21
|
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
|
22
|
NeuriteNet: A convolutional neural network for assessing morphological parameters of neurite growth. J Neurosci Methods 2021; 363:109349. [PMID: 34480956 DOI: 10.1016/j.jneumeth.2021.109349] [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: 05/27/2021] [Revised: 08/19/2021] [Accepted: 08/30/2021] [Indexed: 11/24/2022]
Abstract
BACKGROUND During development or regeneration, neurons extend processes (i.e., neurites) via mechanisms that can be readily analyzed in culture. However, defining the impact of a drug or genetic manipulation on such mechanisms can be challenging due to the complex arborization and heterogeneous patterns of neurite growth in vitro. New Method: NeuriteNet is a Convolutional Neural Network (CNN) sorting model that uses a novel adaptation of the XRAI saliency map overlay, which is a region-based attribution method. NeuriteNet compares neuronal populations based on differences in neurite growth patterns, sorts them into respective groups, and overlays a saliency map indicating which areas differentiated the image for the sorting procedure. RESULTS In this study, we demonstrate that NeuriteNet effectively sorts images corresponding to dissociated neurons into control and treatment groups according to known morphological differences. Furthermore, the saliency map overlay highlights the distinguishing features of the neuron when sorting the images into treatment groups. NeuriteNet also identifies novel morphological differences in neurons cultured from control and genetically modified mouse strains. Comparison with Existing Methods: Unlike other neurite analysis platforms, NeuriteNet does not require manual manipulations, such as segmentation of neurites prior to analysis, and is more accurate than experienced researchers for categorizing neurons according to their pattern of neurite growth. CONCLUSIONS NeuriteNet can be used to effectively screen for morphological differences in a heterogeneous group of neurons and to provide feedback on the key features distinguishing those groups via the saliency map overlay.
Collapse
|
23
|
Rogachevskii IV, Plakhova VB, Penniyaynen VA, Terekhin SG, Podzorova SA, Krylov BV. New approaches to the design of analgesic medicinal substances. Can J Physiol Pharmacol 2021; 100:43-52. [PMID: 34425056 DOI: 10.1139/cjpp-2021-0286] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
A gamma-pyrone derivative, comenic acid, activates the opioid-like receptor-mediated signaling pathway that modulates the NaV1.8 channels in the primary sensory neuron membrane. These channels are responsible for the generation of the nociceptive signal; therefore, gamma-pyrones have great therapeutic potential as analgesics, and this effect deserves a deeper understanding. The novelty of our approach to the design of a medicinal substance is based on a combination of the data obtained from living neurons using very sensitive physiological methods and the results of quantum chemical calculations. This approach allows the correlation of the molecular structure of gamma-pyrones with their ability to evoke a physiological response of the neuron. Comenic acid can bind to two calcium cations. One of them is chelated by the carbonyl and hydroxyl functional groups, while the other forms a salt bond with the carboxylate anion. Calcium-bound gamma-pyrones have fundamentally different electrostatic properties from free gamma-pyrone molecules. These two calcium ions are key elements involved in ligand-receptor binding. It is very likely that ion-ionic interactions between these cations and anionic functional groups of the opioid-like receptor activate the latter. The calculated intercationic distance of 9.5 Å is a structural criterion for effective ligand-receptor binding of calcium-bound gamma-pyrones.
Collapse
Affiliation(s)
- Ilia V Rogachevskii
- Pavlov Institute of Physiology of the Russian Academy of Sciences, 6 Makarova Emb., Saint Petersburg 199034, Russia.,Pavlov Institute of Physiology of the Russian Academy of Sciences, 6 Makarova Emb., Saint Petersburg 199034, Russia
| | - Vera B Plakhova
- Pavlov Institute of Physiology of the Russian Academy of Sciences, 6 Makarova Emb., Saint Petersburg 199034, Russia.,Pavlov Institute of Physiology of the Russian Academy of Sciences, 6 Makarova Emb., Saint Petersburg 199034, Russia
| | - Valentina A Penniyaynen
- Pavlov Institute of Physiology of the Russian Academy of Sciences, 6 Makarova Emb., Saint Petersburg 199034, Russia.,Pavlov Institute of Physiology of the Russian Academy of Sciences, 6 Makarova Emb., Saint Petersburg 199034, Russia
| | - Stanislav G Terekhin
- Pavlov Institute of Physiology of the Russian Academy of Sciences, 6 Makarova Emb., Saint Petersburg 199034, Russia.,Pavlov Institute of Physiology of the Russian Academy of Sciences, 6 Makarova Emb., Saint Petersburg 199034, Russia
| | - Svetlana A Podzorova
- Pavlov Institute of Physiology of the Russian Academy of Sciences, 6 Makarova Emb., Saint Petersburg 199034, Russia.,Pavlov Institute of Physiology of the Russian Academy of Sciences, 6 Makarova Emb., Saint Petersburg 199034, Russia
| | - Boris V Krylov
- Pavlov Institute of Physiology of the Russian Academy of Sciences, 6 Makarova Emb., Saint Petersburg 199034, Russia.,Pavlov Institute of Physiology of the Russian Academy of Sciences, 6 Makarova Emb., Saint Petersburg 199034, Russia
| |
Collapse
|
24
|
Dusan M, Jastrow C, Alyce MM, Yingkai W, Shashikanth M, Andelain E, Christine BM, Stuart BM, Oliver BG, Michael MZ, Nicolas VH, Damien KJ, Rainer HV. Differentiation of the 50B11 dorsal ganglion cells into NGF and GDNF responsive nociceptor subtypes. Mol Pain 2021; 16:1744806920970368. [PMID: 33307981 PMCID: PMC7745567 DOI: 10.1177/1744806920970368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The embryonic rat dorsal root ganglion (DRG) neuron-derived 50B11 cell line is a promising sensory neuron model expressing markers characteristic of NGF and GDNF-dependent C-fibre nociceptors. Whether these cells have the capacity to develop into distinct nociceptive subtypes based on NGF- or GDNF-dependence has not been investigated. Here we show that by augmenting forskolin (FSK) and growth factor supplementation with NGF or GDNF, 50B11 cultures can be driven to acquire differential functional responses to common nociceptive agonists capsaicin and ATP respectively. In addition, to previous studies, we also demonstrate that a differentiated neuronal phenotype can be maintained for up to 7 days. Western blot analysis of nociceptive marker proteins further demonstrates that the 50B11 cells partially recapitulate the functional phenotypes of classical NGF-dependent (peptidergic) and GDNF-dependent (non-peptidergic) neuronal subtypes described in DRGs. Further, 50B11 cells differentiated with NGF/FSK, but not GDNF/FSK, show sensitization to acute prostaglandin E2 treatment. Finally, RNA-Seq analysis confirms that differentiation with NGF/FSK or GDNF/FSK produces two 50B11 cell subtypes with distinct transcriptome expression profiles. Gene ontology comparison of the two subtypes of differentiated 50B11 cells to rodent DRG neurons studies shows significant overlap in matching or partially matching categories. This transcriptomic analysis will aid future suitability assessment of the 50B11 cells as a high-throughput nociceptor model for a broad range of experimental applications. In conclusion, this study shows that the 50B11 cell line is capable of partially recapitulating features of two distinct types of embryonic NGF and GDNF-dependent nociceptor-like cells.
Collapse
Affiliation(s)
- Matusica Dusan
- Anatomy and Histology, Flinders Health & Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Canlas Jastrow
- Anatomy and Histology, Flinders Health & Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Martin M Alyce
- Human Physiology, Flinders Health & Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Wei Yingkai
- Flinders Centre for Innovation in Cancer, College of Medicine and Public Health, Flinders University and Medical Centre, Bedford Park, Adelaide, South Australia, Australia
| | - Marri Shashikanth
- Visceral Pain Research Group, College of Medicine and Public Health, South Australian Health and Medical Research Institute, Flinders University, Adelaide, South Australia, Australia
| | - Erickson Andelain
- Flow Cytometry Facility, Department of Molecular Medicine and Genetics, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Barry M Christine
- Anatomy and Histology, Flinders Health & Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Brierley M Stuart
- Flow Cytometry Facility, Department of Molecular Medicine and Genetics, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Best G Oliver
- Flow Cytometry Facility, Department of Molecular Medicine and Genetics, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Michael Z Michael
- Visceral Pain Research Group, College of Medicine and Public Health, South Australian Health and Medical Research Institute, Flinders University, Adelaide, South Australia, Australia
| | - Voelcker H Nicolas
- Flinders Centre for Innovation in Cancer, College of Medicine and Public Health, Flinders University and Medical Centre, Bedford Park, Adelaide, South Australia, Australia
| | - Keating J Damien
- Human Physiology, Flinders Health & Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Haberberger V Rainer
- Anatomy and Histology, Flinders Health & Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| |
Collapse
|
25
|
Herreros P, Ballesteros-Esteban LM, Laguna MF, Leyva I, Sendiña-Nadal I, Holgado M. Neuronal circuits on a chip for biological network monitoring. Biotechnol J 2021; 16:e2000355. [PMID: 33984186 DOI: 10.1002/biot.202000355] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 03/16/2021] [Accepted: 04/28/2021] [Indexed: 11/09/2022]
Abstract
Cultured neuronal networks (CNNs) are a robust model to closely investigate neuronal circuits' formation and monitor their structural properties evolution. Typically, neurons are cultured in plastic plates or, more recently, in microfluidic platforms with potentially a wide variety of neuroscience applications. As a biological protocol, cell culture integration with a microfluidic system provides benefits such as accurate control of cell seeding area, culture medium renewal, or lower exposure to contamination. The objective of this report is to present a novel neuronal network on a chip device, including a chamber, fabricated from PDMS, vinyl and glass connected to a microfluidic platform to perfuse the continuous flow of culture medium. Network growth is compared in chips and traditional Petri dishes to validate the microfluidic chip performance. The network assessment is performed by computing relevant topological measures like the number of connected neurons, the clustering coefficient, and the shortest path between any pair of neurons throughout the culture's life. The results demonstrate that neuronal circuits on a chip have a more stable network structure and lifespan than developing in conventional settings, and therefore this setup is an advantageous alternative to current culture methods. This technology could lead to challenging applications such as batch drug testing of in vitro cell culture models. From the engineering perspective, a device's advantage is the chance to develop custom designs more efficiently than other microfluidic systems.
