1
|
Chen R, Liu Y, Qian L, Yi M, Yin H, Wang S, Xiang B. Sodium channels as a new target for pain treatment. Front Pharmacol 2025; 16:1573254. [PMID: 40206072 PMCID: PMC11979154 DOI: 10.3389/fphar.2025.1573254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Accepted: 03/07/2025] [Indexed: 04/11/2025] Open
Abstract
Voltage-gated sodium channels, especially the Nav1.7, Nav1.8, and Nav1.9 subtypes, play a crucial role in the transmission of pain signals. Nav1.7 is considered a threshold channel that regulates the generation of action potentials and is closely associated with various hereditary pain disorders. Nav1.8 primarily participates in inflammatory and neuropathic pain within the peripheral nervous system. Its characteristic of not involving the central nervous system makes it a potential target for non-addictive analgesics. Nav1.9 has shown significant involvement in cold pain sensing and small fiber neuropathy, although its mechanism of action is still under further investigation. Currently, despite promising results from preclinical studies, sodium channel inhibitors have not fully met expectations in clinical trials due to issues such as drug selectivity, dosing, and safety. The development of Nav1.7 and Nav1.8 inhibitors faces challenges such as drug intolerance, insufficient target occupancy, and off-target side effects. Future research may promote the development of non-opioid analgesics through combined inhibition strategies targeting multiple Nav subtypes, as well as improving drug selectivity and bioavailability. Overall, sodium channel inhibitors remain a key area of research in pain management, but their clinical application prospects still require further exploration.
Collapse
Affiliation(s)
- Rui Chen
- Department of Anesthesiology, Chengdu Fifth People’s Hospital (The Second Clinical Medical College, Geriatric Diseases Institute of Chengdu/Cancer Prevention and Treatment Institute of Chengdu, Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, China
| | - Yiran Liu
- Nursing Department, Cujin Community Health Service Center, Chengdu, China
| | - Liu Qian
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Mingliang Yi
- Department of Anesthesiology, Chengdu Fifth People’s Hospital (The Second Clinical Medical College, Geriatric Diseases Institute of Chengdu/Cancer Prevention and Treatment Institute of Chengdu, Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, China
| | - Hong Yin
- Department of Anesthesiology, Chengdu Fifth People’s Hospital (The Second Clinical Medical College, Geriatric Diseases Institute of Chengdu/Cancer Prevention and Treatment Institute of Chengdu, Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, China
| | - Shun Wang
- Department of Anesthesiology, Clinical Medical College and The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Bingbing Xiang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
2
|
Köster PA, Leipold E, Tigerholm J, Maxion A, Namer B, Stiehl T, Lampert A. Nociceptor sodium channels shape subthreshold phase, upstroke, and shoulder of action potentials. J Gen Physiol 2025; 157:e202313526. [PMID: 39836077 PMCID: PMC11748974 DOI: 10.1085/jgp.202313526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 09/30/2024] [Accepted: 12/18/2024] [Indexed: 01/22/2025] Open
Abstract
Voltage-gated sodium channels (VGSCs) in the peripheral nervous system shape action potentials (APs) and thereby support the detection of sensory stimuli. Most of the nine mammalian VGSC subtypes are expressed in nociceptors, but predominantly, three are linked to several human pain syndromes: while Nav1.7 is suggested to be a (sub-)threshold channel, Nav1.8 is thought to support the fast AP upstroke. Nav1.9, as it produces large persistent currents, is attributed a role in determining the resting membrane potential. We characterized the gating of Nav1.1-Nav1.3 and Nav1.5-Nav1.9 in manual patch clamp with a focus on the AP subthreshold depolarization phase. Nav1.9 exhibited the most hyperpolarized activation, while its fast inactivation resembled the depolarized inactivation of Nav1.8. For some VGSCs (e.g., Nav1.1 and Nav1.2), a positive correlation between ramp current and window current was detected. Using a modified Hodgkin-Huxley model that accounts for the time needed for inactivation to occur, we used the acquired data to simulate two nociceptive nerve fiber types (an Aδ- and a mechano-insensitive C-nociceptor) containing VGSC conductances according to published human RNAseq data. Our simulations suggest that Nav1.9 is supporting both the AP upstroke and its shoulder. A reduced threshold for AP generation was induced by enhancing Nav1.7 conductivity or shifting its activation to more hyperpolarized potentials, as observed in Nav1.7-related pain disorders. Here, we provide a comprehensive, comparative functional characterization of VGSCs relevant in nociception and describe their gating with Hodgkin-Huxley-like models, which can serve as a tool to study their specific contributions to AP shape and sodium channel-related diseases.
Collapse
Affiliation(s)
- Phil Alexander Köster
- Institute for Neurophysiology, Uniklinik RWTH Aachen University, Aachen, Germany
- Scientific Center for Neuropathic Pain Aachen SCN, Uniklinik RWTH Aachen University, Aachen, Germany
| | - Enrico Leipold
- Department of Anesthesiology and Intensive Care and CBBM-Center of Brain, Behavior and Metabolism, University of Luebeck, Lübeck, Germany
| | - Jenny Tigerholm
- Scientific Center for Neuropathic Pain Aachen SCN, Uniklinik RWTH Aachen University, Aachen, Germany
- Joint Research Center for Computational Biomedicine (JRCC), Uniklinik RWTH Aachen University, Aachen, Germany
| | - Anna Maxion
- Institute for Neurophysiology, Uniklinik RWTH Aachen University, Aachen, Germany
- Scientific Center for Neuropathic Pain Aachen SCN, Uniklinik RWTH Aachen University, Aachen, Germany
- Interdisciplinary Center for Clinical Research (IZKF), Faculty of Medicine, Research Group Neurosciences, Uniklinik RWTH Aachen University, Aachen, Germany
| | - Barbara Namer
- Institute for Neurophysiology, Uniklinik RWTH Aachen University, Aachen, Germany
- Scientific Center for Neuropathic Pain Aachen SCN, Uniklinik RWTH Aachen University, Aachen, Germany
- Interdisciplinary Center for Clinical Research (IZKF), Faculty of Medicine, Research Group Neurosciences, Uniklinik RWTH Aachen University, Aachen, Germany
| | - Thomas Stiehl
- Scientific Center for Neuropathic Pain Aachen SCN, Uniklinik RWTH Aachen University, Aachen, Germany
- Joint Research Center for Computational Biomedicine (JRCC), Uniklinik RWTH Aachen University, Aachen, Germany
- Institute for Computational Biomedicine and Disease Modelling With Focus on Phase Transitions Between Phenotypes, Uniklinik RWTH Aachen University, Aachen, Germany
| | - Angelika Lampert
- Institute for Neurophysiology, Uniklinik RWTH Aachen University, Aachen, Germany
- Scientific Center for Neuropathic Pain Aachen SCN, Uniklinik RWTH Aachen University, Aachen, Germany
| |
Collapse
|
3
|
Zhang B, Shi X, Liu X, Liu Y, Li X, Wang Q, Huang D, Zhao W, Cui J, Cao Y, Chai X, Wang J, Zhang Y, Wang X, Jia Q. Discovery of E0199: A novel compound targeting both peripheral Na V and K V7 channels to alleviate neuropathic pain. J Pharm Anal 2025; 15:101132. [PMID: 39906690 PMCID: PMC11791318 DOI: 10.1016/j.jpha.2024.101132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 10/07/2024] [Accepted: 10/22/2024] [Indexed: 02/06/2025] Open
Abstract
This research study focuses on addressing the limitations of current neuropathic pain (NP) treatments by developing a novel dual-target modulator, E0199, targeting both NaV1.7, NaV1.8, and NaV1.9 and KV7 channels, a crucial regulator in controlling NP symptoms. The objective of the study was to synthesize a compound capable of modulating these channels to alleviate NP. Through an experimental design involving both in vitro and in vivo methods, E0199 was tested for its efficacy on ion channels and its therapeutic potential in a chronic constriction injury (CCI) mouse model. The results demonstrated that E0199 significantly inhibited NaV1.7, NaV1.8, and NaV1.9 channels with a particularly low half maximal inhibitory concentration (IC50) for NaV1.9 by promoting sodium channel inactivation, and also effectively increased KV7.2/7.3, KV7.2, and KV7.5 channels, excluding KV7.1 by promoting potassium channel activation. This dual action significantly reduced the excitability of dorsal root ganglion neurons and alleviated pain hypersensitivity in mice at low doses, indicating a potent analgesic effect without affecting heart and skeletal muscle ion channels critically. The safety of E0199 was supported by neurobehavioral evaluations. Conclusively, E0199 represents a ground-breaking approach in NP treatment, showcasing the potential of dual-target small-molecule compounds in providing a more effective and safe therapeutic option for NP. This study introduces a promising direction for the future development of NP therapeutics.
Collapse
Affiliation(s)
- Boxuan Zhang
- Department of Pharmaceutical Chemistry, College of Pharmacy, Hebei Medical University, Shijiazhuang, 050017, China
| | - Xiaoxing Shi
- Department of Pharmacology, College of Basic Medical, Hebei Medical University, Shijiazhuang, 050017, China
| | - Xingang Liu
- Department of Pharmaceutical Chemistry, College of Pharmacy, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yan Liu
- Department of Pharmaceutical Experimental Teaching Center, College of Pharmacy, Hebei Medical University, Shijiazhuang, 050017, China
| | - Xuedong Li
- Department of Pharmaceutical Chemistry, College of Pharmacy, Hebei Medical University, Shijiazhuang, 050017, China
| | - Qi Wang
- Shijiazhuang Xianyu Digital Biotechnology Co., Ltd., College of Software, Hebei Normal University, Shijiazhuang, 050024, China
| | - Dongyang Huang
- Department of Pharmacology, College of Basic Medical, Hebei Medical University, Shijiazhuang, 050017, China
| | - Weidong Zhao
- Department of Pharmacology, College of Basic Medical, Hebei Medical University, Shijiazhuang, 050017, China
| | - Junru Cui
- The Center for New Drug Safety Evaluation and Research, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yawen Cao
- Department of Pharmacology, College of Basic Medical, Hebei Medical University, Shijiazhuang, 050017, China
| | - Xu Chai
- Department of Pharmacology, College of Basic Medical, Hebei Medical University, Shijiazhuang, 050017, China
| | - Jiahao Wang
- Department of Pharmacology, College of Basic Medical, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yang Zhang
- Department of Pharmaceutical Chemistry, College of Pharmacy, Hebei Medical University, Shijiazhuang, 050017, China
| | - Xiangyu Wang
- Hebei Medical University Postdoctoral Mobile Station of Basic Medical, Hebei Medical University, Shijiazhuang, 050017, China
- Departments of Clinic Pharmacy, College of Pharmacy, Hebei Medical University, Shijiazhuang, 050017, China
| | - Qingzhong Jia
- Department of Pharmaceutical Chemistry, College of Pharmacy, Hebei Medical University, Shijiazhuang, 050017, China
| |
Collapse
|
4
|
Que Z, Olivero-Acosta MI, Robinson M, Chen I, Zhang J, Wettschurack K, Wu J, Xiao T, Otterbacher CM, Shankar V, Harlow H, Hong S, Zirkle B, Wang M, Cui N, Mandal P, Chen X, Deming B, Halurkar M, Zhao Y, Rochet JC, Xu R, Brewster AL, Wu LJ, Yuan C, Skarnes WC, Yang Y. Human iPSC-derived microglia sense and dampen hyperexcitability of cortical neurons carrying the epilepsy-associated SCN2A-L1342P mutation. J Neurosci 2024; 45:e2027232024. [PMID: 39557580 PMCID: PMC11735681 DOI: 10.1523/jneurosci.2027-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 10/16/2024] [Accepted: 11/08/2024] [Indexed: 11/20/2024] Open
Abstract
Neuronal hyperexcitability is a hallmark of epilepsy. It has been recently shown in rodent models of seizures that microglia, the brain's resident immune cells, can respond to and modulate neuronal excitability. However, how human microglia interact with human neurons to regulate hyperexcitability mediated by an epilepsy-causing genetic mutation found in patients is unknown. The SCN2A gene is responsible for encoding the voltage-gated sodium channel Nav1.2, one of the leading contributors to monogenic epilepsies. Previously, we demonstrated that the recurring Nav1.2-L1342P mutation leads to hyperexcitability in a male donor (KOLF2.1) hiPSC-derived cortical neuron model. Microglia originate from a different lineage (yolk sac) and are not naturally present in hiPSCs-derived neuronal cultures. To study how microglia respond to neurons carrying a disease-causing mutation and influence neuronal excitability, we established a co-culture model comprising hiPSC-derived neurons and microglia. We found that microglia display increased branch length and enhanced process-specific calcium signal when co-cultured with Nav1.2-L1342P neurons. Moreover, the presence of microglia significantly lowered the repetitive action potential firing and current density of sodium channels in neurons carrying the mutation. Additionally, we showed that co-culturing with microglia led to a reduction in sodium channel expression within the axon initial segment of Nav1.2-L1342P neurons. Furthermore, we demonstrated that Nav1.2-L1342P neurons release a higher amount of glutamate compared to control neurons. Our work thus reveals a critical role of human iPSCs-derived microglia in sensing and dampening hyperexcitability mediated by an epilepsy-causing mutation.Significance Statement Seizure studies in mouse models have highlighted the role of microglia in modulating neuronal activity, particularly in the promotion or suppression of seizures. However, a gap persists in comprehending the influence of human microglia on intrinsically hyperexcitable neurons carrying epilepsy-associated pathogenic mutations. This research addresses this gap by investigating human microglia and their impact on neuronal functions. Our findings demonstrate that microglia exhibit dynamic morphological alterations and calcium fluctuations in the presence of neurons carrying an epilepsy-associated SCN2A mutation. Furthermore, microglia suppressed the excitability of hyperexcitable neurons, suggesting a potential beneficial role. This study underscores the role of microglia in the regulation of abnormal neuronal activity, providing insights into therapeutic strategies for neurological conditions associated with hyperexcitability.