Collapse
Affiliation(s)
- Pedro Herreros
- Group of Optics, Photonics and Biophotonics (GOFB), Center for Biomedical Technology, Universidad Politécnica de Madrid, Madrid, Spain.,Group of Organ and Tissue on-a-chip and In-Vitro Detection, Health Research Institute of the Hospital Clínico San Carlos, Madrid, Spain
| | - Luis M Ballesteros-Esteban
- Complex Systems Group & GISC, Universidad Rey Juan Carlos, Madrid, Spain.,Group of Biological Networks, Center for Biomedical Technology, Universidad Politécnica de Madrid, Madrid, Spain
| | - María Fe Laguna
- Group of Optics, Photonics and Biophotonics (GOFB), Center for Biomedical Technology, Universidad Politécnica de Madrid, Madrid, Spain.,Group of Organ and Tissue on-a-chip and In-Vitro Detection, Health Research Institute of the Hospital Clínico San Carlos, Madrid, Spain.,Departamento de Física Aplicada e Ingeniería de Materiales, Escuela Técnica Superior de Ingenieros Industriales, Universidad Politécnica de Madrid, Madrid, Spain
| | - Inmaculada Leyva
- Complex Systems Group & GISC, Universidad Rey Juan Carlos, Madrid, Spain.,Group of Biological Networks, Center for Biomedical Technology, Universidad Politécnica de Madrid, Madrid, Spain
| | - Irene Sendiña-Nadal
- Complex Systems Group & GISC, Universidad Rey Juan Carlos, Madrid, Spain.,Group of Biological Networks, Center for Biomedical Technology, Universidad Politécnica de Madrid, Madrid, Spain
| | - Miguel Holgado
- Group of Optics, Photonics and Biophotonics (GOFB), Center for Biomedical Technology, Universidad Politécnica de Madrid, Madrid, Spain.,Group of Organ and Tissue on-a-chip and In-Vitro Detection, Health Research Institute of the Hospital Clínico San Carlos, Madrid, Spain.,Departamento de Física Aplicada e Ingeniería de Materiales, Escuela Técnica Superior de Ingenieros Industriales, Universidad Politécnica de Madrid, Madrid, Spain
| |
Collapse
|
26
|
Buckle T, Hensbergen AW, van Willigen DM, Bosse F, Bauwens K, Pelger RCM, van Leeuwen FWB. Intraoperative visualization of nerves using a myelin protein-zero specific fluorescent tracer. EJNMMI Res 2021; 11:50. [PMID: 34052912 PMCID: PMC8164657 DOI: 10.1186/s13550-021-00792-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 05/17/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Surgically induced nerve damage is a common but debilitating side effect in oncological surgery. With the aim to use fluorescence guidance to enable nerve-sparing interventions in future surgery, a fluorescent tracer was developed that specifically targets myelin protein zero (P0). RESULTS Truncated homotypic P0 protein-based peptide sequences were C-terminally functionalized with the far-red cyanine dye Cy5. The lead compound Cy5-P0101-125 was selected after initial solubility, (photo)physical and in vitro evaluation (including P0-blocking experiments). Cy5-P0101-125 (KD = 105 ± 17 nM) allowed in vitro and ex vivo P0-related staining. Furthermore, Cy5-P0101-125 enabled in vivo fluorescence imaging of the Sciatic nerve in mice after local intravenous (i.v.) administration and showed compatibility with a clinical fluorescence laparoscope during evaluation in a porcine model undergoing robot-assisted surgery. Biodistribution data revealed that i.v. administered [111In]In-DTPA-P0101-125 does not enter the central nervous system (CNS). CONCLUSION P0101-125 has proven to be a potent nerve-specific agent that is able to target P0/myelin under in vitro, ex vivo, and in vivo conditions without posing a threat for CNS-related toxicity.
Collapse
Affiliation(s)
- Tessa Buckle
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands.
| | - Albertus W Hensbergen
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands
| | - Danny M van Willigen
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands
| | - Frank Bosse
- Neurologische Klinik, Heinrich-Heine University Dusseldorf, Düsseldorf, Germany
| | | | - Rob C M Pelger
- Department of Urology, Leiden University Medical Center, Leiden, The Netherlands
| | - Fijs W B van Leeuwen
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2300 RC, Leiden, The Netherlands.
- ORSI Academy, Melle, Belgium.
| |
Collapse
|
27
|
Schutte SC, Kadakia F, Davidson S. Skin-Nerve Co-Culture Systems for Disease Modeling and Drug Discovery. Tissue Eng Part C Methods 2021; 27:89-99. [PMID: 33349133 DOI: 10.1089/ten.tec.2020.0296] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Prominent clinical problems related to the skin-nerve interface include barrier dysfunction and erythema, but it is the symptoms of pain and itch that most often lead patients to seek medical treatment. Tissue-engineered innervated skin models provide an excellent solution for studying the mechanisms underlying neurocutaneous disorders for drug screening, and cutaneous device development. Innervated skin substitutes provide solutions beyond traditional monolayer cultures and have advantages that make them preferable to in vivo animal studies for certain applications, such as measuring somatosensory transduction. The tissue-engineered innervated skin models replicate the complex stratified epidermis that provides barrier function in native skin, a feature that is lacking in monolayer co-cultures, while allowing for a level of detail in measurement of nerve morphology and function that cannot be achieved in animal models. In this review, the advantages and disadvantages of different cell sources and scaffold materials will be discussed and a presentation of the current state of the field is reviewed. Impact statement A review of the current state of innervated skin substitutes and the considerations that need to be addressed when developing these models. Tissue-engineered skin substitutes are customizable and provide barrier function allowing for screening of topical drugs and for studying nerve function.
Collapse
Affiliation(s)
- Stacey C Schutte
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, Ohio, USA
| | - Feni Kadakia
- Department of Anesthesiology, Pain Research Center, and Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Steve Davidson
- Department of Anesthesiology, Pain Research Center, and Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
28
|
Cong M, Shen M, Wu X, Li Y, Wang L, He Q, Shi H, Ding F. Improvement of sensory neuron growth and survival via negatively regulating PTEN by miR-21-5p-contained small extracellular vesicles from skin precursor-derived Schwann cells. Stem Cell Res Ther 2021; 12:80. [PMID: 33494833 PMCID: PMC7831194 DOI: 10.1186/s13287-020-02125-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 12/25/2020] [Indexed: 12/11/2022] Open
Abstract
Background Patients with peripheral nerve injury (PNI) often suffer from hypoxic ischemic impairments, in particular when combined with vascular damage, causing neuronal dysfunction and death. Increasing attention has been paid on skin precursor-derived Schwann cells (SKP-SCs), and previous study has shown that SKP-SCs could promote sensory recovery after cell therapy for PNI, resembling the effect of naive SCs, and SKP-SC-derived extracellular vesicles (SKP-SC-EVs) are putatively supposed to be promising therapeutic agents for neural regeneration. Methods SKPs were induced to differentiate towards SCs with cocktail factors (N2, neuregulin-1β, and forskolin) in vitro. SKP-SC-EVs were isolated by exoEasy Maxi Kit and characterized by morphology and phenotypic markers of EVs. Rat sensory neurons from dorsal root ganglions (DRGs) were primarily cultured in regular condition or exposed to oxygen-glucose-deprivation (OGD) condition. SKP-SC-EVs were applied to DRGs or sensory neurons, with LY294002 (a PI3K inhibitor) added; the effect on neurite outgrowth and cell survival was observed. Moreover, microRNA (miR) candidate contained in SKP-SC-EVs was screened out, and miR-mimics were transfected into DRG neurons; meanwhile, the negative regulation of PTEN/PI3K/Akt axis and downstream signaling molecules were determined. Results It was shown that SKP-SC-EVs could improve the neurite outgrowth of DRGs and sensory neurons. Furthermore, SKP-SC-EVs enhanced the survival of sensory neurons after OGD exposure by alleviating neuronal apoptosis and strengthening cell viability, and the expression of GAP43 (a neuron functional protein) in neurons was upregulated. Moreover, the neuro-reparative role of SKP-SC-EVs was implicated in the activation of PI3K/Akt, mTOR, and p70S6k, as well as the reduction of Bax/Bcl-2 ratio, that was compromised by LY294002 to some extent. In addition, transferring miR-21-5p mimics into sensory neurons could partly protect them from OGD-induced impairment. Conclusions Sum up, SKP-SC-EVs could improve neurite outgrowth of DRG sensory neurons in physiological and pathological condition. Moreover, the in vitro therapeutic potential of SKP-SC-EVs on the survival and restoration of OGD-injured sensory neurons was evidenced to be associated with miR-21-5p contained in the small EVs and miR-21-5p/PTEN/PI3K/Akt axis. Graphic abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s13287-020-02125-4.
Collapse
Affiliation(s)
- Meng Cong
- School of Biology and Basic Medical Sciences, Soochow University, Suzhou, 215123, China.,Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education and Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, China
| | - Mi Shen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education and Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, China
| | - Xia Wu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education and Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, China
| | - Yan Li
- School of Biology and Basic Medical Sciences, Soochow University, Suzhou, 215123, China.,Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education and Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, China
| | - Liting Wang
- Department of Pathophysiology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong, 226001, China
| | - Qianru He
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education and Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, China
| | - Haiyan Shi
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education and Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, China. .,Department of Pathophysiology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong, 226001, China.
| | - Fei Ding
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education and Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, China. .,Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, China.
| |
Collapse
|
29
|
Magadmi RM, Alsulaimani MA, Al-Rafiah AR, Esmat A. The Neuroprotective Effect of Carvedilol on Diabetic Neuropathy: An In Vitro Study. J Diabetes Res 2021; 2021:6927025. [PMID: 33532503 PMCID: PMC7834839 DOI: 10.1155/2021/6927025] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 12/23/2020] [Accepted: 01/06/2021] [Indexed: 01/22/2023] Open
Abstract
Diabetic neuropathy serves as a major complication for diabetic patients across the world. The use of effective treatment is integral for reducing the health complications for diabetic patients. This study has evaluated the carvedilol potential neuroprotective effect on diabetic neuropathy. An in vitro model of diabetic neuropathy was used, including dorsal root ganglia (DRG) that were cultured from male adult mice C57BL. These were incubated for about twenty-four hours in high glucose (HG) media (45 mM). Some cells were incubated with carvedilol (10 μM). Neuronal viability, neuronal morphology, and activating transcription factor 3 (AFT3) were measured. The cell viability was decreased, along with neuronal length, soma area, and soma perimeter with HG media. Also, there was an overexpression of ATF3, which is a neuronal stress response marker. The pretreatment with carvedilol increased the viability of DRG as compared to HG-treated cells. Also, it significantly protected the DRG from HG-induced morphology changes. Though it shows a decrease in AFT3 expression, the statistical results were insignificant. The current study demonstrates the neuroprotective effect of carvedilol against HG-induced DN using an in vitro model. This could be through carvedilol antioxidant effects.
Collapse
Affiliation(s)
- Rania M. Magadmi
- Department of Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Neuroscience Unit, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mujahid A. Alsulaimani
- Department of Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Pharmacy, Ministry of Health, Taif, Saudi Arabia
| | - Aziza Rashed Al-Rafiah
- Department of Pharmacy, Ministry of Health, Taif, Saudi Arabia
- Medical Laboratory Technology Department, Faculty of Applied Medical Sciences, King Abdulaziz University, Saudi Arabia
| | - Ahmed Esmat
- Department of Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| |
Collapse
|
30
|
De Gregorio C, Ezquer F. Sensory neuron cultures derived from adult db/db mice as a simplified model to study type-2 diabetes-associated axonal regeneration defects. Dis Model Mech 2021; 14:dmm.046334. [PMID: 33262160 PMCID: PMC7847260 DOI: 10.1242/dmm.046334] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 11/17/2020] [Indexed: 12/11/2022] Open
Abstract
Diabetic neuropathy (DN) is an early common complication of diabetes mellitus (DM), leading to chronic pain, sensory loss and muscle atrophy. Owing to its multifactorial etiology, neuron in vitro cultures have been proposed as simplified systems for DN studies. However, the most used models currently available do not recreate the chronic and systemic damage suffered by peripheral neurons of type-2 DM (T2DM) individuals. Here, we cultured neurons derived from dorsal root ganglia from 6-month-old diabetic db/db-mice, and evaluated their morphology by the Sholl method as an easy-to-analyze readout of neuronal function. We showed that neurons obtained from diabetic mice exhibited neuritic regeneration defects in basal culture conditions, compared to neurons from non-diabetic mice. Next, we evaluated the morphological response to common neuritogenic factors, including nerve growth factor NGF and Laminin-1 (also called Laminin-111). Neurons derived from diabetic mice exhibited reduced regenerative responses to these factors compared to neurons from non-diabetic mice. Finally, we analyzed the neuronal response to a putative DN therapy based on the secretome of mesenchymal stem cells (MSC). Neurons from diabetic mice treated with the MSC secretome displayed a significant improvement in neuritic regeneration, but still reduced when compared to neurons derived from non-diabetic mice. This in vitro model recapitulates many alterations observed in sensory neurons of T2DM individuals, suggesting the possibility of studying neuronal functions without the need of adding additional toxic factors to culture plates. This model may be useful for evaluating intrinsic neuronal responses in a cell-autonomous manner, and as a throughput screening for the pre-evaluation of new therapies for DN. Summary: Morphological characterization of a model for evaluating neuritic regeneration in vitro in dorsal root ganglion primary neurons derived from type-2 diabetic mice with an advanced stage of diabetic neuropathy.
Collapse
Affiliation(s)
- Cristian De Gregorio
- Center for Regenerative Medicine, School of Medicine Clínica Alemana-Universidad del Desarrollo, Santiago, 7690000 Chile
| | - Fernando Ezquer
- Center for Regenerative Medicine, School of Medicine Clínica Alemana-Universidad del Desarrollo, Santiago, 7690000 Chile
| |
Collapse
|
31
|
Ita ME, Ghimire P, Welch RL, Troche HR, Winkelstein BA. Intra-articular collagenase in the spinal facet joint induces pain, DRG neuron dysregulation and increased MMP-1 absent evidence of joint destruction. Sci Rep 2020; 10:21965. [PMID: 33319791 PMCID: PMC7738551 DOI: 10.1038/s41598-020-78811-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 11/17/2020] [Indexed: 12/11/2022] Open
Abstract
Degeneration is a hallmark of painful joint disease and is mediated by many proteases that degrade joint tissues, including collagenases. We hypothesized that purified bacterial collagenase would initiate nociceptive cascades in the joint by degrading the capsular ligament's matrix and activating innervating pain fibers. Intra-articular collagenase in the rat facet joint was investigated for its effects on behavioral sensitivity, joint degeneration, and nociceptive pathways in the peripheral and central nervous systems. In parallel, a co-culture collagen gel model of the ligament was used to evaluate effects of collagenase on microscale changes to the collagen fibers and embedded neurons. Collagenase induced sensitivity within one day, lasting for 3 weeks (p < 0.001) but did not alter ligament structure, cartilage health, or chondrocyte homeostasis. Yet, nociceptive mediators were increased in the periphery (substance P, pERK, and MMP-1; p ≤ 0.039) and spinal cord (substance P and MMP-1; p ≤ 0.041). The collagen loss (p = 0.008) induced by exposing co-cultures to collagenase was accompanied by altered neuronal activity (p = 0.002) and elevated neuronal MMP-1 (p < 0.001), suggesting microscale collagen degradation mediates sensitivity in vivo. The induction of sustained sensitivity and nociception without joint damage may explain the clinical disconnect in which symptomatic joint pain patients present without radiographic evidence of joint destruction.
Collapse
Affiliation(s)
- Meagan E Ita
- Department of Bioengineering, University of Pennsylvania, 210 S. 33rd Street, 240 Skirkanich Hall, Philadelphia, PA, 19104-6392, USA
| | - Prabesh Ghimire
- Department of Bioengineering, University of Pennsylvania, 210 S. 33rd Street, 240 Skirkanich Hall, Philadelphia, PA, 19104-6392, USA
| | - Rachel L Welch
- Department of Bioengineering, University of Pennsylvania, 210 S. 33rd Street, 240 Skirkanich Hall, Philadelphia, PA, 19104-6392, USA
| | - Harrison R Troche
- Department of Bioengineering, University of Pennsylvania, 210 S. 33rd Street, 240 Skirkanich Hall, Philadelphia, PA, 19104-6392, USA
| | - Beth A Winkelstein
- Department of Bioengineering, University of Pennsylvania, 210 S. 33rd Street, 240 Skirkanich Hall, Philadelphia, PA, 19104-6392, USA.
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
32
|
Aravamudhan P, Raghunathan K, Dermody TS. Confocal Microscopy of Reovirus Transport in Living Dorsal Root Ganglion Neurons. Bio Protoc 2020; 10:e3825. [PMID: 33659477 DOI: 10.21769/bioprotoc.3825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 09/23/2020] [Accepted: 09/28/2020] [Indexed: 11/02/2022] Open
Abstract
Neurotropic reoviruses repurpose host machinery to traffic over long distances in neuronal processes and access distal replication sites. Understanding mechanisms of neuronal transmission is facilitated by using simplified in vitro primary neuronal culture models. Advances in the design of compartmentalized microfluidic devices lend robustness to neuronal culture models by enabling compartmentalization and manipulation of distinct neuronal processes. Here, we describe a streamlined methodology to culture sensory neurons dissociated from dorsal root ganglia of embryonic rats in microfluidic devices. We further describe protocols to exogenously label reovirus and image, track, and analyze transport of single reovirus particles in living neurons. These techniques can be adapted to study directed axonal transport of other neurotropic viruses and neuronal factors involved in signaling and pathology.
Collapse
Affiliation(s)
- Pavithra Aravamudhan
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, USA.,Center for Microbial Pathogenesis, UPMC Children's Hospital of Pittsburgh, Pittsburgh, USA
| | - Krishnan Raghunathan
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, USA.,Center for Microbial Pathogenesis, UPMC Children's Hospital of Pittsburgh, Pittsburgh, USA
| | - Terence S Dermody
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, USA.,Center for Microbial Pathogenesis, UPMC Children's Hospital of Pittsburgh, Pittsburgh, USA.,Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, USA
| |
Collapse
|
33
|
Verma P, Eaton M, Kienle A, Flockerzi D, Yang Y, Ramkrishna D. Examining Sodium and Potassium Channel Conductances Involved in Hyperexcitability of Chemotherapy-Induced Peripheral Neuropathy: A Mathematical and Cell Culture-Based Study. Front Comput Neurosci 2020; 14:564980. [PMID: 33178002 PMCID: PMC7593680 DOI: 10.3389/fncom.2020.564980] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 08/02/2020] [Indexed: 11/13/2022] Open
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a prevalent, painful side effect which arises due to a number of chemotherapy agents. CIPN can have a prolonged effect on quality of life. Chemotherapy treatment is often reduced or stopped altogether because of the severe pain. Currently, there are no FDA-approved treatments for CIPN partially due to its complex pathogenesis in multiple pathways involving a variety of channels, specifically, voltage-gated ion channels. One aspect of neuropathic pain in vitro is hyperexcitability in dorsal root ganglia (DRG) peripheral sensory neurons. Our study employs bifurcation theory to investigate the role of voltage-gated ion channels in inducing hyperexcitability as a consequence of spontaneous firing due to the common chemotherapy agent paclitaxel. Our mathematical investigation of a reductionist DRG neuron model comprised of sodium channel Nav1.7, sodium channel Nav1.8, delayed rectifier potassium channel, A-type transient potassium channel, and a leak channel suggests that Nav1.8 and delayed rectifier potassium channel conductances are critical for hyperexcitability of small DRG neurons. Introducing paclitaxel into the model, our bifurcation analysis predicts that hyperexcitability is highest for a medium dose of paclitaxel, which is supported by multi-electrode array (MEA) recordings. Furthermore, our findings using MEA reveal that Nav1.8 blocker A-803467 and delayed rectifier potassium enhancer L-alpha-phosphatidyl-D-myo-inositol 4,5-diphosphate, dioctanoyl (PIP2) can reduce paclitaxel-induced hyperexcitability of DRG neurons. Our approach can be readily extended and used to investigate various other contributors of hyperexcitability in CIPN.