Collapse
Affiliation(s)
- Zhefu Que
- Borsch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907
- Purdue Institute for Integrative Neuroscience (PIIN), Purdue University, West Lafayette, Indiana 47907
| | - Maria I. Olivero-Acosta
- Borsch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907
- Purdue Institute for Integrative Neuroscience (PIIN), Purdue University, West Lafayette, Indiana 47907
| | - Morgan Robinson
- Borsch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907
- Purdue Institute for Integrative Neuroscience (PIIN), Purdue University, West Lafayette, Indiana 47907
- Department of Chemical Engineering, Purdue University, West Lafayette, Indiana 47907
| | - Ian Chen
- Borsch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907
- Purdue Institute for Integrative Neuroscience (PIIN), Purdue University, West Lafayette, Indiana 47907
| | - Jingliang Zhang
- Borsch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907
- Purdue Institute for Integrative Neuroscience (PIIN), Purdue University, West Lafayette, Indiana 47907
| | - Kyle Wettschurack
- Borsch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907
- Purdue Institute for Integrative Neuroscience (PIIN), Purdue University, West Lafayette, Indiana 47907
| | - Jiaxiang Wu
- Borsch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907
- Purdue Institute for Integrative Neuroscience (PIIN), Purdue University, West Lafayette, Indiana 47907
| | - Tiange Xiao
- Borsch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907
- Purdue Institute for Integrative Neuroscience (PIIN), Purdue University, West Lafayette, Indiana 47907
| | - Conrad Max Otterbacher
- Borsch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907
- Purdue Institute for Integrative Neuroscience (PIIN), Purdue University, West Lafayette, Indiana 47907
| | - Vinayak Shankar
- Borsch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907
- Purdue Institute for Integrative Neuroscience (PIIN), Purdue University, West Lafayette, Indiana 47907
| | - Hope Harlow
- Borsch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907
- Purdue Institute for Integrative Neuroscience (PIIN), Purdue University, West Lafayette, Indiana 47907
| | - Seoyong Hong
- Borsch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907
- Purdue Institute for Integrative Neuroscience (PIIN), Purdue University, West Lafayette, Indiana 47907
| | - Benjamin Zirkle
- Borsch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907
- Purdue Institute for Integrative Neuroscience (PIIN), Purdue University, West Lafayette, Indiana 47907
| | - Muhan Wang
- Borsch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907
- Purdue Institute for Integrative Neuroscience (PIIN), Purdue University, West Lafayette, Indiana 47907
| | - Ningren Cui
- Borsch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907
- Purdue Institute for Integrative Neuroscience (PIIN), Purdue University, West Lafayette, Indiana 47907
| | - Purba Mandal
- Borsch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907
- Purdue Institute for Integrative Neuroscience (PIIN), Purdue University, West Lafayette, Indiana 47907
| | - Xiaoling Chen
- Borsch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907
- Purdue Institute for Integrative Neuroscience (PIIN), Purdue University, West Lafayette, Indiana 47907
| | - Brody Deming
- Borsch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907
- Purdue Institute for Integrative Neuroscience (PIIN), Purdue University, West Lafayette, Indiana 47907
| | - Manasi Halurkar
- Borsch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907
- Purdue Institute for Integrative Neuroscience (PIIN), Purdue University, West Lafayette, Indiana 47907
| | - Yuanrui Zhao
- Borsch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907
- Purdue Institute for Integrative Neuroscience (PIIN), Purdue University, West Lafayette, Indiana 47907
| | - Jean-Christophe Rochet
- Borsch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907
- Purdue Institute for Integrative Neuroscience (PIIN), Purdue University, West Lafayette, Indiana 47907
| | - Ranjie Xu
- Purdue University College of Veterinary Medicine, West Lafayette, Indiana 47907
| | - Amy L. Brewster
- Department of Biological Sciences, Southern Methodist University, Dallas, Texas 75205
| | - Long-jun Wu
- Department of Neurology, Mayo Clinic, Rochester, Minnesota 55905
| | - Chongli Yuan
- Department of Chemical Engineering, Purdue University, West Lafayette, Indiana 47907
| | - William C. Skarnes
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut 06032
| | - Yang Yang
- Borsch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907
- Purdue Institute for Integrative Neuroscience (PIIN), Purdue University, West Lafayette, Indiana 47907
| |
Collapse
|
5
|
Zebochin I, Denk F, Nochi Z. Modeling neuropathic pain in a dish. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 179:233-278. [PMID: 39580214 DOI: 10.1016/bs.irn.2024.10.003] [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: 11/25/2024]
Abstract
The study of pain mechanisms has advanced significantly with the development of innovative in vitro models. This chapter explores those already used in or potentially useful for neuropathic pain research, emphasizing the complementary roles of animal and human cellular models to enhance translational success. Traditional animal models have provided foundational insights into the neurobiology of pain and remain invaluable for understanding complex pain pathways. However, integrating human cellular models addresses the need for better replication of human nociceptors. The chapter details methodologies for culturing rodent and human primary sensory neurons, including isolation and culture techniques, advantages, and limitations. It highlights the application of these models in neuropathic pain research, such as identifying pain-associated receptors and ion channels. Recent advancements in using induced pluripotent stem cell (iPSC)-derived sensory neurons are also discussed. Finally, the chapter explores advanced in vitro models, including 2D co-cultures and 3D organoids, and their implications for studying neuropathic pain. These models offer significant advantages for drug screening and ethical research practices, providing a more accurate representation of human pain pathways and paving the way for innovative therapeutic strategies. Despite challenges such as limited access to viable human tissue and variability between samples, these in vitro models, alongside traditional animal models, are indispensable for advancing our understanding of neuropathic pain and developing effective treatments.
Collapse
Affiliation(s)
- Irene Zebochin
- Wolfson Sensory Pain and Regeneration Centre (SPaRC), King's College London
| | - Franziska Denk
- Wolfson Sensory Pain and Regeneration Centre (SPaRC), King's College London
| | - Zahra Nochi
- Danish Pain Research Centre, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
6
|
Syben A, Weber S, Appelmann I, Rolke R. [N-Check: nerve check to document chemotherapy-induced peripheral neuropathy (CIPN) in incurable cancer]. Schmerz 2024:10.1007/s00482-024-00828-8. [PMID: 39313711 DOI: 10.1007/s00482-024-00828-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/05/2024] [Indexed: 09/25/2024]
Abstract
Chemotherapy-induced polyneuropathy (CIPN) encompasses a spectrum of symptoms ranging from hypoesthesia with impaired gait, stance and fine motor skills to painful dysesthesia and allodynia and significantly impairs the quality of life of those affected. In the present pilot study, quantitative sensory testing (QST) was used to investigate CIPN as a common adverse effect of cytostatic drugs in patients with incurable cancer. The QST is a standardized examination procedure that is not yet routinely used in cancer patients. It is used to examine thermal and mechanical perception and pain thresholds to record the subjectively experienced pain phenotype. In the N‑Check pilot project, the QST was used before and after tumor-specific, potentially CIPN-inducing treatment and the data collected was compared in a pre-post analysis. In addition, the specific effects of CIPN on the health-related quality of life of patients treated primarily with a palliative intention were recorded using the Functional Assessment for Cancer Therapy-General (FACT-G) questionnaire. Overall, the patients showed significant heat hypoalgesia after chemotherapy as a sign of damage to small nerve fibers. In addition, there were signs of deterioration of the quality of life. The feasibility of QST in patients with incurable cancer and palliative, neurotoxic chemotherapy was demonstrated in this pilot study.
Collapse
Affiliation(s)
- Annabell Syben
- Klinik für Palliativmedizin, Uniklinik der RWTH, Medizinische Fakultät, RWTH Aachen University, Pauwelsstraße 30, 52074, Aachen, Deutschland
| | - Sascha Weber
- Klinik für Palliativmedizin, Uniklinik der RWTH, Medizinische Fakultät, RWTH Aachen University, Pauwelsstraße 30, 52074, Aachen, Deutschland
- Klinik für Psychiatrie, Psychotherapie und Psychosomatik, Medizinische Fakultät, RWTH Aachen University, Aachen, Deutschland
| | - Iris Appelmann
- Klinik für Palliativmedizin, Uniklinik der RWTH, Medizinische Fakultät, RWTH Aachen University, Pauwelsstraße 30, 52074, Aachen, Deutschland
| | - Roman Rolke
- Klinik für Palliativmedizin, Uniklinik der RWTH, Medizinische Fakultät, RWTH Aachen University, Pauwelsstraße 30, 52074, Aachen, Deutschland.
| |
Collapse
|
7
|
Kesdoğan AB, Neureiter A, Gaebler AJ, Kalia AK, Körner J, Lampert A. Analgesic effect of Botulinum toxin in neuropathic pain is sodium channel independent. Neuropharmacology 2024; 253:109967. [PMID: 38657946 DOI: 10.1016/j.neuropharm.2024.109967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 02/26/2024] [Accepted: 04/20/2024] [Indexed: 04/26/2024]
Abstract
Botulinum neurotoxin type A BoNT/A is used off-label as a third line therapy for neuropathic pain. However, the mechanism of action remains unclear. In recent years, the role of voltage-gated sodium channels (Nav) in neuropathic pain became evident and it was suggested that block of sodium channels by BoNT/A would contribute to its analgesic effect. We assessed sodium channel function in the presence of BoNT/A in heterologously expressed Nav1.7, Nav1.3, and the neuronal cell line ND7/23 by high throughput automated and manual patch-clamp. We used both the full protein and the isolated catalytic light chain LC/A for acute or long-term extracellular or intracellular exposure. To assess the toxin's effect in a human cellular system, we differentiated induced pluripotent stem cells (iPSC) into sensory neurons from a healthy control and a patient suffering from a hereditary neuropathic pain syndrome (inherited erythromelalgia) carrying the Nav1.7/p.Q875E-mutation and carried out multielectrode-array measurements. Both BoNT/A and the isolated catalytic light chain LC/A showed limited effects in heterologous expression systems and the neuronal cell line ND7/23. Spontaneous activity in iPSC derived sensory neurons remained unaltered upon BoNT/A exposure both in neurons from the healthy control and the mutation carrying patient. BoNT/A may not specifically be beneficial in pain syndromes linked to sodium channel variants. The favorable effects of BoNT/A in neuropathic pain are likely based on mechanisms other than sodium channel blockage and new approaches to understand BoNT/A's therapeutic effects are necessary.
Collapse
Affiliation(s)
- Aylin B Kesdoğan
- Institute of Neurophysiology, RWTH Aachen University Hospital, Pauwelsstr. 30, 52074, Aachen, Germany; Scientific Center for Neuropathic Pain Research Aachen, SCN(Aachen), RWTH Aachen University Hospital, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Anika Neureiter
- Institute of Neurophysiology, RWTH Aachen University Hospital, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Arnim J Gaebler
- Institute of Neurophysiology, RWTH Aachen University Hospital, Pauwelsstr. 30, 52074, Aachen, Germany; Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, RWTH Aachen University Hospital, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Anil K Kalia
- Institute of Neurophysiology, RWTH Aachen University Hospital, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Jannis Körner
- Institute of Neurophysiology, RWTH Aachen University Hospital, Pauwelsstr. 30, 52074, Aachen, Germany; Department of Anesthesiology, Medical Faculty, RWTH Aachen University Hospital, Pauwelsstr. 30, 52074, Aachen, Germany; Department of Intensive and Intermediate Care, Medical Faculty, RWTH Aachen University Hospital, Pauwelsstr. 30, 52074, Aachen, Germany; Scientific Center for Neuropathic Pain Research Aachen, SCN(Aachen), RWTH Aachen University Hospital, Pauwelsstr. 30, 52074, Aachen, Germany.
| | - Angelika Lampert
- Institute of Neurophysiology, RWTH Aachen University Hospital, Pauwelsstr. 30, 52074, Aachen, Germany; Scientific Center for Neuropathic Pain Research Aachen, SCN(Aachen), RWTH Aachen University Hospital, Pauwelsstr. 30, 52074, Aachen, Germany
| |
Collapse
|
8
|
Chuinsiri N, Siraboriphantakul N, Kendall L, Yarova P, Nile CJ, Song B, Obara I, Durham J, Telezhkin V. Calcium-sensing receptor regulates Kv7 channels via G i/o protein signalling and modulates excitability of human induced pluripotent stem cell-derived nociceptive-like neurons. Br J Pharmacol 2024; 181:2676-2696. [PMID: 38627101 DOI: 10.1111/bph.16349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 01/29/2024] [Accepted: 02/15/2024] [Indexed: 07/03/2024] Open
Abstract
BACKGROUND AND PURPOSE Neuropathic pain, a debilitating condition with unmet medical needs, can be characterised as hyperexcitability of nociceptive neurons caused by dysfunction of ion channels. Voltage-gated potassium channels type 7 (Kv7), responsible for maintaining neuronal resting membrane potential and thus excitability, reside under tight control of G protein-coupled receptors (GPCRs). Calcium-sensing receptor (CaSR) is a GPCR that regulates the activity of numerous ion channels, but whether CaSR can control Kv7 channel function has been unexplored until now. EXPERIMENTAL APPROACH Experiments were conducted in recombinant cell models, mouse dorsal root ganglia (DRG) neurons and human induced pluripotent stem cell (hiPSC)-derived nociceptive-like neurons using patch-clamp electrophysiology and molecular biology techniques. KEY RESULTS Our results demonstrate that CaSR is expressed in recombinant cell models, hiPSC-derived nociceptive-like neurons and mouse DRG neurons, and its activation induced depolarisation via Kv7.2/7.3 channel inhibition. The CaSR-Kv7.2/7.3 channel crosslink was mediated via the Gi/o protein-adenylate cyclase-cyclicAMP-protein kinase A signalling cascade. Suppression of CaSR function demonstrated a potential to rescue hiPSC-derived nociceptive-like neurons from algogenic cocktail-induced hyperexcitability. CONCLUSION AND IMPLICATIONS This study demonstrates that the CaSR-Kv7.2/7.3 channel crosslink, via a Gi/o protein signalling pathway, effectively regulates neuronal excitability, providing a feasible pharmacological target for neuronal hyperexcitability management in neuropathic pain.
Collapse
Affiliation(s)
- Nontawat Chuinsiri
- School of Dental Sciences, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- Institute of Dentistry, Suranaree University of Technology, Nakhon Ratchasima, Thailand
- Oral Health Center, Suranaree University of TechnologyHospital, Suranaree University of Technology, Nakhon Ratchasima, Thailand
| | | | - Luke Kendall
- School of Dental Sciences, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Polina Yarova
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Christopher J Nile
- School of Dental Sciences, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Bing Song
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Cardiff, UK
| | - Ilona Obara
- School of Pharmacy, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Justin Durham
- School of Dental Sciences, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Vsevolod Telezhkin
- School of Dental Sciences, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
9
|
Lee KH, Kim UJ, Cha M, Lee BH. Inhibiting Nav1.7 channels in pulpitis: An in vivo study on neuronal hyperexcitability. Biochem Biophys Res Commun 2024; 717:150044. [PMID: 38718567 DOI: 10.1016/j.bbrc.2024.150044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 04/30/2024] [Indexed: 05/21/2024]
Abstract
Pulpitis constitutes a significant challenge in clinical management due to its impact on peripheral nerve tissue and the persistence of chronic pain. Despite its clinical importance, the correlation between neuronal activity and the expression of voltage-gated sodium channel 1.7 (Nav1.7) in the trigeminal ganglion (TG) during pulpitis is less investigated. The aim of this study was to examine the relationship between experimentally induced pulpitis and Nav1.7 expression in the TG and to investigate the potential of selective Nav1.7 modulation to attenuate TG abnormal activity associated with pulpitis. Acute pulpitis was induced at the maxillary molar (M1) using allyl isothiocyanate (AITC). The mice were divided into three groups: control, pulpitis model, and pulpitis model treated with ProTx-II, a selective Nav1.7 channel inhibitor. After three days following the surgery, we conducted a recording and comparative analysis of the neural activity of the TG utilizing in vivo optical imaging. Then immunohistochemistry and Western blot were performed to assess changes in the expression levels of extracellular signal-regulated kinase (ERK), c-Fos, collapsin response mediator protein-2 (CRMP2), and Nav1.7 channels. The optical imaging result showed significant neurological excitation in pulpitis TGs. Nav1.7 expressions exhibited upregulation, accompanied by signaling molecular changes suggestive of inflammation and neuroplasticity. In addition, inhibition of Nav1.7 led to reduced neural activity and subsequent decreases in ERK, c-Fos, and CRMP2 levels. These findings suggest the potential for targeting overexpressed Nav1.7 channels to alleviate pain associated with pulpitis, providing practical pain management strategies.
Collapse
Affiliation(s)
- Kyung Hee Lee
- Department of Dental Hygiene, Division of Health Science, Dongseo University, Busan, 47011, Republic of Korea
| | - Un Jeng Kim
- Department of Physiology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Myeounghoon Cha
- Department of Physiology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
| | - Bae Hwan Lee
- Department of Physiology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea; Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
| |
Collapse
|
10
|
Liu Y, Balaji R, de Toledo MAS, Ernst S, Hautvast P, Kesdoğan AB, Körner J, Zenke M, Neureiter A, Lampert A. The pain target Na V1.7 is expressed late during human iPS cell differentiation into sensory neurons as determined in high-resolution imaging. Pflugers Arch 2024; 476:975-992. [PMID: 38538988 PMCID: PMC11139713 DOI: 10.1007/s00424-024-02945-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 02/16/2024] [Accepted: 03/11/2024] [Indexed: 06/01/2024]
Abstract
Human-induced pluripotent stem cells (iPS cells) are efficiently differentiated into sensory neurons. These cells express the voltage-gated sodium channel NaV1.7, which is a validated pain target. NaV1.7 deficiency leads to pain insensitivity, whereas NaV1.7 gain-of-function mutants are associated with chronic pain. During differentiation, the sensory neurons start spontaneous action potential firing around day 22, with increasing firing rate until day 40. Here, we used CRISPR/Cas9 genome editing to generate a HA-tag NaV1.7 to follow its expression during differentiation. We used two protocols to generate sensory neurons: the classical small molecule approach and a directed differentiation methodology and assessed surface NaV1.7 expression by Airyscan high-resolution microscopy. Our results show that maturation of at least 49 days is necessary to observe robust NaV1.7 surface expression in both protocols. Electric activity of the sensory neurons precedes NaV1.7 surface expression. A clinically effective NaV1.7 blocker is still missing, and we expect this iPS cell model system to be useful for drug discovery and disease modeling.
Collapse
Affiliation(s)
- Yi Liu
- Institute of Neurophysiology, Uniklinik RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Rachna Balaji
- Institute of Neurophysiology, Uniklinik RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Marcelo A Szymanski de Toledo
- Department of Hematology, Oncology, and Stem Cell Transplantation, Uniklinik RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Sabrina Ernst
- Confocal Microscopy Facility, Interdisciplinary Center for Clinical Research IZKF, Uniklinik RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Petra Hautvast
- Institute of Neurophysiology, Uniklinik RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Aylin B Kesdoğan
- Institute of Neurophysiology, Uniklinik RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Jannis Körner
- Institute of Neurophysiology, Uniklinik RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany
- Department of Anaesthesiology, Uniklinik RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany
- Department of Intensive and Intermediate Care, Uniklinik RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany
- Scientific Center for Neuropathic Pain Research Aachen, SCN-Aachen Uniklinik RWTH Aachen, Aachen, Germany
| | - Martin Zenke
- Department of Hematology, Oncology, and Stem Cell Transplantation, Uniklinik RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Anika Neureiter
- Institute of Neurophysiology, Uniklinik RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany.
| | - Angelika Lampert
- Institute of Neurophysiology, Uniklinik RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany.
- Scientific Center for Neuropathic Pain Research Aachen, SCN-Aachen Uniklinik RWTH Aachen, Aachen, Germany.
| |
Collapse
|
11
|
Zurek NA, Ehsanian R, Goins AE, Adams IM, Petersen T, Goyal S, Shilling M, Westlund KN, Alles SRA. Electrophysiological Analyses of Human Dorsal Root Ganglia and Human Induced Pluripotent Stem Cell-derived Sensory Neurons From Male and Female Donors. THE JOURNAL OF PAIN 2024; 25:104451. [PMID: 38154622 PMCID: PMC11128351 DOI: 10.1016/j.jpain.2023.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/15/2023] [Accepted: 12/16/2023] [Indexed: 12/30/2023]
Abstract
Human induced pluripotent stem cell-derived sensory neurons (hiPSC-SNs) and human dorsal root ganglia neurons (hDRG-N) are popular tools in the field of pain research; however, few groups make use of both approaches. For screening and analgesic validation purposes, important characterizations can be determined of the similarities and differences between hDRG-N and hiPSC-SNs. This study focuses specifically on the electrophysiology properties of hDRG-N in comparison to hiPSC-SNs. We also compared hDRG-N and hiPSC-SNs from both male and female donors to evaluate potential sex differences. We recorded neuronal size, rheobase, resting membrane potential, input resistance, and action potential waveform properties from 83 hiPSCs-SNs (2 donors) and 108 hDRG-N neurons (8 donors). We observed several statistically significant electrophysiological differences between hDRG-N and hiPSC-SNs, such as size, rheobase, input resistance, and several action potential waveform properties. Correlation analysis also revealed many properties that were positively or negatively correlated, some of which were differentially correlated between hDRG-N and hiPSC-SNs. This study shows several differences between hDRG-N and hiPSC-SNs and allows a better understanding of the advantages and disadvantages of both for use in pain research. We hope this study will be a valuable resource for pain researchers considering the use of these human in vitro systems for mechanistic studies and/or drug development projects. PERSPECTIVE: hiPSC-SNs and hDRG-N are popular tools in the field of pain research. This study allows for a better functional understanding of the pros and cons of both tools.
Collapse
Affiliation(s)
- Nesia A Zurek
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Reza Ehsanian
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Aleyah E Goins
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Ian M Adams
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Timothy Petersen
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Sachin Goyal
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Mark Shilling
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Karin N Westlund
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Sascha R A Alles
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico.
| |
Collapse
|
12
|
Scheliga S, Dohrn MF, Habel U, Lampert A, Rolke R, Lischka A, van den Braak N, Spehr M, Jo HG, Kellermann T. Reduced Gray Matter Volume and Cortical Thickness in Patients With Small-Fiber Neuropathy. THE JOURNAL OF PAIN 2024; 25:104457. [PMID: 38211845 DOI: 10.1016/j.jpain.2024.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 12/08/2023] [Accepted: 01/02/2024] [Indexed: 01/13/2024]
Abstract
Small-fiber neuropathy (SFN) is defined by degeneration or dysfunction of peripheral sensory nerve endings. Central correlates have been identified on the level of gray matter volume (GMV) and cortical thickness (CT) changes. However, across SFN etiologies knowledge about a common structural brain signature is still lacking. Therefore, we recruited 26 SFN patients and 25 age- and sex-matched healthy controls to conduct voxel-based- and surface-based morphometry. Across all patients, we found reduced GMV in widespread frontal regions, left caudate, insula and superior parietal lobule. Surface-based morphometry analysis revealed reduced CT in the right precentral gyrus of SFN patients. In a region-based approach, patients had reduced GMV in the left caudate. Since pathogenic gain-of-function variants in voltage-gated sodium channels (Nav) have been associated with SFN pathophysiology, we explored brain morphological patterns in a homogenous subsample of patients carrying rare heterozygous missense variants. Whole brain- and region-based approaches revealed GMV reductions in the bilateral caudate for Nav variant carriers. Further research is needed to analyze the specific role of Nav variants for structural brain alterations. Together, we conclude that SFN patients have specific GMV and CT alterations, potentially forming potential new central biomarkers for this condition. Our results might help to better understand underlying or compensatory mechanisms of chronic pain perception in the future. PERSPECTIVE: This study reveals structural brain changes in small-fiber neuropathy (SFN) patients, particularly in frontal regions, caudate, insula, and parietal lobule. Notably, individuals with SFN and specific Nav variants exhibit bilateral caudate abnormalities. These findings may serve as potential central biomarkers for SFN and provide insights into chronic pain perception mechanisms.
Collapse
Affiliation(s)
- Sebastian Scheliga
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty RWTH Aachen University, Aachen, Germany
| | - Maike F Dohrn
- Department of Neurology, Medical Faculty RWTH Aachen University, Aachen, Germany
| | - Ute Habel
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty RWTH Aachen University, Aachen, Germany; Institute of Neuroscience and Medicine: JARA-Institute Brain Structure Function Relationship (INM 10), Research Center Jülich, Jülich, Germany
| | - Angelika Lampert
- Institute of Neurophysiology, Medical Faculty RWTH Aachen University, Aachen, Germany
| | - Roman Rolke
- Department of Palliative Medicine, Medical Faculty RWTH Aachen University, Aachen, Germany
| | - Annette Lischka
- Institute for Human Genetics and Genomic Medicine, Medical Faculty RWTH Aachen University, Aachen, Germany
| | | | - Marc Spehr
- Department of Chemosensation, RWTH Aachen University, Institute for Biology II, Aachen, Germany
| | - Han-Gue Jo
- School of Computer Information and Communication Engineering, Kunsan National University, Gunsan, South Korea
| | - Thilo Kellermann
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty RWTH Aachen University, Aachen, Germany; Institute of Neuroscience and Medicine: JARA-Institute Brain Structure Function Relationship (INM 10), Research Center Jülich, Jülich, Germany
| |
Collapse
|
13
|
Kalia AK, Rösseler C, Granja-Vazquez R, Ahmad A, Pancrazio JJ, Neureiter A, Zhang M, Sauter D, Vetter I, Andersson A, Dussor G, Price TJ, Kolber BJ, Truong V, Walsh P, Lampert A. How to differentiate induced pluripotent stem cells into sensory neurons for disease modelling: a functional assessment. Stem Cell Res Ther 2024; 15:99. [PMID: 38581069 PMCID: PMC10998320 DOI: 10.1186/s13287-024-03696-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 03/13/2024] [Indexed: 04/07/2024] Open
Abstract
BACKGROUND Human induced pluripotent stem cell (iPSC)-derived peripheral sensory neurons present a valuable tool to model human diseases and are a source for applications in drug discovery and regenerative medicine. Clinically, peripheral sensory neuropathies can result in maladies ranging from a complete loss of pain to severe painful neuropathic disorders. Sensory neurons are located in the dorsal root ganglion and are comprised of functionally diverse neuronal types. Low efficiency, reproducibility concerns, variations arising due to genetic factors and time needed to generate functionally mature neuronal populations from iPSCs remain key challenges to study human nociception in vitro. Here, we report a detailed functional characterization of iPSC-derived sensory neurons with an accelerated differentiation protocol ("Anatomic" protocol) compared to the most commonly used small molecule approach ("Chambers" protocol). Anatomic's commercially available RealDRG™ were further characterized for both functional and expression phenotyping of key nociceptor markers. METHODS Multiple iPSC clones derived from different reprogramming methods, genetics, age, and somatic cell sources were used to generate sensory neurons. Manual patch clamp was used to functionally characterize both control and patient-derived neurons. High throughput techniques were further used to demonstrate that RealDRGs™ derived from the Anatomic protocol are amenable to high throughput technologies for disease modelling. RESULTS The Anatomic protocol rendered a purer culture without the use of mitomycin C to suppress non-neuronal outgrowth, while Chambers differentiations yielded a mix of cell types. Chambers protocol results in predominantly tonic firing when compared to Anatomic protocol. Patient-derived nociceptors displayed higher frequency firing compared to control subject with both, Chambers and Anatomic differentiation approaches, underlining their potential use for clinical phenotyping as a disease-in-a-dish model. RealDRG™ sensory neurons show heterogeneity of nociceptive markers indicating that the cells may be useful as a humanized model system for translational studies. CONCLUSIONS We validated the efficiency of two differentiation protocols and their potential application for functional assessment and thus understanding the disease mechanisms from patients suffering from pain disorders. We propose that both differentiation methods can be further exploited for understanding mechanisms and development of novel treatments in pain disorders.