Collapse
Affiliation(s)
- Parul Verma
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, United States
| | - Muriel Eaton
- Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, United States
| | - Achim Kienle
- Process Synthesis and Dynamics Group, Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
- Chair for Automation/Modeling, Otto von Guericke University, Magdeburg, Germany
| | - Dietrich Flockerzi
- Max Planck Institute for Dynamics of Complex Technical Systems, Magdeburg, Germany
- Otto von Guericke University, Magdeburg, Germany
| | - Yang Yang
- Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, United States
| | - Doraiswami Ramkrishna
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, United States
| |
Collapse
|
34
|
Bae B, Gruner HN, Lynch M, Feng T, So K, Oliver D, Mastick GS, Yan W, Pieraut S, Miura P. Elimination of Calm1 long 3'-UTR mRNA isoform by CRISPR-Cas9 gene editing impairs dorsal root ganglion development and hippocampal neuron activation in mice. RNA (NEW YORK, N.Y.) 2020; 26:1414-1430. [PMID: 32522888 PMCID: PMC7491327 DOI: 10.1261/rna.076430.120] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 06/06/2020] [Indexed: 05/04/2023]
Abstract
The majority of mouse and human genes are subject to alternative cleavage and polyadenylation (APA), which most often leads to the expression of two or more alternative length 3' untranslated region (3'-UTR) mRNA isoforms. In neural tissues, there is enhanced expression of APA isoforms with longer 3'-UTRs on a global scale, but the physiological relevance of these alternative 3'-UTR isoforms is poorly understood. Calmodulin 1 (Calm1) is a key integrator of calcium signaling that generates short (Calm1-S) and long (Calm1-L) 3'-UTR mRNA isoforms via APA. We found Calm1-L expression to be largely restricted to neural tissues in mice including the dorsal root ganglion (DRG) and hippocampus, whereas Calm1-S was more broadly expressed. smFISH revealed that both Calm1-S and Calm1-L were subcellularly localized to neural processes of primary hippocampal neurons. In contrast, cultured DRG showed restriction of Calm1-L to soma. To investigate the in vivo functions of Calm1-L, we implemented a CRISPR-Cas9 gene editing strategy to delete a small region encompassing the Calm1 distal poly(A) site. This eliminated Calm1-L expression while maintaining expression of Calm1-S Mice lacking Calm1-L (Calm1ΔL/ΔL ) exhibited disorganized DRG migration in embryos, and reduced experience-induced neuronal activation in the adult hippocampus. These data indicate that Calm1-L plays functional roles in the central and peripheral nervous systems.
Collapse
Affiliation(s)
- Bongmin Bae
- Department of Biology, University of Nevada, Reno, Nevada 89557, USA
| | - Hannah N Gruner
- Department of Biology, University of Nevada, Reno, Nevada 89557, USA
| | - Maebh Lynch
- Department of Biology, University of Nevada, Reno, Nevada 89557, USA
| | - Ting Feng
- Department of Biology, University of Nevada, Reno, Nevada 89557, USA
| | - Kevin So
- Department of Biology, University of Nevada, Reno, Nevada 89557, USA
| | - Daniel Oliver
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, Nevada 89557, USA
| | - Grant S Mastick
- Department of Biology, University of Nevada, Reno, Nevada 89557, USA
| | - Wei Yan
- Department of Biology, University of Nevada, Reno, Nevada 89557, USA
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, Nevada 89557, USA
| | - Simon Pieraut
- Department of Biology, University of Nevada, Reno, Nevada 89557, USA
| | - Pedro Miura
- Department of Biology, University of Nevada, Reno, Nevada 89557, USA
| |
Collapse
|
35
|
Johnstone AFM, Mack CM, Valdez MC, Shafer TJ, LoPachin RM, Herr DW, Kodavanti PRS. Acute in vitro effects on embryonic rat dorsal root ganglion (DRG) cultures by in silico predicted neurotoxic chemicals: Evaluations on cytotoxicity, neurite length, and neurophysiology. Toxicol In Vitro 2020; 69:104989. [PMID: 32882341 DOI: 10.1016/j.tiv.2020.104989] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 08/25/2020] [Accepted: 08/26/2020] [Indexed: 11/25/2022]
Abstract
The Hard-Soft Acid and Base hypothesis can be used to predict the potential bio-reactivity (electrophilicity) of a chemical with intracellular proteins, resulting in neurotoxicity. Twelve chemicals predicted to be neurotoxic were evaluated in vitro in rat dorsal root ganglia (DRG) for effects on cytotoxicity (%LDH), neuronal structure (total neurite length/neuron, NLPN), and neurophysiology (mean firing rate, MFR). DRGs were treated acutely on days in vitro (DIV) 7 (1-100 μM) with test chemical; %LDH and NLPN were measured after 48 h. 4-cyclohexylhexanone (4-C) increased %LDH release at 50 (29%) and 100 μM (56%), citronellal (Cit) and 1-bromopropane increased %LDH at 100 μM (22% and 26%). 4-C, Cit, 2,5 Hexanedione (2,5Hex), phenylacetylaldehyde (PAA) and 2-ethylhexanal decreased mean NLPN at 48 h; 50 and 100 μM for 4-C (28% and 60%), 100 μM Cit (52%), 100 μM 2,5- Hex (37%) 100 μM PAA (41%) and 100 μM for 2-ethylhexanal (23%). Separate DRG cultures were treated on DIV 14 and changes in MFR measured. Four compounds decreased MFR at 50 or 100 μM: Acrylamide (-83%), 3,4-dichloro-1-butene (-93%), 4-C (-89%) and hexane (-79%, 50 μM). Changes in MFR and NLPN occurred in absence of cytotoxicity. While the current study showed little cytotoxicity, it gave insight to initial changes in MFR. Results provide insight for future chronic exposure experiments to evaluate neurotoxicity.
Collapse
Affiliation(s)
- Andrew F M Johnstone
- Clinical Research Branch, Public Health and Integrated Toxicology Division, CPHEA/ORD, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA.
| | - Cina M Mack
- Neurological and Endocrine Toxicology Branch, Public Health and Integrated Toxicology Division, CPHEA/ORD, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA
| | - Matthew C Valdez
- Oak Ridge Institute for Science and Education, U.S. Department of Energy, Oak Ridge, TN 37831, USA
| | - Timothy J Shafer
- Rapid Assay Development Branch, Biomolecular and Computational Toxicology Division, CCTE/ORD, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA
| | - Richard M LoPachin
- Department of Anesthesiology, Montefiore Medical Center, Albert Einstein College of Medicine, 111 E. 210th St, Bronx, NY 10467, United States of America
| | - David W Herr
- Neurological and Endocrine Toxicology Branch, Public Health and Integrated Toxicology Division, CPHEA/ORD, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA
| | - Prasada Rao S Kodavanti
- Neurological and Endocrine Toxicology Branch, Public Health and Integrated Toxicology Division, CPHEA/ORD, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA
| |
Collapse
|
36
|
Zhao J, Ding Y, He R, Huang K, Liu L, Jiang C, Liu Z, Wang Y, Yan X, Cao F, Huang X, Peng Y, Ren R, He Y, Cui T, Zhang Q, Zhang X, Liu Q, Li Y, Ma Z, Yi X. Dose-effect relationship and molecular mechanism by which BMSC-derived exosomes promote peripheral nerve regeneration after crush injury. Stem Cell Res Ther 2020; 11:360. [PMID: 32811548 PMCID: PMC7437056 DOI: 10.1186/s13287-020-01872-8] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 07/28/2020] [Accepted: 08/03/2020] [Indexed: 12/13/2022] Open
Abstract
Background The development of new treatment strategies to improve peripheral nerve repair after injury, especially those that accelerate axonal nerve regeneration, is very important. The aim of this study is to elucidate the molecular mechanisms of how bone marrow stromal cell (BMSC)-derived exosomes (EXOs) participate in peripheral nerve regeneration and whether the regenerative effect of EXOs is correlated with dose. Method BMSCs were transfected with or without an siRNA targeting Ago2 (SiAgo2). EXOs extracted from the BMSCs were administered to dorsal root ganglion (DRG) neurons in vitro. After 48 h of culture, the neurite length was measured. Moreover, EXOs at four different doses were injected into the gastrocnemius muscles of rats with sciatic nerve crush injury. The sciatic nerve functional index (SFI) and latency of thermal pain (LTP) of the hind leg sciatic nerve were measured before the operation and at 7, 14, 21, and 28 days after the operation. Then, the number and diameter of the regenerated fibers in the injured distal sciatic nerve were quantified. Seven genes associated with nerve regeneration were investigated by qRT-PCR in DRG neurons extracted from rats 7 days after the sciatic nerve crush. Results We showed that after 48 h of culture, the mean number of neurites and the length of cultured DRG neurons in the SiAgo2-BMSC-EXO and SiAgo2-BMSC groups were smaller than that in the untreated and siRNA control groups. The average number and diameter of regenerated axons, LTP, and SFI in the group with 0.9 × 1010 particles/ml EXOs were better than those in other groups, while the group that received a minimum EXO dose (0.4 × 1010 particles/ml) was not significantly different from the PBS group. The expression of PMP22, VEGFA, NGFr, and S100b in DRGs from the EXO-treated group was significantly higher than that in the PBS control group. No significant difference was observed in the expression of HGF and Akt1 among the groups. Conclusions These results showed that BMSC-derived EXOs can promote the regeneration of peripheral nerves and that the mechanism may involve miRNA-mediated regulation of regeneration-related genes, such as VEGFA. Finally, a dose-effect relationship between EXO treatment and nerve regeneration was shown.
Collapse
Affiliation(s)
- Jiuhong Zhao
- Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China.,Department of Anatomy, Hainan Medical University, Haikou, China
| | - Yali Ding
- School of Medicine, Tibet University, Lhasa, China
| | - Rui He
- Department of Anatomy, Hainan Medical University, Haikou, China.,Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, China
| | - Kui Huang
- Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China
| | - Lu Liu
- Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China
| | - Chaona Jiang
- Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China
| | - Zhuozhou Liu
- Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China
| | - Yuanlan Wang
- Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China
| | - Xiaokai Yan
- Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China
| | - Fuyang Cao
- Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China
| | - Xueying Huang
- Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China
| | - Yanan Peng
- Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China.,Department of Anatomy, Hainan Medical University, Haikou, China
| | - Rui Ren
- Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China.,Department of Anatomy, Hainan Medical University, Haikou, China
| | - Yuebin He
- Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China.,Department of Anatomy, Hainan Medical University, Haikou, China
| | - Tianwei Cui
- Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China.,Department of Anatomy, Hainan Medical University, Haikou, China
| | - Quanpeng Zhang
- Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China.,Department of Anatomy, Hainan Medical University, Haikou, China
| | - Xianfang Zhang
- Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China.,Department of Anatomy, Hainan Medical University, Haikou, China
| | - Qibing Liu
- Department of Anatomy, Hainan Medical University, Haikou, China
| | - Yunqing Li
- Department of Anatomy, Hainan Medical University, Haikou, China
| | - Zhijian Ma
- Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China. .,Department of Anatomy, Hainan Medical University, Haikou, China.
| | - Xinan Yi
- Key Laboratory of Brain Science Research & Transformation in Tropical Environment of Hainan Province, Hainan Medical University, Haikou, China. .,Department of Anatomy, Hainan Medical University, Haikou, China.
| |
Collapse
|
37
|
Chakrabarti S, Ai M, Henson FM, Smith ESJ. Peripheral mechanisms of arthritic pain: A proposal to leverage large animals for in vitro studies. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2020; 8:100051. [PMID: 32817908 PMCID: PMC7426561 DOI: 10.1016/j.ynpai.2020.100051] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/22/2020] [Accepted: 07/22/2020] [Indexed: 04/14/2023]
Abstract
Pain arising from musculoskeletal disorders such as arthritis is one of the leading causes of disability. Whereas the past 20-years has seen an increase in targeted therapies for rheumatoid arthritis (RA), other arthritis conditions, especially osteoarthritis, remain poorly treated. Although modulation of central pain pathways occurs in chronic arthritis, multiple lines of evidence indicate that peripherally driven pain is important in arthritic pain. To understand the peripheral mechanisms of arthritic pain, various in vitro and in vivo models have been developed, largely in rodents. Although rodent models provide numerous advantages for studying arthritis pathogenesis and treatment, the anatomy and biomechanics of rodent joints differ considerably to those of humans. By contrast, the anatomy and biomechanics of joints in larger animals, such as dogs, show greater similarity to human joints and thus studying them can provide novel insight for arthritis research. The purpose of this article is firstly to review models of arthritis and behavioral outcomes commonly used in large animals. Secondly, we review the existing in vitro models and assays used to study arthritic pain, primarily in rodents, and discuss the potential for adopting these strategies, as well as likely limitations, in large animals. We believe that exploring peripheral mechanisms of arthritic pain in vitro in large animals has the potential to reduce the veterinary burden of arthritis in commonly afflicted species like dogs, as well as to improve translatability of pain research into the clinic.