Collapse
Affiliation(s)
- Anil Kumar Kalia
- Institute of Neurophysiology, Uniklinik RWTH Aachen University, Pauwelsstr. 30, 52074, Aachen, Germany
- Research Training Group 2416 MultiSenses-MultiScales, RWTH Aachen University, Aachen, Germany
| | - Corinna Rösseler
- Institute of Neurophysiology, Uniklinik RWTH Aachen University, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Rafael Granja-Vazquez
- Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, 75080, USA
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Ayesha Ahmad
- Department of Neuroscience, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Joseph J Pancrazio
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Anika Neureiter
- Institute of Neurophysiology, Uniklinik RWTH Aachen University, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Mei Zhang
- Sophion Bioscience Inc., Bedford, MA, 01730, USA
| | | | - Irina Vetter
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, 4072, Australia
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD, 4102, Australia
| | - Asa Andersson
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Gregory Dussor
- Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, 75080, USA
- Department of Neuroscience, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Theodore J Price
- Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, 75080, USA
- Department of Neuroscience, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Benedict J Kolber
- Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, 75080, USA
- Department of Neuroscience, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Vincent Truong
- Anatomic Incorporated, 2112 Broadway Street NE #135, Minneapolis, MN, 55413, USA
| | - Patrick Walsh
- Anatomic Incorporated, 2112 Broadway Street NE #135, Minneapolis, MN, 55413, USA
| | - Angelika Lampert
- Institute of Neurophysiology, Uniklinik RWTH Aachen University, Pauwelsstr. 30, 52074, Aachen, Germany.
- Research Training Group 2416 MultiSenses-MultiScales, RWTH Aachen University, Aachen, Germany.
- Scientific Center for Neuropathic Pain Aachen - SCN-Aachen, Uniklinik RWTH Aachen University, 52074, Aachen, Germany.
| |
Collapse
|
14
|
Gomez K, Allen HN, Duran P, Loya-Lopez S, Calderon-Rivera A, Moutal A, Tang C, Nelson TS, Perez-Miller S, Khanna R. Targeted transcriptional upregulation of SENP1 by CRISPR activation enhances deSUMOylation pathways to elicit antinociception in the spinal nerve ligation model of neuropathic pain. Pain 2024; 165:866-883. [PMID: 37862053 PMCID: PMC11389604 DOI: 10.1097/j.pain.0000000000003080] [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: 05/09/2023] [Accepted: 08/04/2023] [Indexed: 10/21/2023]
Abstract
ABSTRACT The voltage-gated sodium channel Na V 1.7 is an essential component of human pain signaling. Changes in Na V 1.7 trafficking are considered critical in the development of neuropathic pain. SUMOylation of collapsin response mediator protein 2 (CRMP2) regulates the membrane trafficking and function of Na V 1.7. Enhanced CRMP2 SUMOylation in neuropathic pain correlates with increased Na V 1.7 activity. Pharmacological and genetic interventions that interfere with CRMP2 SUMOylation in rodents with neuropathic pain have been shown to reverse mechanical allodynia. Sentrin or SUMO-specific proteases (SENPs) are vital for balancing SUMOylation and deSUMOylation of substrates. Overexpression of SENP1 and/or SENP2 in CRMP2-expressing cells results in increased deSUMOylation and decreased membrane expression and currents of Na V 1.7. Although SENP1 is present in the spinal cord and dorsal root ganglia, its role in regulating Na V 1.7 function and pain is not known. We hypothesized that favoring SENP1 expression can enhance CRMP2 deSUMOylation to modulate Na V 1.7 channels. In this study, we used a clustered regularly interspaced short palindromic repeats activation (CRISPRa) SENP1 lentivirus to overexpress SENP1 in dorsal root ganglia neurons. We found that SENP1 lentivirus reduced CRMP2 SUMOylation, Na V 1.7-CRMP2 interaction, and Na V 1.7 membrane expression. SENP1 overexpression decreased Na V 1.7 currents through clathrin-mediated endocytosis, directly linked to CRMP2 deSUMOylation. Moreover, enhancing SENP1 expression did not affect the activity of TRPV1 channels or voltage-gated calcium and potassium channels. Intrathecal injection of CRISPRa SENP1 lentivirus reversed mechanical allodynia in male and female rats with spinal nerve injury. These results provide evidence that the pain-regulating effects of SENP1 overexpression involve, in part, the modulation of Na V 1.7 channels through the indirect mechanism of CRMP2 deSUMOylation.
Collapse
Affiliation(s)
- Kimberly Gomez
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, United States
- NYU Pain Research Center, New York, NY, United States
| | - Heather N Allen
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, United States
- NYU Pain Research Center, New York, NY, United States
| | - Paz Duran
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, United States
- NYU Pain Research Center, New York, NY, United States
| | - Santiago Loya-Lopez
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, United States
- NYU Pain Research Center, New York, NY, United States
| | - Aida Calderon-Rivera
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, United States
- NYU Pain Research Center, New York, NY, United States
| | - Aubin Moutal
- School of Medicine, Department of Pharmacology and Physiology, Saint Louis University, Saint Louis, MO, United States
| | - Cheng Tang
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, United States
- NYU Pain Research Center, New York, NY, United States
| | - Tyler S Nelson
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, United States
- NYU Pain Research Center, New York, NY, United States
| | - Samantha Perez-Miller
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, United States
- NYU Pain Research Center, New York, NY, United States
| | - Rajesh Khanna
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, United States
- NYU Pain Research Center, New York, NY, United States
- Department of Neuroscience and Physiology and Neuroscience Institute, School of Medicine, New York University, New York, NY, United States
| |
Collapse
|
15
|
Loya-Lopez SI, Allen HN, Duran P, Calderon-Rivera A, Gomez K, Kumar U, Shields R, Zeng R, Dwivedi A, Saurabh S, Korczeniewska OA, Khanna R. Intranasal CRMP2-Ubc9 inhibitor regulates Na V 1.7 to alleviate trigeminal neuropathic pain. Pain 2024; 165:573-588. [PMID: 37751532 PMCID: PMC10922202 DOI: 10.1097/j.pain.0000000000003053] [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: 07/11/2023] [Accepted: 07/25/2023] [Indexed: 09/28/2023]
Abstract
ABSTRACT Dysregulation of voltage-gated sodium Na V 1.7 channels in sensory neurons contributes to chronic pain conditions, including trigeminal neuropathic pain. We previously reported that chronic pain results in part from increased SUMOylation of collapsin response mediator protein 2 (CRMP2), leading to an increased CRMP2/Na V 1.7 interaction and increased functional activity of Na V 1.7. Targeting this feed-forward regulation, we developed compound 194 , which inhibits CRMP2 SUMOylation mediated by the SUMO-conjugating enzyme Ubc9. We further demonstrated that 194 effectively reduces the functional activity of Na V 1.7 channels in dorsal root ganglia neurons and alleviated inflammatory and neuropathic pain. Here, we used a comprehensive array of approaches, encompassing biochemical, pharmacological, genetic, electrophysiological, and behavioral analyses, to assess the functional implications of Na V 1.7 regulation by CRMP2 in trigeminal ganglia (TG) neurons. We confirmed the expression of Scn9a , Dpysl2 , and UBE2I within TG neurons. Furthermore, we found an interaction between CRMP2 and Na V 1.7, with CRMP2 being SUMOylated in these sensory ganglia. Disrupting CRMP2 SUMOylation with compound 194 uncoupled the CRMP2/Na V 1.7 interaction, impeded Na V 1.7 diffusion on the plasma membrane, and subsequently diminished Na V 1.7 activity. Compound 194 also led to a reduction in TG neuron excitability. Finally, when intranasally administered to rats with chronic constriction injury of the infraorbital nerve, 194 significantly decreased nociceptive behaviors. Collectively, our findings underscore the critical role of CRMP2 in regulating Na V 1.7 within TG neurons, emphasizing the importance of this indirect modulation in trigeminal neuropathic pain.
Collapse
Affiliation(s)
- Santiago I. Loya-Lopez
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York, United States of America
- NYU Pain Research Center, 433 First Avenue, New York, NY 10010, United States of America
| | - Heather N. Allen
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York, United States of America
- NYU Pain Research Center, 433 First Avenue, New York, NY 10010, United States of America
| | - Paz Duran
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York, United States of America
- NYU Pain Research Center, 433 First Avenue, New York, NY 10010, United States of America
| | - Aida Calderon-Rivera
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York, United States of America
- NYU Pain Research Center, 433 First Avenue, New York, NY 10010, United States of America
| | - Kimberly Gomez
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York, United States of America
- NYU Pain Research Center, 433 First Avenue, New York, NY 10010, United States of America
| | - Upasana Kumar
- Center for Orofacial Pain and Temporomandibular Disorders, Department of Diagnostic Sciences, Rutgers School of Dental Medicine, Newark, NJ 07101, United States of America
| | - Rory Shields
- Rutgers School of Graduate Studies, Newark Health Science Campus, Newark, NJ 07101, United States of America
| | - Rui Zeng
- Department of Chemistry, College of Arts and Sciences, New York University, 100 Washington Square East, New York, NY 10003, United States of America
| | - Akshat Dwivedi
- Department of Chemistry, College of Arts and Sciences, New York University, 100 Washington Square East, New York, NY 10003, United States of America
| | - Saumya Saurabh
- Department of Chemistry, College of Arts and Sciences, New York University, 100 Washington Square East, New York, NY 10003, United States of America
| | - Olga A. Korczeniewska
- Center for Orofacial Pain and Temporomandibular Disorders, Department of Diagnostic Sciences, Rutgers School of Dental Medicine, Newark, NJ 07101, United States of America
- Rutgers School of Graduate Studies, Newark Health Science Campus, Newark, NJ 07101, United States of America
| | - Rajesh Khanna
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York, United States of America
- NYU Pain Research Center, 433 First Avenue, New York, NY 10010, United States of America
- Department of Neuroscience and Physiology and Neuroscience Institute, School of Medicine, New York University, New York, NY, 10010, USA
| |
Collapse
|
16
|
Black BJ, Ghazal RE, Lojek N, Williams V, Rajput JS, Lawson JM. Phenotypic Screening of Prospective Analgesics Among FDA-Approved Compounds using an iPSC-Based Model of Acute and Chronic Inflammatory Nociception. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2303724. [PMID: 38189546 PMCID: PMC10953557 DOI: 10.1002/advs.202303724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 11/26/2023] [Indexed: 01/09/2024]
Abstract
Classical target-based drug screening is low-throughput, largely subjective, and costly. Phenotypic screening based on in vitro models is increasingly being used to identify candidate compounds that modulate complex cell/tissue functions. Chronic inflammatory nociception, and subsequent chronic pain conditions, affect peripheral sensory neuron activity (e.g., firing of action potentials) through myriad pathways, and remain unaddressed in regard to effective, non-addictive management/treatment options. Here, a chronic inflammatory nociception model is demonstrated based on induced pluripotent stem cell (iPSC) sensory neurons and glia, co-cultured on microelectrode arrays (MEAs). iPSC sensory co-cultures exhibit coordinated spontaneous extracellular action potential (EAP) firing, reaching a stable baseline after ≈27 days in vitro (DIV). Spontaneous and evoked EAP metrics are significantly modulated by 24-h incubation with tumor necrosis factor-alpha (TNF-α), representing an inflammatory phenotype. Compared with positive controls (lidocaine), this model is identified as an "excellent" stand-alone assay based on a modified Z' assay quality metric. This model is then used to screen 15 cherry-picked, off-label, Food and Drug Administration (FDA)-approved compounds; 10 of 15 are identified as "hits". Both hits and "misses" are discussed in turn. In total, this data suggests that iPSC sensory co-cultures on MEAs may represent a moderate-to-high-throughput assay for drug discovery targeting inflammatory nociception.
Collapse
Affiliation(s)
- Bryan James Black
- Department of Biomedical EngineeringFrancis College of EngineeringUniversity of Massachusetts LowellLowellMA01854USA
| | - Rasha El Ghazal
- Department of Biomedical EngineeringFrancis College of EngineeringUniversity of Massachusetts LowellLowellMA01854USA
| | - Neal Lojek
- Department of Biomedical EngineeringFrancis College of EngineeringUniversity of Massachusetts LowellLowellMA01854USA
| | - Victoria Williams
- Department of Biomedical EngineeringFrancis College of EngineeringUniversity of Massachusetts LowellLowellMA01854USA
| | - Jai Singh Rajput
- Department of Biomedical EngineeringFrancis College of EngineeringUniversity of Massachusetts LowellLowellMA01854USA
| | - Jennifer M. Lawson
- Department of Biomedical EngineeringFrancis College of EngineeringUniversity of Massachusetts LowellLowellMA01854USA
| |
Collapse
|
17
|
Zurek NA, Ehsanian R, Goins AE, Adams IM, Petersen T, Goyal S, Shilling M, Westlund KN, Alles SRA. Electrophysiological analyses of human dorsal root ganglia and human induced pluripotent stem cell-derived sensory neurons from male and female donors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.03.565343. [PMID: 37961669 PMCID: PMC10635102 DOI: 10.1101/2023.11.03.565343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Human induced pluripotent stem cell-derived sensory neurons (hiPSC-SNs) and human dorsal root ganglia (hDRG) neurons are popular tools in the field of pain research; however, few groups make use of both approaches. For screening and analgesic validation purposes, important characterizations can be determined of the similarities and differences between hDRG and hiPSC-SNs. This study focuses specifically on electrophysiology properties of hDRG in comparison to hiPSC-SNs. We also compared hDRG and hiPSC-SNs from both male and female donors to evaluate potential sex differences. We recorded neuronal size, rheobase, resting membrane potential, input resistance, and action potential waveform properties from 83 hiPSCs-SNs (2 donors) and 108 hDRG neurons (9 donors). We observed several statistically significant electrophysiological differences between hDRG and hiPSC-SNs, such as size, rheobase, input resistance, and several actional potential (AP) waveform properties. Correlation analysis also revealed many properties that were positively or negatively correlated, some of which were differentially correlated between hDRG and hiPSC-SNs. This study shows several differences between hDRG and hiPSC-SNs and allows better understanding of the advantages and disadvantages of both for use in pain research. We hope this study will be a valuable resource for pain researchers considering the use of these human in vitro systems for mechanistic studies and/or drug development projects.