Collapse
Affiliation(s)
- Sampurna Chakrabarti
- Department of Neuroscience, Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, Germany
- Department of Pharmacology, University of Cambridge, UK
| | - Minji Ai
- Department of Veterinary Medicine, University of Cambridge, UK
| | | | | |
Collapse
|
38
|
Wangzhou A, McIlvried LA, Paige C, Barragan-Iglesias P, Shiers S, Ahmad A, Guzman CA, Dussor G, Ray PR, Gereau RW, Price TJ. Pharmacological target-focused transcriptomic analysis of native vs cultured human and mouse dorsal root ganglia. Pain 2020; 161:1497-1517. [PMID: 32197039 PMCID: PMC7305999 DOI: 10.1097/j.pain.0000000000001866] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Dorsal root ganglion (DRG) neurons detect sensory inputs and are crucial for pain processing. They are often studied in vitro as dissociated cell cultures with the assumption that this reasonably represents in vivo conditions. However, to the best of our knowledge, no study has directly compared genome-wide transcriptomes of DRG tissue in vivo versus in vitro or between laboratories and culturing protocols. Comparing RNA sequencing-based transcriptomes of native to cultured (4 days in vitro) human or mouse DRG, we found that the overall expression levels of many ion channels and G-protein-coupled receptors specifically expressed in neurons are markedly lower although still expressed in culture. This suggests that most pharmacological targets expressed in vivo are present under the condition of dissociated cell culture, but with changes in expression levels. The reduced relative expression for neuronal genes in human DRG cultures is likely accounted for by increased expression of genes in fibroblast-like and other proliferating cells, consistent with their mitotic status in these cultures. We found that the expression of a subset of genes typically expressed in neurons increased in human and mouse DRG cultures relative to the intact ganglion, including genes associated with nerve injury or inflammation in preclinical models such as BDNF, MMP9, GAL, and ATF3. We also found a striking upregulation of a number of inflammation-associated genes in DRG cultures, although many were different between mouse and human. Our findings suggest an injury-like phenotype in DRG cultures that has important implications for the use of this model system for pain drug discovery.
Collapse
Affiliation(s)
- Andi Wangzhou
- The University of Texas at Dallas, School of Behavioral and
Brain Sciences and Center for Advanced Pain Studies, 800 W Campbell Rd. Richardson,
TX, 75080, USA
| | - Lisa A. McIlvried
- Washington University Pain Center and Department of
Anesthesiology, Washington University School of Medicine
| | - Candler Paige
- The University of Texas at Dallas, School of Behavioral and
Brain Sciences and Center for Advanced Pain Studies, 800 W Campbell Rd. Richardson,
TX, 75080, USA
| | - Paulino Barragan-Iglesias
- The University of Texas at Dallas, School of Behavioral and
Brain Sciences and Center for Advanced Pain Studies, 800 W Campbell Rd. Richardson,
TX, 75080, USA
| | - Stephanie Shiers
- The University of Texas at Dallas, School of Behavioral and
Brain Sciences and Center for Advanced Pain Studies, 800 W Campbell Rd. Richardson,
TX, 75080, USA
| | - Ayesha Ahmad
- The University of Texas at Dallas, School of Behavioral and
Brain Sciences and Center for Advanced Pain Studies, 800 W Campbell Rd. Richardson,
TX, 75080, USA
| | - Carolyn A. Guzman
- The University of Texas at Dallas, School of Behavioral and
Brain Sciences and Center for Advanced Pain Studies, 800 W Campbell Rd. Richardson,
TX, 75080, USA
| | - Gregory Dussor
- The University of Texas at Dallas, School of Behavioral and
Brain Sciences and Center for Advanced Pain Studies, 800 W Campbell Rd. Richardson,
TX, 75080, USA
| | - Pradipta R. Ray
- The University of Texas at Dallas, School of Behavioral and
Brain Sciences and Center for Advanced Pain Studies, 800 W Campbell Rd. Richardson,
TX, 75080, USA
| | - Robert W. Gereau
- Washington University Pain Center and Department of
Anesthesiology, Washington University School of Medicine
| | - Theodore J. Price
- The University of Texas at Dallas, School of Behavioral and
Brain Sciences and Center for Advanced Pain Studies, 800 W Campbell Rd. Richardson,
TX, 75080, USA
| |
Collapse
|
39
|
Atmaramani R, Pancrazio JJ, Black BJ. Adaptation of robust Z' factor for assay quality assessment in microelectrode array based screening using adult dorsal root ganglion neurons. J Neurosci Methods 2020; 339:108699. [PMID: 32224158 DOI: 10.1016/j.jneumeth.2020.108699] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 03/04/2020] [Accepted: 03/23/2020] [Indexed: 01/02/2023]
Abstract
BACKGROUND Cell-based assays comprising primary sensory neurons cultured in vitro are an emerging tool for the screening and identification of potential analgesic compounds and chronic pain treatments. High-content screening (HCS) platforms for drug screening are characterized by a measure of assay quality indicator, such as the Z'-factor, which considers the signal dynamic range and data variation using control compounds only. Although widely accepted as a quality metric in high throughput screening (HTS), standard Z'-factor are not well-suited to indicate the quality of complex cell-based assays. NEW METHOD The present study describes a method to assess assay quality in the context of extracellular recordings from dorsal root ganglion (DRG) sensory neurons cultured on multi-well microelectrode arrays. Data transformations are applied to electrophysiological parameters, such as electrode and well spike rates, for valid normality assumptions and suitability for use as a sample signal. Importantly, using transformed well-wide metrics, a robust version of the Z'-factor was applied, based on the median and median absolute deviation, to indicate assay quality and assess hit identification of putative pharmacological compounds. RESULTS Application of appropriately scaled data and robust statistics ensured insensitivity to data variation and approximation of normal distribution. The use median and median absolute deviation of log transformed well spike rates in computing the Z'-factor revealed a value of 0.61, which is accepted as an "excellent assay." Known antagonists of nociceptor-specific voltage-gated sodium ion channels were identified as true hits in the present assay format under both spontaneous and thermally stimulated conditions. COMPARISON WITH EXISTING METHODS The present approach demonstrated a large signal dynamic range and reduced sensitivity to data variation compared to standard Z'-factor used widely in HTS. CONCLUSION Overall, the present study provides a statistical basis for the implementation of a HCS platform utilizing adult DRG neurons on microelectrode arrays.
Collapse
Affiliation(s)
- Rahul Atmaramani
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA; Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Joseph J Pancrazio
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA; Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Bryan J Black
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA; Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA.
| |
Collapse
|
40
|
Atmaramani RR, Black BJ, de la Peña JB, Campbell ZT, Pancrazio JJ. Conserved Expression of Nav1.7 and Nav1.8 Contribute to the Spontaneous and Thermally Evoked Excitability in IL-6 and NGF-Sensitized Adult Dorsal Root Ganglion Neurons In Vitro. Bioengineering (Basel) 2020; 7:bioengineering7020044. [PMID: 32429423 PMCID: PMC7356605 DOI: 10.3390/bioengineering7020044] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 05/08/2020] [Accepted: 05/12/2020] [Indexed: 02/06/2023] Open
Abstract
Sensory neurons respond to noxious stimuli by relaying information from the periphery to the central nervous system via action potentials driven by voltage-gated sodium channels, specifically Nav1.7 and Nav1.8. These channels play a key role in the manifestation of inflammatory pain. The ability to screen compounds that modulate voltage-gated sodium channels using cell-based assays assumes that key channels present in vivo is maintained in vitro. Prior electrophysiological work in vitro utilized acutely dissociated tissues, however, maintaining this preparation for long periods is difficult. A potential alternative involves multi-electrode arrays which permit long-term measurements of neural spike activity and are well suited for assessing persistent sensitization consistent with chronic pain. Here, we demonstrate that the addition of two inflammatory mediators associated with chronic inflammatory pain, nerve growth factor (NGF) and interleukin-6 (IL-6), to adult DRG neurons increases their firing rates on multi-electrode arrays in vitro. Nav1.7 and Nav1.8 proteins are readily detected in cultured neurons and contribute to evoked activity. The blockade of both Nav1.7 and Nav1.8, has a profound impact on thermally evoked firing after treatment with IL-6 and NGF. This work underscores the utility of multi-electrode arrays for pharmacological studies of sensory neurons and may facilitate the discovery and mechanistic analyses of anti-nociceptive compounds.
Collapse
Affiliation(s)
- Rahul R. Atmaramani
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA; (R.R.A.); (B.J.B.)
- Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA; (J.B.d.l.P.); (Z.T.C.)
| | - Bryan J. Black
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA; (R.R.A.); (B.J.B.)
- Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA; (J.B.d.l.P.); (Z.T.C.)
| | - June Bryan de la Peña
- Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA; (J.B.d.l.P.); (Z.T.C.)
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Zachary T. Campbell
- Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA; (J.B.d.l.P.); (Z.T.C.)
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Joseph J. Pancrazio
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA; (R.R.A.); (B.J.B.)