Collapse
|
18
|
Kalia AK, Rösseler C, Granja-Vazquez R, Ahmad A, Pancrazio JJ, Neureiter A, Zhang M, Sauter D, Vetter I, Andersson A, Dussor G, Price TJ, Kolber BJ, Truong V, Walsh P, Lampert A. How to differentiate induced pluripotent stem cells into sensory neurons for disease modelling: a comparison of two protocols. RESEARCH SQUARE 2023:rs.3.rs-3127017. [PMID: 37961300 PMCID: PMC10635298 DOI: 10.21203/rs.3.rs-3127017/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Background Human induced pluripotent stem cell (iPSC)-derived peripheral sensory neurons present a valuable tool to model human diseases and are a source for applications in drug discovery and regenerative medicine. Clinically, peripheral sensory neuropathies can result in maladies ranging from a complete loss of pain to severe painful neuropathic symptoms. Sensory neurons are located in the dorsal root ganglion and are comprised of functionally diverse neuronal types. Low efficiency, reproducibility concerns, variations arising due to genetic factors and time needed to generate functionally mature neuronal populations from iPSCs for disease modelling remain key challenges to study human nociception in vitro. Here, we report a detailed characterization of iPSC-derived sensory neurons with an accelerated differentiation protocol ("Anatomic" protocol) compared to the most commonly used small molecule approach ("Chambers" protocol). Methods Multiple iPSC clones derived from different reprogramming methods, genetics, age, and somatic cell sources were used to generate sensory neurons. Expression profiling of sensory neurons was performed with Immunocytochemistry and in situ hybridization techniques. Manual patch clamp and high throughput cellular screening systems (Fluorescence imaging plate reader, automated patch clamp and multi-well microelectrode arrays recordings) were applied to functionally characterize the generated sensory neurons. Results The Anatomic protocol rendered a purer culture without the use of mitomycin C to suppress non-neuronal outgrowth, while Chambers differentiations yielded a mix of cell types. High throughput systems confirmed functional expression of Na+ and K+ ion channels. Multi-well microelectrode recordings display spontaneously active neurons with sensitivity to increased temperature indicating expression of heat sensitive ion channels. Patient-derived nociceptors displayed higher frequency firing compared to control subject with both, Chambers and Anatomic differentiation approaches, underlining their potential use for clinical phenotyping as a disease-in-a-dish model. Conclusions We validated the efficiency of two differentiation protocols and their potential application for understanding the disease mechanisms from patients suffering from pain disorders. We propose that both differentiation methods can be further exploited for understanding mechanisms and development of novel treatments in pain disorders.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Mei Zhang
- Sophion Bioscience A/S: Biolin Scientific AB
| | | | - Irina Vetter
- The University of Queensland Institute for Molecular Bioscience
| | - Asa Andersson
- The University of Queensland Institute for Molecular Bioscience
| | | | | | | | | | | | | |
Collapse
|
19
|
Smith PA. Neuropathic pain; what we know and what we should do about it. FRONTIERS IN PAIN RESEARCH 2023; 4:1220034. [PMID: 37810432 PMCID: PMC10559888 DOI: 10.3389/fpain.2023.1220034] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 09/05/2023] [Indexed: 10/10/2023] Open
Abstract
Neuropathic pain can result from injury to, or disease of the nervous system. It is notoriously difficult to treat. Peripheral nerve injury promotes Schwann cell activation and invasion of immunocompetent cells into the site of injury, spinal cord and higher sensory structures such as thalamus and cingulate and sensory cortices. Various cytokines, chemokines, growth factors, monoamines and neuropeptides effect two-way signalling between neurons, glia and immune cells. This promotes sustained hyperexcitability and spontaneous activity in primary afferents that is crucial for onset and persistence of pain as well as misprocessing of sensory information in the spinal cord and supraspinal structures. Much of the current understanding of pain aetiology and identification of drug targets derives from studies of the consequences of peripheral nerve injury in rodent models. Although a vast amount of information has been forthcoming, the translation of this information into the clinical arena has been minimal. Few, if any, major therapeutic approaches have appeared since the mid 1990's. This may reflect failure to recognise differences in pain processing in males vs. females, differences in cellular responses to different types of injury and differences in pain processing in humans vs. animals. Basic science and clinical approaches which seek to bridge this knowledge gap include better assessment of pain in animal models, use of pain models which better emulate human disease, and stratification of human pain phenotypes according to quantitative assessment of signs and symptoms of disease. This can lead to more personalized and effective treatments for individual patients. Significance statement: There is an urgent need to find new treatments for neuropathic pain. Although classical animal models have revealed essential features of pain aetiology such as peripheral and central sensitization and some of the molecular and cellular mechanisms involved, they do not adequately model the multiplicity of disease states or injuries that may bring forth neuropathic pain in the clinic. This review seeks to integrate information from the multiplicity of disciplines that seek to understand neuropathic pain; including immunology, cell biology, electrophysiology and biophysics, anatomy, cell biology, neurology, molecular biology, pharmacology and behavioral science. Beyond this, it underlines ongoing refinements in basic science and clinical practice that will engender improved approaches to pain management.
Collapse
Affiliation(s)
- Peter A. Smith
- Neuroscience and Mental Health Institute and Department of Pharmacology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
20
|
Gerardo‐Nava JL, Jansen J, Günther D, Klasen L, Thiebes AL, Niessing B, Bergerbit C, Meyer AA, Linkhorst J, Barth M, Akhyari P, Stingl J, Nagel S, Stiehl T, Lampert A, Leube R, Wessling M, Santoro F, Ingebrandt S, Jockenhoevel S, Herrmann A, Fischer H, Wagner W, Schmitt RH, Kiessling F, Kramann R, De Laporte L. Transformative Materials to Create 3D Functional Human Tissue Models In Vitro in a Reproducible Manner. Adv Healthc Mater 2023; 12:e2301030. [PMID: 37311209 PMCID: PMC11468549 DOI: 10.1002/adhm.202301030] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/21/2023] [Indexed: 06/15/2023]
Abstract
Recreating human tissues and organs in the petri dish to establish models as tools in biomedical sciences has gained momentum. These models can provide insight into mechanisms of human physiology, disease onset, and progression, and improve drug target validation, as well as the development of new medical therapeutics. Transformative materials play an important role in this evolution, as they can be programmed to direct cell behavior and fate by controlling the activity of bioactive molecules and material properties. Using nature as an inspiration, scientists are creating materials that incorporate specific biological processes observed during human organogenesis and tissue regeneration. This article presents the reader with state-of-the-art developments in the field of in vitro tissue engineering and the challenges related to the design, production, and translation of these transformative materials. Advances regarding (stem) cell sources, expansion, and differentiation, and how novel responsive materials, automated and large-scale fabrication processes, culture conditions, in situ monitoring systems, and computer simulations are required to create functional human tissue models that are relevant and efficient for drug discovery, are described. This paper illustrates how these different technologies need to converge to generate in vitro life-like human tissue models that provide a platform to answer health-based scientific questions.
Collapse
|
21
|
Loya-Lopez SI, Allen HN, Duran P, Calderon-Rivera A, Gomez K, Kumar U, Shields R, Zeng R, Dwivedi A, Saurabh S, Korczeniewska OA, Khanna R. Intranasal CRMP2-Ubc9 Inhibitor Regulates Na V 1.7 to Alleviate Trigeminal Neuropathic Pain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.16.549195. [PMID: 37502910 PMCID: PMC10370107 DOI: 10.1101/2023.07.16.549195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Dysregulation of voltage-gated sodium Na V 1.7 channels in sensory neurons contributes to chronic pain conditions, including trigeminal neuropathic pain. We previously reported that chronic pain results in part from increased SUMOylation of collapsin response mediator protein 2 (CRMP2), leading to an increased CRMP2/Na V 1.7 interaction and increased functional activity of Na V 1.7. Targeting this feed-forward regulation, we developed compound 194 , which inhibits CRMP2 SUMOylation mediated by the SUMO-conjugating enzyme Ubc9. We further demonstrated that 194 effectively reduces the functional activity of Na V 1.7 channels in dorsal root ganglia neurons and alleviated inflammatory and neuropathic pain. Here, we employed a comprehensive array of investigative approaches, encompassing biochemical, pharmacological, genetic, electrophysiological, and behavioral analyses, to assess the functional implications of Na V 1.7 regulation by CRMP2 in trigeminal ganglia (TG) neurons. We confirmed the expression of Scn9a , Dpysl2 , and UBE2I within TG neurons. Furthermore, we found an interaction between CRMP2 and Na V 1.7, with CRMP2 being SUMOylated in these sensory ganglia. Disrupting CRMP2 SUMOylation with compound 194 uncoupled the CRMP2/Na V 1.7 interaction, impeded Na V 1.7 diffusion on the plasma membrane, and subsequently diminished Na V 1.7 activity. Compound 194 also led to a reduction in TG neuron excitability. Finally, when intranasally administered to rats with chronic constriction injury of the infraorbital nerve (CCI-ION), 194 significantly decreased nociceptive behaviors. Collectively, our findings underscore the critical role of CRMP2 in regulating Na V 1.7 within TG neurons, emphasizing the importance of this indirect modulation in trigeminal neuropathic pain.
Collapse
|
22
|
Li L, Li P, Guo J, Wu Y, Zeng Q, Li N, Huang X, He Y, Ai W, Sun W, Liu T, Xiong D, Xiao L, Sun Y, Zhou Q, Kuang H, Wang Z, Jiang C. Up-regulation of oxytocin receptors on peripheral sensory neurons mediates analgesia in chemotherapy-induced neuropathic pain. Br J Pharmacol 2023. [PMID: 36702458 DOI: 10.1111/bph.16042] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 01/19/2023] [Accepted: 01/22/2023] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND AND PURPOSE Chemotherapy-induced neuropathic pain (CINP) currently has limited effective treatment. Although the roles of oxytocin (OXT) and the oxytocin receptor (OXTR) in central analgesia have been well documented, the expression and function of OXTR in the peripheral nervous system remain unclear. Here, we evaluated the peripheral antinociceptive profiles of OXTR in CINP. EXPERIMENTAL APPROACH Paclitaxel (PTX) was used to establish CINP. Quantitative real-time polymerase chain reaction (qRT-PCR), in situ hybridization, and immunohistochemistry were used to observe OXTR expression in dorsal root ganglia (DRG). The antinociceptive effects of OXT were assessed by hot-plate and von Frey tests. Whole-cell patch clamp was performed to record sodium currents, excitability of DRG neurons, and excitatory synapse transmission. KEY RESULTS Expression of OXTR in DRG neurons was enhanced significantly after PTX treatment. Activation of OXTR exhibited antinociceptive effects, by decreasing the hyperexcitability of DRG neurons in PTX-treated mice. Additionally, OXTR activation up-regulated the phosphorylation of protein kinase C (pPKC) and, in turn, impaired voltage-gated sodium currents, particularly the voltage-gated sodium channel 1.7 (NaV 1.7) current, that plays an indispensable role in PTX-induced neuropathic pain. OXT suppressed excitatory transmission in the spinal dorsal horn as well as excitatory inputs from primary afferents in PTX-treated mice. CONCLUSION AND IMPLICATIONS The OXTR in small-sized DRG neurons is up-regulated in CINP and its activation relieved CINP by inhibiting the neural excitability by impairment of NaV 1.7 currents via pPKC. Our results suggest that OXTR on peripheral sensory neurons is a potential therapeutic target to relieve CINP.
Collapse
Affiliation(s)
- Lixuan Li
- Guangdong Medical University, Zhanjiang, Guangdong, China.,Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, The 6th Affiliated Hospital of Shenzhen University Medical School, Shenzhen, Guangdong, China
| | - Pupu Li
- Department of Medical Oncology, The 6th Affiliated Hospital of Shenzhen University Medical School, Shenzhen, Guangdong, China
| | - Jing Guo
- Department of Endocrinology and Metabolism, Shenzhen University General Hospital and Shenzhen University Academy of Clinical Medical Sciences, Shenzhen University, Shenzhen, Guangdong, China
| | - Yifei Wu
- Department of Medical Neuroscience, Key University Laboratory of Metabolism and Health of Guangdong, SUSTech Center for Pain Medicine, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Qian Zeng
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, The 6th Affiliated Hospital of Shenzhen University Medical School, Shenzhen, Guangdong, China
| | - Nan Li
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, The 6th Affiliated Hospital of Shenzhen University Medical School, Shenzhen, Guangdong, China
| | - Xiaoting Huang
- Medical Research Center, The 6th Affiliated Hospital of Shenzhen University Medical School, Shenzhen, Guangdong, China
| | - Yongshen He
- Medical Research Center, The 6th Affiliated Hospital of Shenzhen University Medical School, Shenzhen, Guangdong, China
| | - Wen Ai
- Medical Research Center, The 6th Affiliated Hospital of Shenzhen University Medical School, Shenzhen, Guangdong, China
| | - Wuping Sun
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, The 6th Affiliated Hospital of Shenzhen University Medical School, Shenzhen, Guangdong, China
| | - Tao Liu
- Department of Pediatrics, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Donglin Xiong
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, The 6th Affiliated Hospital of Shenzhen University Medical School, Shenzhen, Guangdong, China
| | - Lizu Xiao
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, The 6th Affiliated Hospital of Shenzhen University Medical School, Shenzhen, Guangdong, China
| | - Yanyan Sun
- Department of Anesthesiology, Shenzhen University General Hospital and Shenzhen University Academy of Clinical Medical Sciences, Shenzhen University, Shenzhen, Guangdong, China
| | - Qiming Zhou
- Department of Medical Oncology, The 6th Affiliated Hospital of Shenzhen University Medical School, Shenzhen, Guangdong, China
| | - Haixia Kuang
- Department of Pediatrics, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Zilong Wang
- Department of Medical Neuroscience, Key University Laboratory of Metabolism and Health of Guangdong, SUSTech Center for Pain Medicine, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Changyu Jiang
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain Medicine, The 6th Affiliated Hospital of Shenzhen University Medical School, Shenzhen, Guangdong, China.,Medical Research Center, The 6th Affiliated Hospital of Shenzhen University Medical School, Shenzhen, Guangdong, China
| |
Collapse
|
23
|
Röderer P, Belu A, Heidrich L, Siobal M, Isensee J, Prolingheuer J, Janocha E, Valdor M, Hagendorf S, Bahrenberg G, Opitz T, Segschneider M, Haupt S, Nitzsche A, Brüstle O, Hucho T. Emergence of nociceptive functionality and opioid signaling in human induced pluripotent stem cell-derived sensory neurons. Pain 2023:00006396-990000000-00249. [PMID: 36727909 DOI: 10.1097/j.pain.0000000000002860] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 11/15/2022] [Indexed: 02/03/2023]
Abstract
ABSTRACT Induced pluripotent stem cells (iPSCs) have enabled the generation of various difficult-to-access cell types such as human nociceptors. A key challenge associated with human iPSC-derived nociceptors (hiPSCdNs) is their prolonged functional maturation. While numerous studies have addressed the expression of classic neuronal markers and ion channels in hiPSCdNs, the temporal development of key signaling cascades regulating nociceptor activity has remained largely unexplored. In this study, we used an immunocytochemical high-content imaging approach alongside electrophysiological staging to assess metabotropic and ionotropic signaling of large scale-generated hiPSCdNs across 70 days of in vitro differentiation. During this period, the resting membrane potential became more hyperpolarized, while rheobase, action potential peak amplitude, and membrane capacitance increased. After 70 days, hiPSCdNs exhibited robust physiological responses induced by GABA, pH shift, ATP, and capsaicin. Direct activation of protein kinase A type II (PKA-II) through adenylyl cyclase stimulation with forskolin resulted in PKA-II activation at all time points. Depolarization-induced activation of PKA-II emerged after 35 days of differentiation. However, effective inhibition of forskolin-induced PKA-II activation by opioid receptor agonists required 70 days of in vitro differentiation. Our results identify a pronounced time difference between early expression of functionally important ion channels and emergence of regulatory metabotropic sensitizing and desensitizing signaling only at advanced stages of in vitro cultivation, suggesting an independent regulation of ionotropic and metabotropic signaling. These data are relevant for devising future studies into the development and regulation of human nociceptor function and for defining time windows suitable for hiPSCdN-based drug discovery.
Collapse
Affiliation(s)
- Pascal Röderer
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty and University Hospital Bonn, Bonn
- LIFE & BRAIN GmbH, Cellomics Unit, Bonn, Germany, Germany
| | - Andreea Belu
- Translational Pain Research, Department of Anaesthesiology and Intensive Care Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Luzia Heidrich
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty and University Hospital Bonn, Bonn
- LIFE & BRAIN GmbH, Cellomics Unit, Bonn, Germany, Germany
| | - Maike Siobal
- Translational Pain Research, Department of Anaesthesiology and Intensive Care Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Jörg Isensee
- Translational Pain Research, Department of Anaesthesiology and Intensive Care Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Jonathan Prolingheuer
- Translational Pain Research, Department of Anaesthesiology and Intensive Care Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | | | | | | | | | - Thoralf Opitz
- Institute of Experimental Epileptology and Cognition Research, University of Bonn, Bonn, Germany
| | - Michaela Segschneider
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty and University Hospital Bonn, Bonn
| | - Simone Haupt
- LIFE & BRAIN GmbH, Cellomics Unit, Bonn, Germany, Germany
| | - Anja Nitzsche
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty and University Hospital Bonn, Bonn
- LIFE & BRAIN GmbH, Cellomics Unit, Bonn, Germany, Germany
| | - Oliver Brüstle
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty and University Hospital Bonn, Bonn
- LIFE & BRAIN GmbH, Cellomics Unit, Bonn, Germany, Germany
| | - Tim Hucho
- Translational Pain Research, Department of Anaesthesiology and Intensive Care Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
24
|
Alsaloum M, Labau JIR, Liu S, Effraim PR, Waxman SG. Stem cell-derived sensory neurons modelling inherited erythromelalgia: normalization of excitability. Brain 2023; 146:359-371. [PMID: 35088838 DOI: 10.1093/brain/awac031] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 12/30/2021] [Accepted: 01/06/2022] [Indexed: 01/11/2023] Open
Abstract
Effective treatment of pain remains an unmet healthcare need that requires new and effective therapeutic approaches. NaV1.7 has been genetically and functionally validated as a mediator of pain. Preclinical studies of NaV1.7-selective blockers have shown limited success and translation to clinical studies has been limited. The degree of NaV1.7 channel blockade necessary to attenuate neuronal excitability and ameliorate pain is an unanswered question important for drug discovery. Here, we utilize dynamic clamp electrophysiology and induced pluripotent stem cell-derived sensory neurons (iPSC-SNs) to answer this question for inherited erythromelalgia, a pain disorder caused by gain-of-function mutations in Nav1.7. We show that dynamic clamp can produce hyperexcitability in iPSC-SNs associated with two different inherited erythromelalgia mutations, NaV1.7-S241T and NaV1.7-I848T. We further show that blockade of approximately 50% of NaV1.7 currents can reverse neuronal hyperexcitability to baseline levels.
Collapse
Affiliation(s)
- Matthew Alsaloum
- Department of Neurology, Yale School of Medicine, New Haven, CT 06510, USA.,Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT 06516, USA.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA.,Yale Medical Scientist Training Program, Yale School of Medicine, New Haven, CT 06510, USA.,Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT 06510, USA
| | - Julie I R Labau
- Department of Neurology, Yale School of Medicine, New Haven, CT 06510, USA.,Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT 06516, USA.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA.,Department of Clinical Epidemiology and Medical Technology Assessment (KEMTA), Maastricht University Medical Centre, Maastricht, The Netherlands.,Department of Clinical Genetics, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Shujun Liu
- Department of Neurology, Yale School of Medicine, New Haven, CT 06510, USA.,Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT 06516, USA.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Philip R Effraim
- Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT 06516, USA.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA.,Department of Anesthesiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Stephen G Waxman
- Department of Neurology, Yale School of Medicine, New Haven, CT 06510, USA.,Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT 06516, USA.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| |
Collapse
|
25
|
Maximizing treatment efficacy through patient stratification in neuropathic pain trials. Nat Rev Neurol 2023; 19:53-64. [PMID: 36400867 DOI: 10.1038/s41582-022-00741-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2022] [Indexed: 11/19/2022]
Abstract
Treatment of neuropathic pain remains inadequate despite the elucidation of multiple pathophysiological mechanisms and the development of promising therapeutic compounds. The lack of success in translating knowledge into clinical practice has discouraged pharmaceutical companies from investing in pain medicine; however, new patient stratification approaches could help bridge the translation gap and develop individualized therapeutic approaches. As we highlight in this article, subgrouping of patients according to sensory profiles and other baseline characteristics could aid the prediction of treatment success. Furthermore, novel outcome measures have been developed for patients with neuropathic pain. The extent to which sensory profiles and outcome measures can be employed in routine clinical practice and clinical trials and across distinct neuropathic pain aetiologies is yet to be determined. Improvements in animal models, drawing on our knowledge of human pain, and robust public-private partnerships will be needed to pave the way to innovative and effective pain medicine in the future.
Collapse
|
26
|
Nimbalkar S, Guo X, Colón A, Jackson M, Akanda N, Patel A, Grillo M, Hickman JJ. Development of a functional human induced pluripotent stem cell-derived nociceptor MEA system as a pain model for analgesic drug testing. Front Cell Dev Biol 2023; 11:1011145. [PMID: 36936691 PMCID: PMC10014464 DOI: 10.3389/fcell.2023.1011145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 02/13/2023] [Indexed: 03/06/2023] Open
Abstract
The control of severe or chronic pain has relied heavily on opioids and opioid abuse and addiction have recently become a major global health crisis. Therefore, it is imperative to develop new pain therapeutics which have comparable efficacy for pain suppression but lack of the harmful effects of opioids. Due to the nature of pain, any in vivo experiment is undesired even in animals. Recent developments in stem cell technology has enabled the differentiation of nociceptors from human induced pluripotent stem cells. This study sought to establish an in vitro functional induced pluripotent stem cells-derived nociceptor culture system integrated with microelectrode arrays for nociceptive drug testing. Nociceptors were differentiated from induced pluripotent stem cells utilizing a modified protocol and a medium was designed to ensure prolonged and stable nociceptor culture. These neurons expressed nociceptor markers as characterized by immunocytochemistry and responded to the exogenous toxin capsaicin and the endogenous neural modulator ATP, as demonstrated with patch clamp electrophysiology. These cells were also integrated with microelectrode arrays for analgesic drug testing to demonstrate their utilization in the preclinical drug screening process. The neural activity was induced by ATP to mimic clinically relevant pathological pain and then the analgesics Lidocaine and the opioid DAMGO were tested individually and both induced immediate silencing of the nociceptive activity. This human-based functional nociceptive system provides a valuable platform for investigating pathological pain and for evaluating effective analgesics in the search of opioid substitutes.
Collapse
Affiliation(s)
- Siddharth Nimbalkar
- Hybrid Systems Lab, University of Central Florida, NanoScience Technology Center, Orlando, FL, United States
| | - Xiufang Guo
- Hybrid Systems Lab, University of Central Florida, NanoScience Technology Center, Orlando, FL, United States
| | - Alisha Colón
- Hybrid Systems Lab, University of Central Florida, NanoScience Technology Center, Orlando, FL, United States
| | | | - Nesar Akanda
- Hybrid Systems Lab, University of Central Florida, NanoScience Technology Center, Orlando, FL, United States
| | - Aakash Patel
- Hybrid Systems Lab, University of Central Florida, NanoScience Technology Center, Orlando, FL, United States
| | - Marcella Grillo
- Hybrid Systems Lab, University of Central Florida, NanoScience Technology Center, Orlando, FL, United States
| | - James J. Hickman
- Hybrid Systems Lab, University of Central Florida, NanoScience Technology Center, Orlando, FL, United States
- Hesperos Inc., Orlando, FL, United States
- *Correspondence: James J. Hickman,
| |
Collapse
|
27
|
Tsai NW, Lin CC, Yeh TY, Chiu YA, Chiu HH, Huang HP, Hsieh ST. An induced pluripotent stem cell-based model identifies molecular targets of vincristine neurotoxicity. Dis Model Mech 2022; 15:dmm049471. [PMID: 36518084 PMCID: PMC10655812 DOI: 10.1242/dmm.049471] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 09/29/2022] [Indexed: 11/19/2023] Open
Abstract
To model peripheral nerve degeneration and investigate molecular mechanisms of neurodegeneration, we established a cell system of induced pluripotent stem cell (iPSC)-derived sensory neurons exposed to vincristine, a drug that frequently causes chemotherapy-induced peripheral neuropathy. Sensory neurons differentiated from iPSCs exhibit distinct neurochemical patterns according to the immunocytochemical phenotypes, and gene expression of peripherin (PRPH, hereafter referred to as Peri) and neurofilament heavy chain (NEFH, hereafter referred to as NF). The majority of iPSC-derived sensory neurons were PRPH positive/NEFH negative, i.e. Peri(+)/NF(-) neurons, whose somata were smaller than those of Peri(+)/NF(+) neurons. On exposure to vincristine, projections from the cell body of a neuron, i.e. neurites, were degenerated quicker than somata, the lethal concentration to kill 50% (LC50) of neurites being below the LC50 for somata, consistent with the clinical pattern of length-dependent neuropathy. We then examined the molecular expression in the MAP kinase signaling pathways of, extracellular signal-regulated kinases 1/2 (MAPK1/3, hereafter referred to as ERK), p38 mitogen-activated protein kinases (MAPK11/12/13/14, hereafter referred to as p38) and c-Jun N-terminal kinases (MAPK8/9/10, hereafter referred to as JNK). Regarding these three cascades, only phosphorylation of JNK was upregulated but not that of p38 or ERK1/2. Furthermore, vincristine-treatment resulted in impaired autophagy and reduced autophagic flux. Rapamycin-treatment reversed the effect of impaired autophagy and JNK activation. These results not only established a platform to study peripheral degeneration of human neurons but also provide molecular mechanisms for neurodegeneration with the potential for therapeutic targets.
Collapse
Affiliation(s)
- Neng-Wei Tsai
- Department of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei 100, Taiwan
| | - Cheng-Chen Lin
- Department of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei 100, Taiwan
| | - Ti-Yen Yeh
- Department of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei 100, Taiwan
| | - Yu-An Chiu
- Department of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei 100, Taiwan
| | - Hsin-Hui Chiu
- Department of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei 100, Taiwan
| | - Hsiang-Po Huang
- Department of Medical Genomics and Proteomics, National Taiwan University College of Medicine, Taipei 100, Taiwan
- Department of Pediatrics, National Taiwan University Children's Hospital, Taipei 100, Taiwan
| | - Sung-Tsang Hsieh
- Department of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei 100, Taiwan
- Department of Brain and Mind Sciences, National Taiwan University College of Medicine, Taipei 100, Taiwan
- Department of Neurology, National Taiwan University Hospital, Taipei 100, Taiwan
| |
Collapse
|
28
|
Mießner H, Seidel J, Smith ESJ. In vitro models for investigating itch. Front Mol Neurosci 2022; 15:984126. [PMID: 36385768 PMCID: PMC9644192 DOI: 10.3389/fnmol.2022.984126] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 10/10/2022] [Indexed: 12/04/2022] Open
Abstract
Itch (pruritus) is a sensation that drives a desire to scratch, a behavior observed in many animals. Although generally short-lasting and not causing harm, there are several pathological conditions where chronic itch is a hallmark symptom and in which prolonged scratching can induce damage. Finding medications to counteract the sensation of chronic itch has proven difficult due to the molecular complexity that involves a multitude of triggers, receptors and signaling pathways between skin, immune and nerve cells. While much has been learned about pruritus from in vivo animal models, they have limitations that corroborate the necessity for a transition to more human disease-like models. Also, reducing animal use should be encouraged in research. However, conducting human in vivo experiments can also be ethically challenging. Thus, there is a clear need for surrogate models to be used in pre-clinical investigation of the mechanisms of itch. Most in vitro models used for itch research focus on the use of known pruritogens. For this, sensory neurons and different types of skin and/or immune cells are stimulated in 2D or 3D co-culture, and factors such as neurotransmitter or cytokine release can be measured. There are however limitations of such simplistic in vitro models. For example, not all naturally occurring cell types are present and there is also no connection to the itch-sensing organ, the central nervous system (CNS). Nevertheless, in vitro models offer a chance to investigate otherwise inaccessible specific cell–cell interactions and molecular pathways. In recent years, stem cell-based approaches and human primary cells have emerged as viable alternatives to standard cell lines or animal tissue. As in vitro models have increased in their complexity, further opportunities for more elaborated means of investigating itch have been developed. In this review, we introduce the latest concepts of itch and discuss the advantages and limitations of current in vitro models, which provide valuable contributions to pruritus research and might help to meet the unmet clinical need for more refined anti-pruritic substances.