- Correspondence: ; Tel.: +1-972-883-2138
| |
Collapse
|
41
|
De Gregorio C, Contador D, Díaz D, Cárcamo C, Santapau D, Lobos-Gonzalez L, Acosta C, Campero M, Carpio D, Gabriele C, Gaspari M, Aliaga-Tobar V, Maracaja-Coutinho V, Ezquer M, Ezquer F. Human adipose-derived mesenchymal stem cell-conditioned medium ameliorates polyneuropathy and foot ulceration in diabetic BKS db/db mice. Stem Cell Res Ther 2020; 11:168. [PMID: 32357914 PMCID: PMC7195803 DOI: 10.1186/s13287-020-01680-0] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 03/31/2020] [Accepted: 04/15/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Diabetic polyneuropathy (DPN) is the most common and early developing complication of diabetes mellitus, and the key contributor for foot ulcers development, with no specific therapies available. Different studies have shown that mesenchymal stem cell (MSC) administration is able to ameliorate DPN; however, limited cell survival and safety reasons hinder its transfer from bench to bedside. MSCs secrete a broad range of antioxidant, neuroprotective, angiogenic, and immunomodulatory factors (known as conditioned medium), which are all decreased in the peripheral nerves of diabetic patients. Furthermore, the abundance of these factors can be boosted in vitro by incubating MSCs with a preconditioning stimulus, enhancing their therapeutic efficacy. We hypothesize that systemic administration of conditioned medium derived from preconditioned MSCs could reverse DPN and prevent foot ulcer formation in a mouse model of type II diabetes mellitus. METHODS Diabetic BKS db/db mice were treated with systemic administration of conditioned medium derived from preconditioned human MSCs; conditioned medium derived from non-preconditioned MSCs or vehicle after behavioral signs of DPN was already present. Conditioned medium or vehicle administration was repeated every 2 weeks for a total of four administrations, and several functional and structural parameters characteristic of DPN were evaluated. Finally, a wound was made in the dorsal surface of both feet, and the kinetics of wound closure, re-epithelialization, angiogenesis, and cell proliferation were evaluated. RESULTS Our molecular, electrophysiological, and histological analysis demonstrated that the administration of conditioned medium derived from non-preconditioned MSCs or from preconditioned MSCs to diabetic BKS db/db mice strongly reverts the established DPN, improving thermal and mechanical sensitivity, restoring intraepidermal nerve fiber density, reducing neuron and Schwann cell apoptosis, improving angiogenesis, and reducing chronic inflammation of peripheral nerves. Furthermore, DPN reversion induced by conditioned medium administration enhances the wound healing process by accelerating wound closure, improving the re-epithelialization of the injured skin and increasing blood vessels in the wound bed in a skin injury model that mimics a foot ulcer. CONCLUSIONS Studies conducted indicate that MSC-conditioned medium administration could be a novel cell-free therapeutic approach to reverse the initial stages of DPN, avoiding the risk of lower limb amputation triggered by foot ulcer formation and accelerating the wound healing process in case it occurs.
Collapse
Affiliation(s)
- Cristian De Gregorio
- Center for Regenerative Medicine, School of Medicine Clínica Alemana-Universidad del Desarrollo, Av. Las Condes 12438, Lo Barnechea, Santiago, Chile
| | - David Contador
- Center for Regenerative Medicine, School of Medicine Clínica Alemana-Universidad del Desarrollo, Av. Las Condes 12438, Lo Barnechea, Santiago, Chile
| | - Diego Díaz
- Center for Regenerative Medicine, School of Medicine Clínica Alemana-Universidad del Desarrollo, Av. Las Condes 12438, Lo Barnechea, Santiago, Chile
| | - Constanza Cárcamo
- Center for Regenerative Medicine, School of Medicine Clínica Alemana-Universidad del Desarrollo, Av. Las Condes 12438, Lo Barnechea, Santiago, Chile
| | - Daniela Santapau
- Center for Regenerative Medicine, School of Medicine Clínica Alemana-Universidad del Desarrollo, Av. Las Condes 12438, Lo Barnechea, Santiago, Chile
| | - Lorena Lobos-Gonzalez
- Center for Regenerative Medicine, School of Medicine Clínica Alemana-Universidad del Desarrollo, Av. Las Condes 12438, Lo Barnechea, Santiago, Chile
| | - Cristian Acosta
- Institute of Histology and Embryology of Mendoza (IHEM-CONICET), School of Medicine, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Mario Campero
- Department of Neurology & Neurosurgery, Hospital José Joaquín Aguirre, Universidad de Chile, Santiago, Chile
| | - Daniel Carpio
- Institute of Anatomy, Histology and Pathology, Universidad Austral de Chile, Valdivia, Chile
| | - Caterina Gabriele
- Research Center for Advanced Biochemistry and Molecular Biology, Department of Experimental and Clinical Medicine, University of Catanzaro, Catanzaro, Italy
| | - Marco Gaspari
- Research Center for Advanced Biochemistry and Molecular Biology, Department of Experimental and Clinical Medicine, University of Catanzaro, Catanzaro, Italy
| | - Victor Aliaga-Tobar
- Advanced Center for Chronic Diseases-ACCDiS, Faculty of Chemical and Pharmaceutical Sciences, Universidad de Chile, Santiago, Chile
| | - Vinicius Maracaja-Coutinho
- Advanced Center for Chronic Diseases-ACCDiS, Faculty of Chemical and Pharmaceutical Sciences, Universidad de Chile, Santiago, Chile
| | - Marcelo Ezquer
- Center for Regenerative Medicine, School of Medicine Clínica Alemana-Universidad del Desarrollo, Av. Las Condes 12438, Lo Barnechea, Santiago, Chile
| | - Fernando Ezquer
- Center for Regenerative Medicine, School of Medicine Clínica Alemana-Universidad del Desarrollo, Av. Las Condes 12438, Lo Barnechea, Santiago, Chile.
| |
Collapse
|
42
|
Triaca V, Fico E, Sposato V, Caioli S, Ciotti MT, Zona C, Mercanti D, La Mendola D, Satriano C, Rizzarelli E, Tirassa P, Calissano P. hNGF Peptides Elicit the NGF-TrkA Signalling Pathway in Cholinergic Neurons and Retain Full Neurotrophic Activity in the DRG Assay. Biomolecules 2020; 10:biom10020216. [PMID: 32024191 PMCID: PMC7072391 DOI: 10.3390/biom10020216] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 01/22/2020] [Accepted: 01/26/2020] [Indexed: 12/18/2022] Open
Abstract
In the last decade, Nerve Growth Factor (NGF)-based clinical approaches have lacked specific and efficient Tyrosine Kinase A (TrkA) agonists for brain delivery. Nowadays, the characterization of novel small peptidomimetic is taking centre stage in preclinical studies, in order to overcome the main size-related limitation in brain delivery of NGF holoprotein for Central Nervous System (CNS) pathologies. Here we investigated the NGF mimetic properties of the human NGF 1–14 sequence (hNGF1–14) and its derivatives, by resorting to primary cholinergic and dorsal root ganglia (DRG) neurons. Briefly, we observed that: 1) hNGF1–14 peptides engage the NGF pathway through TrkA phosphorylation at tyrosine 490 (Y490), and activation of ShcC/PI3K and Plc-γ/MAPK signalling, promoting AKT-dependent survival and CREB-driven neuronal activity, as seen by levels of the immediate early gene c-Fos, of the cholinergic marker Choline Acetyltransferase (ChAT), and of Brain Derived Neurotrophic Factor (BDNF); 2) their NGF mimetic activity is lost upon selective TrkA inhibition by means of GW441756; 3) hNGF1–14 peptides are able to sustain DRG survival and differentiation in absence of NGF. Furthermore, the acetylated derivative Ac-hNGF1–14 demonstrated an optimal NGF mimetic activity in both neuronal paradigms and an electrophysiological profile similar to NGF in cholinergic neurons. Cumulatively, the findings here reported pinpoint the hNGF1–14 peptide, and in particular its acetylated derivative, as novel, specific and low molecular weight TrkA specific agonists in both CNS and PNS primary neurons.
Collapse
Affiliation(s)
- Viviana Triaca
- Institute of Biochemistry and Cell Biology, National Research Council (CNR-IBBC), International Campus A. Buzzati Traverso, Via E. Ramarini 32, Monterotondo, 00015 Rome, Italy
- Correspondence: ; Tel.: +39-06-90091357
| | - Elena Fico
- Institute of Biochemistry and Cell Biology, National Research Council (CNR-IBBC), at Department of Sense Organs, University of Rome “ La Sapienza”, Viale del Policlinico 155, 00161 Rome, Italy; (E.F.); (M.T.C.); (D.M.); (P.T.)
| | - Valentina Sposato
- European Brain Research Institute (EBRI Foundation), Viale Regina Elena 295, 00161 Rome, Italy; (V.S.); (P.C.)
| | - Silvia Caioli
- IRCCS S. Lucia Foundation, Via del Fosso di Fiorano 64, 00143 Rome, Italy; (S.C.); (C.Z.)
| | - Maria Teresa Ciotti
- Institute of Biochemistry and Cell Biology, National Research Council (CNR-IBBC), at Department of Sense Organs, University of Rome “ La Sapienza”, Viale del Policlinico 155, 00161 Rome, Italy; (E.F.); (M.T.C.); (D.M.); (P.T.)
| | - Cristina Zona
- IRCCS S. Lucia Foundation, Via del Fosso di Fiorano 64, 00143 Rome, Italy; (S.C.); (C.Z.)
- Department of Systems Medicine, University of Rome “TorVergata”, Via Montpellier 1, 00133 Rome, Italy
| | - Delio Mercanti
- Institute of Biochemistry and Cell Biology, National Research Council (CNR-IBBC), at Department of Sense Organs, University of Rome “ La Sapienza”, Viale del Policlinico 155, 00161 Rome, Italy; (E.F.); (M.T.C.); (D.M.); (P.T.)
| | - Diego La Mendola
- Department of Pharmacy, University of Pisa, via Bonanno Pisano 6, 56126 Pisa, Italy;
| | - Cristina Satriano
- Department of Chemical Sciences, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy; (C.S.); (E.R.)
| | - Enrico Rizzarelli
- Department of Chemical Sciences, University of Catania, Viale Andrea Doria 6, 95125 Catania, Italy; (C.S.); (E.R.)