Collapse
Affiliation(s)
- Hendrik Mießner
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
- Dermatological Skin Care, Beiersdorf AG, Hamburg, Germany
| | - Judith Seidel
- Dermatological Skin Care, Beiersdorf AG, Hamburg, Germany
| | - Ewan St. John Smith
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
- *Correspondence: Ewan St. John Smith,
| |
Collapse
|
29
|
Middleton SJ, Perini I, Themistocleous AC, Weir GA, McCann K, Barry AM, Marshall A, Lee M, Mayo LM, Bohic M, Baskozos G, Morrison I, Löken LS, McIntyre S, Nagi SS, Staud R, Sehlstedt I, Johnson RD, Wessberg J, Wood JN, Woods CG, Moqrich A, Olausson H, Bennett DL. Nav1.7 is required for normal C-low threshold mechanoreceptor function in humans and mice. Brain 2022; 145:3637-3653. [PMID: 34957475 PMCID: PMC9586547 DOI: 10.1093/brain/awab482] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 11/03/2021] [Accepted: 11/24/2021] [Indexed: 11/15/2022] Open
Abstract
Patients with bi-allelic loss of function mutations in the voltage-gated sodium channel Nav1.7 present with congenital insensitivity to pain (CIP), whilst low threshold mechanosensation is reportedly normal. Using psychophysics (n = 6 CIP participants and n = 86 healthy controls) and facial electromyography (n = 3 CIP participants and n = 8 healthy controls), we found that these patients also have abnormalities in the encoding of affective touch, which is mediated by the specialized afferents C-low threshold mechanoreceptors (C-LTMRs). In the mouse, we found that C-LTMRs express high levels of Nav1.7. Genetic loss or selective pharmacological inhibition of Nav1.7 in C-LTMRs resulted in a significant reduction in the total sodium current density, an increased mechanical threshold and reduced sensitivity to non-noxious cooling. The behavioural consequence of loss of Nav1.7 in C-LTMRs in mice was an elevation in the von Frey mechanical threshold and less sensitivity to cooling on a thermal gradient. Nav1.7 is therefore not only essential for normal pain perception but also for normal C-LTMR function, cool sensitivity and affective touch.
Collapse
Affiliation(s)
- Steven J Middleton
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Irene Perini
- Center for Social and Affective Neuroscience, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- Center for Medical Image Science and Visualization, Linköping, Sweden
| | - Andreas C Themistocleous
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
- Brain Function Research Group, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Greg A Weir
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Kirsty McCann
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Allison M Barry
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Andrew Marshall
- Institute of Aging and Chronic Disease, University of Liverpool, L3 5DA Liverpool, UK
| | - Michael Lee
- University Division of Anaesthesia, University of Cambridge, Cambridge NHS Foundation Trust Hospitals, Hills Road, Cambridge CB2 0QQ, UK
| | - Leah M Mayo
- Center for Social and Affective Neuroscience, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Manon Bohic
- Aix-Marseille-Université, CNRS, Institute de Biologie du Développement de Marseille, UMR 7288, case 907, 13288 Marseille Cedex 09, France
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Georgios Baskozos
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - India Morrison
- Center for Social and Affective Neuroscience, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Line S Löken
- Department of Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Sarah McIntyre
- Center for Social and Affective Neuroscience, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Saad S Nagi
- Center for Social and Affective Neuroscience, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Roland Staud
- Department of Physiological Sciences, University of Florida College of Veterinary Medicine, Gainesville, FL, USA
| | - Isac Sehlstedt
- Department of Psychology, University of Gothenburg, Gothenburg, Sweden
| | - Richard D Johnson
- Department of Physiology, University of Gothenburg, Gothenburg, Sweden
- Department of Physiological Sciences, University of Florida College of Veterinary Medicine, Gainesville, FL, USA
| | - Johan Wessberg
- Department of Physiology, University of Gothenburg, Gothenburg, Sweden
| | - John N Wood
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, UK
| | - Christopher G Woods
- Cambridge Institute for Medical Research, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Aziz Moqrich
- Aix-Marseille-Université, CNRS, Institute de Biologie du Développement de Marseille, UMR 7288, case 907, 13288 Marseille Cedex 09, France
| | - Håkan Olausson
- Center for Social and Affective Neuroscience, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - David L Bennett
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| |
Collapse
|
30
|
Labau JIR, Andelic M, Faber CG, Waxman SG, Lauria G, Dib-Hajj SD. Recent advances for using human induced-pluripotent stem cells as pain-in-a-dish models of neuropathic pain. Exp Neurol 2022; 358:114223. [PMID: 36100046 DOI: 10.1016/j.expneurol.2022.114223] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 08/15/2022] [Accepted: 09/05/2022] [Indexed: 11/28/2022]
Abstract
Neuropathic pain is amongst the most common non-communicable disorders and the poor effectiveness of current treatment is an unmet need. Although pain is a universal experience, there are significant inter-individual phenotypic differences. Developing models that can accurately recapitulate the clinical pain features is crucial to better understand underlying pathophysiological mechanisms and find innovative treatments. Current data from heterologous expression systems that investigate properties of specific molecules involved in pain signaling, and from animal models, show limited success with their translation into the development of novel treatments for pain. This is in part because they do not recapitulate the native environment in which a particular molecule functions, and due to species-specific differences in the properties of several key molecules that are involved in pain signaling. The limited availability of post-mortem tissue, in particular dorsal root ganglia (DRG), has hampered research using human cells in pre-clinical studies. Human induced-pluripotent stem cells (iPSCs) have emerged as an exciting alternative platform to study patient-specific diseases. Sensory neurons that are derived from iPSCs (iPSC-SNs) have provided new avenues towards elucidating peripheral pathophysiological mechanisms, the potential for development of personalized treatments, and as a cell-based system for high-throughput screening for discovering novel analgesics. Nevertheless, reprogramming and differentiation protocols to obtain nociceptors have mostly yielded immature homogenous cell populations that do not recapitulate the heterogeneity of native sensory neurons. To close the gap between native human tissue and iPSCs, alternative strategies have been developed. We will review here recent developments in differentiating iPSC-SNs and their use in pre-clinical translational studies. Direct conversion of stem cells into the cells of interest has provided a more cost- and time-saving method to improve reproducibility and diversity of sensory cell types. Furthermore, multi-cellular strategies that mimic in vivo microenvironments for cell maturation, by improving cell contact and communication (co-cultures), reproducing the organ complexity and architecture (three-dimensional organoid), and providing iPSCs with the full spatiotemporal context and nutrients needed for acquiring a mature phenotype (xenotransplantation), have led to functional sensory neuron-like systems. Finally, this review touches on novel prospective strategies, including fluorescent-tracking to select the differentiated neurons of relevance, and dynamic clamp, an electrophysiological method that allows direct manipulation of ionic conductances that are missing in iPSC-SNs.
Collapse
Affiliation(s)
- Julie I R Labau
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA; Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA; Department of Toxicogenomics, Clinical Genomics, Maastricht University Medical Centre+, Maastricht, the Netherlands; School of Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Mirna Andelic
- School of Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands; Department of Neurology, Neuroalgology Unit, Fondazione IRCCS Istituto Neurologico "Carlo Besta", Milan, Italy
| | - Catharina G Faber
- School of Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA; Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA
| | - Giuseppe Lauria
- Department of Clinical Neurosciences, Fondazione IRCCS Istituto Neurologico "Carlo Besta", Milan, Italy; Department of Medical Biotechnology and Translational Medicine, University of Milan, Italy.
| | - Sulayman D Dib-Hajj
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA; Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA.
| |
Collapse
|
31
|
Zeidler M, Kummer KK, Kress M. Towards bridging the translational gap by improved modeling of human nociception in health and disease. Pflugers Arch 2022; 474:965-978. [PMID: 35655042 PMCID: PMC9393146 DOI: 10.1007/s00424-022-02707-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 05/18/2022] [Indexed: 11/09/2022]
Abstract
Despite numerous studies which have explored the pathogenesis of pain disorders in preclinical models, there is a pronounced translational gap, which is at least partially caused by differences between the human and rodent nociceptive system. An elegant way to bridge this divide is the exploitation of human-induced pluripotent stem cell (iPSC) reprogramming into human iPSC-derived nociceptors (iDNs). Several protocols were developed and optimized to model nociceptive processes in health and disease. Here we provide an overview of the different approaches and summarize the knowledge obtained from such models on pain pathologies associated with monogenetic sensory disorders so far. In addition, novel perspectives offered by increasing the complexity of the model systems further to better reflect the natural environment of nociceptive neurons by involving other cell types in 3D model systems are described.
Collapse
Affiliation(s)
- Maximilian Zeidler
- Institute of Physiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Kai K Kummer
- Institute of Physiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Michaela Kress
- Institute of Physiology, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
32
|
Loya-López SI, Duran P, Ran D, Calderon-Rivera A, Gomez K, Moutal A, Khanna R. Cell specific regulation of NaV1.7 activity and trafficking in rat nodose ganglia neurons. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2022; 12:100109. [PMID: 36531612 PMCID: PMC9755031 DOI: 10.1016/j.ynpai.2022.100109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/10/2022] [Accepted: 11/10/2022] [Indexed: 06/17/2023]
Abstract
The voltage-gated sodium NaV1.7 channel sets the threshold for electrogenesis. Mutations in the gene encoding human NaV1.7 (SCN9A) cause painful neuropathies or pain insensitivity. In dorsal root ganglion (DRG) neurons, activity and trafficking of NaV1.7 are regulated by the auxiliary collapsin response mediator protein 2 (CRMP2). Specifically, preventing addition of a small ubiquitin-like modifier (SUMO), by the E2 SUMO-conjugating enzyme Ubc9, at lysine-374 (K374) of CRMP2 reduces NaV1.7 channel trafficking and activity. We previously identified a small molecule, designated 194, that prevented CRMP2 SUMOylation by Ubc9 to reduce NaV1.7 surface expression and currents, leading to a reduction in spinal nociceptive transmission, and culminating in normalization of mechanical allodynia in models of neuropathic pain. In this study, we investigated whether NaV1.7 control via CRMP2-SUMOylation is conserved in nodose ganglion (NG) neurons. This study was motivated by our desire to develop 194 as a safe, non-opioid substitute for persistent pain, which led us to wonder how 194 would impact NaV1.7 in NG neurons, which are responsible for driving the cough reflex. We found functioning NaV1.7 channels in NG neurons; however, they were resistant to downregulation via either CRMP2 knockdown or pharmacological inhibition of CRMP2 SUMOylation by 194. CRMP2 SUMOylation and interaction with NaV1.7 was consered in NG neurons but the endocytic machinery was deficient in the endocytic adaptor protein Numb. Overexpression of Numb rescued CRMP2-dependent regulation on NaV1.7, rendering NG neurons sensitive to 194. Altogether, these data point at the existence of cell-specific mechanisms regulating NaV1.7 trafficking.
Collapse
Affiliation(s)
- Santiago I. Loya-López
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, USA
- NYU Pain Research Center, 433 First Avenue, New York, NY 10010, USA
| | - Paz Duran
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, USA
- NYU Pain Research Center, 433 First Avenue, New York, NY 10010, USA
| | - Dongzhi Ran
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Aida Calderon-Rivera
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, USA
- NYU Pain Research Center, 433 First Avenue, New York, NY 10010, USA
| | - Kimberly Gomez
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, USA
- NYU Pain Research Center, 433 First Avenue, New York, NY 10010, USA
| | - Aubin Moutal
- School of Medicine, Department of Pharmacology and Physiology, Saint Louis University, Saint Louis, MO 63104, USA
| | - Rajesh Khanna
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, USA
- NYU Pain Research Center, 433 First Avenue, New York, NY 10010, USA
| |
Collapse
|
33
|
Chase R, de la Peña JB, Smith PR, Lawson J, Lou TF, Stanowick AD, Black BJ, Campbell ZT. Global analyses of mRNA expression in human sensory neurons reveal eIF5A as a conserved target for inflammatory pain. FASEB J 2022; 36:e22422. [PMID: 35747924 DOI: 10.1096/fj.202101933rr] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 05/28/2022] [Accepted: 06/08/2022] [Indexed: 11/11/2022]
Abstract
Nociceptors are a type of sensory neuron that are integral to most forms of pain. Targeted disruption of nociceptor sensitization affords unique opportunities to prevent pain. An emerging model for nociceptors are sensory neurons derived from human stem cells. Here, we subjected five groups to high-throughput sequencing: human induced pluripotent stem cells (hiPSCs) prior to differentiation, mature hiPSC-derived sensory neurons, mature co-cultures containing hiPSC-derived astrocytes and sensory neurons, mouse dorsal root ganglion (DRG) tissues, and mouse DRG cultures. Co-culture of nociceptors and astrocytes promotes expression of transcripts enriched in DRG tissues. Comparisons of the hiPSC models to tissue samples reveal that many key transcripts linked to pain are present. Markers indicative of a range of neuronal subtypes present in the DRG were detected in mature hiPSCs. Intriguingly, translation factors were maintained at consistently high expression levels across species and culture systems. As a proof of concept for the utility of this resource, we validated expression of eukaryotic initiation factor 5A (eIF5A) in DRG tissues and hiPSC samples. eIF5A is subject to a unique posttranslational hypusine modification required for its activity. Inhibition of hypusine biosynthesis prevented hyperalgesic priming by inflammatory mediators in vivo and diminished hiPSC activity in vitro. Collectively, our results illuminate the transcriptomes of hiPSC sensory neuron models. We provide a demonstration for this resource through our investigation of eIF5A. Our findings reveal hypusine as a potential target for inflammation associated pain in males.