- Institute of Crystallography, National Research Council (CNR-IC), Via Paolo Gaifami 18, 95126 Catania, Italy
| | - Paola Tirassa
- Institute of Biochemistry and Cell Biology, National Research Council (CNR-IBBC), at Department of Sense Organs, University of Rome “ La Sapienza”, Viale del Policlinico 155, 00161 Rome, Italy; (E.F.); (M.T.C.); (D.M.); (P.T.)
| | - Pietro Calissano
- European Brain Research Institute (EBRI Foundation), Viale Regina Elena 295, 00161 Rome, Italy; (V.S.); (P.C.)
| |
Collapse
|
43
|
Griso O, Puccio H. Primary Cultures of Pure Embryonic Dorsal Root Ganglia Sensory Neurons as a New Cellular Model for Friedreich's Ataxia. Methods Mol Biol 2020; 2056:241-253. [PMID: 31586352 DOI: 10.1007/978-1-4939-9784-8_15] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Peripheral neuropathies can have various origins, from genetic to acquired causes, and affect altogether a large group of people in the world. Current available therapies aim at helping the disease symptoms but not to correct or stop the development of the disease. Primary neuronal cultures represent an essential tool in the study of events related to peripheral neuropathies as they allow to isolate the affected cell types, often originating in complex tissues in which they account for only a few percentage of cells. They provide a powerful system to identifying or testing compounds with potential therapeutic effect in the treatment of those diseases. Friedreich's ataxia is an autosomal recessive neurodegenerative disorder, which is characterized by a progressive spinocerebellar and sensory ataxia. Proprioceptive neurons of the dorsal root ganglia (DRG) are the primary affected cells. The disease is triggered by a mutation in the gene FXN which leads to a reduction of the frataxin protein. In order to study the neurophysiopathology of the disease at the cellular and molecular levels, we have established a model of primary cultures of DRG sensory neurons in which we induce the loss of the frataxin protein. With such a model we can alleviate the issues related to the complexity of DRG tissues and low amount of sensory neuron material in adult mouse. Hereby, we provide a protocol of detailed and optimized methods to obtain high yield of healthy mouse DRG sensory neuron in culture.
Collapse
Affiliation(s)
- Olivier Griso
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France
- INSERM, U1258, Illkirch, France
- CNRS, UMR7104, Illkirch, France
- Université de Strasbourg, Strasbourg, France
| | - Hélène Puccio
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France.
- INSERM, U1258, Illkirch, France.
- CNRS, UMR7104, Illkirch, France.
- Université de Strasbourg, Strasbourg, France.
| |
Collapse
|
44
|
Shulha O, Çiçek SS, Wangensteen H, Kroes J, Mäder M, Girreser U, Sendker J, Jöhrer K, Greil R, Schühly W, Mangoni A, Grauso L, van Thriel C, Zidorn C. Lignans and sesquiterpene lactones from Hypochaeris radicata subsp. neapolitana (Asteraceae, Cichorieae). PHYTOCHEMISTRY 2019; 165:112047. [PMID: 31203102 DOI: 10.1016/j.phytochem.2019.05.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 05/24/2019] [Accepted: 05/30/2019] [Indexed: 06/09/2023]
Abstract
Four undescribed lignans and two undescribed sesquiterpenic acids, together with three known compounds (hypochoeroside C, hypochoeroside D, and 5-O-caffeoylshikimic acid) were isolated from the roots of Hypochaeris radicata subsp. neapolitana (Asteraceae, Cichorieae). The lignans were identified as 4-(3,4-dihydroxybenzyl)-2-(3,4-dihydroxyphenyl)tetrahydrofuran-3-carboxy-O-β-D-glucopyranoside, 4-(3,4-dihydroxybenzyl)-2-(3,4-dihydroxyphenyl)tetrahydrofuran-3-carboxy-O-β-D-glucopyranosyl-2'-O-methacrylate, (7S,8R,8'R)-7-(3,4-dihydroxyphenyl)-3',4'-dihydroxy-7,8,7',8'-tetrahydronaphtho [8,8'-c]furan-1(3H)-one, and (7S,8R,8'R)-7-(3,4-dihydroxyphenyl)-3',4'-dihydroxy-8'-(hydroxymethyl)-7,8,7',8'-tetrahydronaphthalen-8-carboxylic acid. The two sesquiterpenic acids were identified as the ring open precursors of hypochoerosides C and D. Structures were elucidated using NMR and HRMS. Absolute configurations of (7S,8R,8'R)-7-(3,4-dihydroxyphenyl)-3',4'-dihydroxy-7,8,7',8'-tetrahydronaphtho [8,8'-c]furan-1(3H)-one and (7S,8R,8'R)-7-(3,4-dihydroxyphenyl)-3',4'-dihydroxy-8'-(hydroxymethyl)-7,8,7',8'-tetrahydronaphthalen-8-carboxylic acid were determined using electronic circular dichroism (ECD) spectroscopy. 4-(3,4-dihydroxybenzyl)-2-(3,4-dihydroxyphenyl)tetrahydrofuran-3-carboxy-O-β-D-glucopyranoside was evaluated for its anti-proliferative activity against myeloma cell lines MM1S, U266, and NCI-H929 and showed cytotoxicity at 100 mM against MM1S strain. No neurotoxicity was observed for major compounds 4-(3,4-dihydroxybenzyl)-2-(3,4-dihydroxyphenyl)tetrahydrofuran-3-carboxy-O-β-D-glucopyranoside, hypochoeroside C, and hypochoeroside D in a fluorescence assay measuring neurite outgrowth in dorsal root ganglion (DRG) neurons. Additionally, compounds 4-(3,4-dihydroxybenzyl)-2-(3,4-dihydroxyphenyl)tetrahydrofuran-3-carboxy-O-β-D-glucopyranoside, hypochoeroside C, hypochoeroside D, and hypochoerosidic acid D were quantified in unstressed and drought-stressed plants using HPLC-DAD. Drought-stressed plants were found to contain lower concentrations of the lignan 4-(3,4-dihydroxybenzyl)-2-(3,4-dihydroxyphenyl)tetrahydrofuran-3-carboxy-O-β-D-glucopyranoside and sesquiterpene lactone hypochoeroside C.
Collapse
Affiliation(s)
- Oleksandr Shulha
- Pharmazeutisches Institut, Abteilung Pharmazeutische Biologie, Christian-Albrechts-Universität zu Kiel, Gutenbergstraße 76, 24118, Kiel, Germany
| | - Serhat Sezai Çiçek
- Pharmazeutisches Institut, Abteilung Pharmazeutische Biologie, Christian-Albrechts-Universität zu Kiel, Gutenbergstraße 76, 24118, Kiel, Germany
| | - Helle Wangensteen
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of Oslo, P.O. Box 1068, 0316, Oslo, Norway
| | - Janina Kroes
- Leibniz-Institut für Arbeitsforschung, TU Dortmund, Ardeystraße 67, 44139, Dortmund, Germany
| | - Malte Mäder
- Pharmazeutisches Institut, Abteilung Pharmazeutische Chemie, Christian-Albrechts-Universität zu Kiel, Gutenbergstraße 76, 24118, Kiel, Germany
| | - Ulrich Girreser
- Pharmazeutisches Institut, Abteilung Pharmazeutische Chemie, Christian-Albrechts-Universität zu Kiel, Gutenbergstraße 76, 24118, Kiel, Germany
| | - Jandirk Sendker
- Institut für Pharmazeutische Biologie und Phytochemie, Universiät Münster, Corrensstraße 48, 48149, Münster, Germany
| | - Karin Jöhrer
- Tyrolean Cancer Research Institute, Innrain 66, 6020, Innsbruck, Austria
| | - Richard Greil
- Tyrolean Cancer Research Institute, Innrain 66, 6020, Innsbruck, Austria; Paracelsus Medical University Salzburg, Department of Internal Medicine III, Laboratory for Immunological and Molecular Cancer Research, Müllner Hauptstraße 48, 5020, Salzburg, Austria
| | - Wolfgang Schühly
- Institut für Zoologie, Universität Graz, Universitätsplatz 2/I, 8010, Graz, Austria
| | - Alfonso Mangoni
- Dipartimento di Farmacia, Università di Napoli Federico II, Via Domenico Montesano 49, 80131, Napoli, Italy
| | - Laura Grauso
- Dipartimento di Agraria, Università di Napoli Federico II, Via Università 100, 80055, Portici (NA), Italy
| | - Christoph van Thriel
- Leibniz-Institut für Arbeitsforschung, TU Dortmund, Ardeystraße 67, 44139, Dortmund, Germany
| | - Christian Zidorn
- Pharmazeutisches Institut, Abteilung Pharmazeutische Biologie, Christian-Albrechts-Universität zu Kiel, Gutenbergstraße 76, 24118, Kiel, Germany.
| |
Collapse
|
45
|
Ita ME, Winkelstein BA. Concentration-Dependent Effects of Fibroblast-Like Synoviocytes on Collagen Gel Multiscale Biomechanics and Neuronal Signaling: Implications for Modeling Human Ligamentous Tissues. J Biomech Eng 2019; 141:091013. [PMID: 31209465 PMCID: PMC6808009 DOI: 10.1115/1.4044051] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 06/12/2019] [Indexed: 12/13/2022]
Abstract
Abnormal loading of a joint's ligamentous capsule causes pain by activating the capsule's nociceptive afferent fibers, which reside in the capsule's collagenous matrix alongside fibroblast-like synoviocytes (FLS) and transmit pain to the dorsal root ganglia (DRG). This study integrated FLS into a DRG-collagen gel model to better mimic the anatomy and physiology of human joint capsules; using this new model, the effect of FLS on multiscale biomechanics and cell physiology under load was investigated. Primary FLS cells were co-cultured with DRGs at low or high concentrations, to simulate variable anatomical FLS densities, and failed in tension. Given their roles in collagen degradation and nociception, matrix-metalloproteinase (MMP-1) and neuronal expression of the neurotransmitter substance P were probed after gel failure. The amount of FLS did not alter (p > 0.3) the gel failure force, displacement, or stiffness. FLS doubled regional strains at both low (p < 0.01) and high (p = 0.01) concentrations. For high FLS, the collagen network showed more reorganization at failure (p < 0.01). Although total MMP-1 and neuronal substance P were the same regardless of FLS concentration before loading, protein expression of both increased after failure, but only in low FLS gels (p ≤ 0.02). The concentration-dependent effect of FLS on microstructure and cellular responses implies that capsule regions with different FLS densities experience variable microenvironments. This study presents a novel DRG-FLS co-culture collagen gel system that provides a platform for investigating the complex biomechanics and physiology of human joint capsules, and is the first relating DRG and FLS interactions between each other and their surrounding collagen network.
Collapse
Affiliation(s)
- Meagan E Ita
- Department of Bioengineering,University of Pennsylvania,240 Skirkanich Hall, 210 South 33rd Street,Philadelphia, PA 19104e-mail:
| | - Beth A Winkelstein
- Mem. ASMEDepartment of Bioengineering,University of Pennsylvania, 240 Skirkanich Hall, 210 South 33rd Street,Philadelphia, PA 19104
- Department of Neurosurgery,University of Pennsylvania,240 Skirkanich Hall, 210 South 33rd Street,Philadelphia, PA 19104e-mail:
| |
Collapse
|
46
|
Magadmi R, Meszaros J, Damanhouri ZA, Seward EP. Secretion of Mast Cell Inflammatory Mediators Is Enhanced by CADM1-Dependent Adhesion to Sensory Neurons. Front Cell Neurosci 2019; 13:262. [PMID: 31275114 PMCID: PMC6591473 DOI: 10.3389/fncel.2019.00262] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 05/24/2019] [Indexed: 01/05/2023] Open
Abstract
Neuroimmune interactions are important in the pathophysiology of many chronic inflammatory diseases, particularly those associated with alterations in sensory processing and pain. Mast cells and sensory neuron nerve endings are found in areas of the body exposed to the external environment, both are specialized to sense potential damage by injury or pathogens and signal to the immune system and nervous system, respectively, to elicit protective responses. Cell adhesion molecule 1 (CADM1), also known as SynCAM1, has previously been identified as an adhesion molecule which may couple mast cells to sensory neurons however, whether this molecule exerts a functional as well as structural role in neuroimmune cross-talk is unknown. Here we show, using a newly developed in vitro co-culture system consisting of murine bone marrow derived mast cells (BMMC) and adult sensory neurons isolated from dorsal root ganglions (DRG), that CADM1 is expressed in mast cells and adult sensory neurons and mediates strong adhesion between the two cell types. Non-neuronal cells in the DRG cultures did not express CADM1, and mast cells did not adhere to them. The interaction of BMMCs with sensory neurons was found to induce mast cell degranulation and IL-6 secretion and to enhance responses to antigen stimulation and activation of FcεRI receptors. Secretion of TNFα in contrast was not affected, nor was secretion evoked by compound 48/80. Co-cultures of BMMCs with HEK 293 cells, which also express CADM1, while also leading to adhesion did not replicate the effects of sensory neurons on mast cells, indicative of a neuron-specific interaction. Application of a CADM1 blocking peptide or knockdown of CADM1 in BMMCs significantly decreased BMMC attachment to sensory neurites and abolished the enhanced secretory responses of mast cells. In conclusion, CADM1 is necessary and sufficient to drive mast cell-sensory neuron adhesion and promote the development of a microenvironment in which neurons enhance mast cell responsiveness to antigen, this interaction could explain why the incidence of painful neuroinflammatory disorders such as irritable bowel syndrome (IBS) are increased in atopic patients.
Collapse
Affiliation(s)
- Rania Magadmi
- Department of Biomedical Science, University of Sheffield, Sheffield, United Kingdom.,Department of Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Judit Meszaros
- Department of Biomedical Science, University of Sheffield, Sheffield, United Kingdom
| | - Zoheir A Damanhouri
- Department of Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Elizabeth P Seward
- Department of Biomedical Science, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
47
|
Booth DG, Beckett AJ, Prior IA, Meijer D. SuperCLEM: an accessible correlative light and electron microscopy approach for investigation of neurons and glia in vitro. Biol Open 2019; 8:bio.042085. [PMID: 31110056 PMCID: PMC6550067 DOI: 10.1242/bio.042085] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The rapid evolution of super-resolution light microscopy has narrowed the gap between light and electron microscopy, allowing the imaging of molecules and cellular structures at high resolution within their normal cellular and tissue context. Multimodal imaging approaches such as correlative light electron microscopy (CLEM) combine these techniques to create a tool with unique imaging capacity. However, these approaches are typically reserved for specialists, and their application to the analysis of neural tissue is challenging. Here we present SuperCLEM, a relatively simple approach that combines super-resolution fluorescence light microscopy (FLM), 3D electron microscopy (3D-EM) and rendering into 3D models. We demonstrate our workflow using neuron-glia cultures from which we first acquire high-resolution fluorescent light images of myelinated axons. After resin embedding and re-identification of the region of interest, serially aligned EM sections are acquired and imaged using a serial block face scanning electron microscope (SBF-SEM). The FLM and 3D-EM datasets are then combined to render 3D models of the myelinated axons. Thus, the SuperCLEM imaging pipeline is a useful new tool for researchers pursuing similar questions in neuronal and other complex tissue culture systems. Summary: We developed an advanced, accessible and adaptable correlative light and electron microscopy pipeline for study of neuron-glia cultures and demonstrate its power by deriving accurate 3D models of myelinated axons.
Collapse
Affiliation(s)
- Daniel G Booth
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Alison J Beckett
- Biomedical Electron Microscopy Unit, Department of Molecular and Cellular Biology, University of Liverpool, Crown Street, Liverpool L69 3BX, UK
| | - Ian A Prior
- Biomedical Electron Microscopy Unit, Department of Molecular and Cellular Biology, University of Liverpool, Crown Street, Liverpool L69 3BX, UK
| | - Dies Meijer
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| |
Collapse
|
48
|
Jiang Y, Parameswaran R, Li X, Carvalho-de-Souza JL, Gao X, Meng L, Bezanilla F, Shepherd GMG, Tian B. Nongenetic optical neuromodulation with silicon-based materials. Nat Protoc 2019; 14:1339-1376. [PMID: 30980031 PMCID: PMC6557640 DOI: 10.1038/s41596-019-0135-9] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 01/10/2019] [Indexed: 01/13/2023]
Abstract
Optically controlled nongenetic neuromodulation represents a promising approach for the fundamental study of neural circuits and the clinical treatment of neurological disorders. Among the existing material candidates that can transduce light energy into biologically relevant cues, silicon (Si) is particularly advantageous due to its highly tunable electrical and optical properties, ease of fabrication into multiple forms, ability to absorb a broad spectrum of light, and biocompatibility. This protocol describes a rational design principle for Si-based structures, general procedures for material synthesis and device fabrication, a universal method for evaluating material photoresponses, detailed illustrations of all instrumentation used, and demonstrations of optically controlled nongenetic modulation of cellular calcium dynamics, neuronal excitability, neurotransmitter release from mouse brain slices, and brain activity in the mouse brain in vivo using the aforementioned Si materials. The entire procedure takes ~4-8 d in the hands of an experienced graduate student, depending on the specific biological targets. We anticipate that our approach can also be adapted in the future to study other systems, such as cardiovascular tissues and microbial communities.
Collapse
Affiliation(s)
- Yuanwen Jiang
- Department of Chemistry, The University of Chicago, Chicago, IL, USA.
- The James Franck Institute, The University of Chicago, Chicago, IL, USA.
| | - Ramya Parameswaran
- The Graduate Program in Biophysical Sciences, The University of Chicago, Chicago, IL, USA
| | - Xiaojian Li
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | | | - Xiang Gao
- The James Franck Institute, The University of Chicago, Chicago, IL, USA
| | - Lingyuan Meng
- Insitute for Molecular Engineering, The University of Chicago, Chicago, IL, USA
| | - Francisco Bezanilla
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL, USA
- Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL, USA
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Gordon M G Shepherd
- Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Bozhi Tian
- Department of Chemistry, The University of Chicago, Chicago, IL, USA.
- The James Franck Institute, The University of Chicago, Chicago, IL, USA.
- Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
49
|
Mobini S, Song YH, McCrary MW, Schmidt CE. Advances in ex vivo models and lab-on-a-chip devices for neural tissue engineering. Biomaterials 2019; 198:146-166. [PMID: 29880219 PMCID: PMC6957334 DOI: 10.1016/j.biomaterials.2018.05.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 04/25/2018] [Accepted: 05/07/2018] [Indexed: 02/08/2023]
Abstract
The technologies related to ex vivo models and lab-on-a-chip devices for studying the regeneration of brain, spinal cord, and peripheral nerve tissues are essential tools for neural tissue engineering and regenerative medicine research. The need for ex vivo systems, lab-on-a-chip technologies and disease models for neural tissue engineering applications are emerging to overcome the shortages and drawbacks of traditional in vitro systems and animal models. Ex vivo models have evolved from traditional 2D cell culture models to 3D tissue-engineered scaffold systems, bioreactors, and recently organoid test beds. In addition to ex vivo model systems, we discuss lab-on-a-chip devices and technologies specifically for neural tissue engineering applications. Finally, we review current commercial products that mimic diseased and normal neural tissues, and discuss the future directions in this field.
Collapse
Affiliation(s)
- Sahba Mobini
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Young Hye Song
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Michaela W McCrary
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Christine E Schmidt
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
50
|
D’Amato AR, Puhl DL, Ziemba AM, Johnson CDL, Doedee J, Bao J, Gilbert RJ. Exploring the effects of electrospun fiber surface nanotopography on neurite outgrowth and branching in neuron cultures. PLoS One 2019; 14:e0211731. [PMID: 30716106 PMCID: PMC6361509 DOI: 10.1371/journal.pone.0211731] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 01/18/2019] [Indexed: 12/19/2022] Open
Abstract
Three aligned, electrospun fiber scaffolds with unique surface features were created from poly-L-lactic acid (PLLA). Fibers without surface nanotopography (smooth fibers), fibers with surface divots (shallow pits), and fibers with surface pits (deeper pits) were fabricated, and fiber alignment, diameter, and density were characterized using scanning electron microscopy (SEM). Whole dorsal root ganglia (DRG) were isolated from rats and placed onto uncoated fibers or fibers coated with laminin. On uncoated fibers, neurite outgrowth was restricted by fibers displaying divoted or pitted nanotopography when compared to neurite outgrowth on smooth fibers. However, neurites extending from whole DRG cultured on laminin-coated fibers were not restricted by divoted or pitted surface nanotopography. Thus, neurites extending on laminin-coated fibers were able to extend long neurites even in the presence of surface divots or pits. To further explore this result, individual neurons isolated from dissociated DRG were seeded onto laminin-coated smooth, pitted, or divoted fibers. Interestingly, neurons on pitted or divoted fibers exhibited a 1.5-fold increase in total neurite length, and a 2.3 or 2.7-fold increase in neurite branching compared to neurons on smooth fibers, respectively. Based on these findings, we conclude that fiber roughness in the form of pits or divots can promote extension and branching of long neurites along aligned electrospun fibers in the presence of an extracellular matrix protein coating. Thus, aligned, electrospun fibers can be crafted to not only direct the extension of axons but to induce unique branching morphologies.
Collapse
Affiliation(s)
- Anthony R. D’Amato
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York,United States of America
| | - Devan L. Puhl
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York,United States of America
| | - Alexis M. Ziemba
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York,United States of America
| | - Christopher D. L. Johnson
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York,United States of America
| | - Janneke Doedee
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York,United States of America
| | - Jonathan Bao
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York,United States of America
| | - Ryan J. Gilbert
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York, United States of America
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York,United States of America
| |
Collapse
|