Collapse
Affiliation(s)
- Rebecca Chase
- Department of Biological Sciences, University of Texas at Dallas, Richardson, Texas, USA
| | - June Bryan de la Peña
- Department of Biological Sciences, University of Texas at Dallas, Richardson, Texas, USA
| | - Patrick R Smith
- Department of Biological Sciences, University of Texas at Dallas, Richardson, Texas, USA
| | - Jennifer Lawson
- Department of Biomedical Engineering, University of Massachusetts Lowell, Lowell, Massachusetts, USA
| | - Tzu-Fang Lou
- Department of Biological Sciences, University of Texas at Dallas, Richardson, Texas, USA
| | - Alexander D Stanowick
- Department of Biological Sciences, University of Texas at Dallas, Richardson, Texas, USA
| | - Bryan J Black
- Department of Biomedical Engineering, University of Massachusetts Lowell, Lowell, Massachusetts, USA
| | - Zachary T Campbell
- Department of Biological Sciences, University of Texas at Dallas, Richardson, Texas, USA.,Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, Texas, USA
| |
Collapse
|
34
|
Zhang XY, Wu X, Zhang P, Gan YH. Prolonged PGE 2 treatment increased TTX-sensitive but not TTX-resistant sodium current in trigeminal ganglionic neurons. Neuropharmacology 2022; 215:109156. [PMID: 35691365 DOI: 10.1016/j.neuropharm.2022.109156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 05/09/2022] [Accepted: 05/31/2022] [Indexed: 10/18/2022]
Abstract
Prostaglandin E2 (PGE2) is an important inflammatory mediator for the initiation and maintenance of inflammatory and neuropathic pain. The acute effect of PGE2 on sodium currents has been widely characterized in sensory neurons; however, the prolonged effect of PGE2 remains to be determined. Here, we performed patch clamp recordings to evaluate the acute and prolonged effects of PGE2 on sodium currents in trigeminal ganglionic (TG) neurons from male Sprague-Dawley rats. We found that 24-h treatment with PGE2 (10 μM) increased the peak sodium current density by approximately 31% in a voltage-dependent manner and shifted the activation curve in a hyperpolarized direction but did not affect steady-state inactivation. Furthermore, treatment with PGE2 for 24 h increased the current density of tetrodotoxin-sensitive (TTX-S) but not TTX-resistant (TTX-R) channels significantly. Interestingly, TTX-S current was increased mostly in medium-sized, but not in small-sized, neurons after 24 h of treatment with PGE2. Moreover, the mRNA level of TTX-S Nav1.1 but not TTX-R Nav1.8 or Nav1.9 was significantly increased after 24 h of treatment with PGE2. In contrast, 5-min treatment with PGE2 (10 μM) increased the peak sodium current density by approximately 29% and increased TTX-R sodium currents, but not TTX-S currents, in both small- and medium-sized TG neurons. Our results presented a differential regulation of subtypes of sodium channels by acute and prolonged treatments of PGE2, which may help to better understand the mechanism of PGE2-mediated orofacial pain development.
Collapse
Affiliation(s)
- Xiao-Yu Zhang
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
| | - Xi Wu
- Academy for Advanced Interdisciplinary Studies, College of Future Technology, Institute of Molecular Medicine, Peking University, Beijing, 100871, PR China
| | - Peng Zhang
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
| | - Ye-Hua Gan
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China.
| |
Collapse
|
35
|
Alsaloum M, Waxman SG. iPSCs and DRGs: stepping stones to new pain therapies. Trends Mol Med 2022; 28:110-122. [PMID: 34933815 PMCID: PMC8810720 DOI: 10.1016/j.molmed.2021.11.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/23/2021] [Accepted: 11/24/2021] [Indexed: 02/03/2023]
Abstract
There is a pressing need for more effective nonaddictive treatment options for pain. Pain signals are transmitted from the periphery into the spinal cord via dorsal root ganglion (DRG) neurons, whose excitability is driven by voltage-gated sodium (NaV) channels. Three NaV channels (NaV1.7, NaV1.8, and NaV1.9), preferentially expressed in DRG neurons, play important roles in pain signaling in humans. Blockade of these channels may provide a novel approach to the treatment of pain, but clinical translation of preclinical results has been challenging, in part due to differences between rodent and human DRG neurons. Human DRG neurons and iPSC-derived sensory neurons (iPSC-SNs) provide new preclinical platforms that may facilitate the development of novel pain therapeutics.
Collapse
Affiliation(s)
- Matthew Alsaloum
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA; Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA; Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, USA; Yale Medical Scientist Training Program, Yale School of Medicine, New Haven, CT, USA; Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT, USA
| | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA; Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA; Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, USA.
| |
Collapse
|
36
|
|
37
|
Abstract
Induced pluripotent stem cells (iPS-cells) have significantly expanded our experimental possibilities, by creating new strategies for the molecular study of human disease and drug development. Treatment of pain has not seen much improvement over the past decade, likely due to species differences in preclinical models. Thus, iPS-cell derived sensory neurons offer a highly welcome translational approach for research and drug development. Although central neuronal differentiation is relatively straightforward, the successful and reliable generation of peripheral neurons requires more complex measures. Here, we describe a small molecule-based protocol for the differentiation of human sensory neurons from iPS-cells which renders functional nociceptor-like cells within several weeks.
Collapse
Affiliation(s)
- Anika Neureiter
- Institute of Physiology, Uniklinik RWTH Aachen, Aachen, Germany
| | - Esther Eberhardt
- Department of Anesthesiology, Uniklinik RWTH Aachen, Aachen, Germany
| | - Angelika Lampert
- Institute of Physiology, Uniklinik RWTH Aachen, Aachen, Germany.
| |
Collapse
|
38
|
Alles SRA, Smith PA. Peripheral Voltage-Gated Cation Channels in Neuropathic Pain and Their Potential as Therapeutic Targets. FRONTIERS IN PAIN RESEARCH 2021; 2:750583. [PMID: 35295464 PMCID: PMC8915663 DOI: 10.3389/fpain.2021.750583] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 11/10/2021] [Indexed: 11/25/2022] Open
Abstract
The persistence of increased excitability and spontaneous activity in injured peripheral neurons is imperative for the development and persistence of many forms of neuropathic pain. This aberrant activity involves increased activity and/or expression of voltage-gated Na+ and Ca2+ channels and hyperpolarization activated cyclic nucleotide gated (HCN) channels as well as decreased function of K+ channels. Because they display limited central side effects, peripherally restricted Na+ and Ca2+ channel blockers and K+ channel activators offer potential therapeutic approaches to pain management. This review outlines the current status and future therapeutic promise of peripherally acting channel modulators. Selective blockers of Nav1.3, Nav1.7, Nav1.8, Cav3.2, and HCN2 and activators of Kv7.2 abrogate signs of neuropathic pain in animal models. Unfortunately, their performance in the clinic has been disappointing; some substances fail to meet therapeutic end points whereas others produce dose-limiting side effects. Despite this, peripheral voltage-gated cation channels retain their promise as therapeutic targets. The way forward may include (i) further structural refinement of K+ channel activators such as retigabine and ASP0819 to improve selectivity and limit toxicity; use or modification of Na+ channel blockers such as vixotrigine, PF-05089771, A803467, PF-01247324, VX-150 or arachnid toxins such as Tap1a; the use of Ca2+ channel blockers such as TTA-P2, TTA-A2, Z 944, ACT709478, and CNCB-2; (ii) improving methods for assessing "pain" as opposed to nociception in rodent models; (iii) recognizing sex differences in pain etiology; (iv) tailoring of therapeutic approaches to meet the symptoms and etiology of pain in individual patients via quantitative sensory testing and other personalized medicine approaches; (v) targeting genetic and biochemical mechanisms controlling channel expression using anti-NGF antibodies such as tanezumab or re-purposed drugs such as vorinostat, a histone methyltransferase inhibitor used in the management of T-cell lymphoma, or cercosporamide a MNK 1/2 inhibitor used in treatment of rheumatoid arthritis; (vi) combination therapy using drugs that are selective for different channel types or regulatory processes; (vii) directing preclinical validation work toward the use of human or human-derived tissue samples; and (viii) application of molecular biological approaches such as clustered regularly interspaced short palindromic repeats (CRISPR) technology.
Collapse
Affiliation(s)
- Sascha R A Alles
- Department of Anesthesiology and Critical Care Medicine, University of New Mexico School of Medicine, Albuquerque, NM, United States
| | - Peter A Smith
- Department of Pharmacology, Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
39
|
Le Cann K, Meents JE, Sudha Bhagavath Eswaran V, Dohrn MF, Bott R, Maier A, Bialer M, Hautvast P, Erickson A, Rolke R, Rothermel M, Körner J, Kurth I, Lampert A. Assessing the impact of pain-linked Nav1.7 variants: An example of two variants with no biophysical effect. Channels (Austin) 2021; 15:208-228. [PMID: 33487118 PMCID: PMC7833769 DOI: 10.1080/19336950.2020.1870087] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/23/2020] [Accepted: 12/23/2020] [Indexed: 11/30/2022] Open
Abstract
Mutations in the voltage-gated sodium channel Nav1.7 are linked to human pain. The Nav1.7/N1245S variant was described before in several patients suffering from primary erythromelalgia and/or olfactory hypersensitivity. We have identified this variant in a pain patient and a patient suffering from severe and life-threatening orthostatic hypotension. In addition, we report a female patient suffering from muscle pain and carrying the Nav1.7/E1139K variant. We tested both Nav1.7 variants by whole-cell voltage-clamp recordings in HEK293 cells, revealing a slightly enhanced current density for the N1245S variant when co-expressed with the β1 subunit. This effect was counteracted by an enhanced slow inactivation. Both variants showed similar voltage dependence of activation and steady-state fast inactivation, as well as kinetics of fast inactivation, deactivation, and use-dependency compared to WT Nav1.7. Finally, homology modeling revealed that the N1245S substitution results in different intramolecular interaction partners. Taken together, these experiments do not point to a clear pathogenic effect of either the N1245S or E1139K variant and suggest they may not be solely responsible for the patients' pain symptoms. As discussed previously for other variants, investigations in heterologous expression systems may not sufficiently mimic the pathophysiological situation in pain patients, and single nucleotide variants in other genes or modulatory proteins are necessary for these specific variants to show their effect. Our findings stress that biophysical investigations of ion channel mutations need to be evaluated with care and should preferably be supplemented with studies investigating the mutations in their context, ideally in human sensory neurons.
Collapse
Affiliation(s)
- Kim Le Cann
- Institute of Physiology, RWTH Aachen University Hospital, Aachen, Germany
| | - Jannis E. Meents
- Institute of Physiology, RWTH Aachen University Hospital, Aachen, Germany
| | | | - Maike F. Dohrn
- Department of Neurology, Medical Faculty, RWTH Aachen University Hospital, Aachen, Germany
| | - Raya Bott
- Institute of Physiology, RWTH Aachen University Hospital, Aachen, Germany
| | - Andrea Maier
- Department of Neurology, Medical Faculty, RWTH Aachen University Hospital, Aachen, Germany
| | - Martin Bialer
- Division of Clinical Metabolism of Medical Genetics and Human Genomics at Northwell Health System, New-York, United States
| | - Petra Hautvast
- Institute of Physiology, RWTH Aachen University Hospital, Aachen, Germany
| | - Andelain Erickson
- Institute of Physiology, RWTH Aachen University Hospital, Aachen, Germany
| | - Roman Rolke
- Department for Palliative Care, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Markus Rothermel
- Department of Chemosensation, AG Neuromodulation, Institute for Biology II, RWTH Aachen University, Aachen, 52074, Germany
| | - Jannis Körner
- Institute of Physiology, RWTH Aachen University Hospital, Aachen, Germany
- Department of Anaesthesiology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Ingo Kurth
- Institute of Human Genetics, Medical Faculty, RWTH Aachen University Hospital, Aachen, Germany
| | - Angelika Lampert
- Institute of Physiology, RWTH Aachen University Hospital, Aachen, Germany
| |
Collapse
|
40
|
Cai S, Moutal A, Yu J, Chew LA, Isensee J, Chawla R, Gomez K, Luo S, Zhou Y, Chefdeville A, Madura C, Perez-Miller S, Bellampalli SS, Dorame A, Scott DD, François-Moutal L, Shan Z, Woodward T, Gokhale V, Hohmann AG, Vanderah TW, Patek M, Khanna M, Hucho T, Khanna R. Selective targeting of NaV1.7 via inhibition of the CRMP2-Ubc9 interaction reduces pain in rodents. Sci Transl Med 2021; 13:eabh1314. [PMID: 34757807 PMCID: PMC11729770 DOI: 10.1126/scitranslmed.abh1314] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The voltage-gated sodium NaV1.7 channel, critical for sensing pain, has been actively targeted by drug developers; however, there are currently no effective and safe therapies targeting NaV1.7. Here, we tested whether a different approach, indirect NaV1.7 regulation, could have antinociceptive effects in preclinical models. We found that preventing addition of small ubiquitin-like modifier (SUMO) on the NaV1.7-interacting cytosolic collapsin response mediator protein 2 (CRMP2) blocked NaV1.7 functions and had antinociceptive effects in rodents. In silico targeting of the SUMOylation site in CRMP2 (Lys374) identified >200 hits, of which compound 194 exhibited selective in vitro and ex vivo NaV1.7 engagement. Orally administered 194 was not only antinociceptive in preclinical models of acute and chronic pain but also demonstrated synergy alongside other analgesics—without eliciting addiction, rewarding properties, or neurotoxicity. Analgesia conferred by 194 was opioid receptor dependent. Our results demonstrate that 194 is a first-in-class protein-protein inhibitor that capitalizes on CRMP2-NaV1.7 regulation to deliver safe analgesia in rodents.
Collapse
Affiliation(s)
- Song Cai
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Aubin Moutal
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Jie Yu
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Lindsey A. Chew
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Jörg Isensee
- Department of Anesthesiology and Intensive Care Medicine, Translational Pain Research, University Hospital of Cologne, University Cologne, Joseph-Stelzmann-Str 9, Cologne D-50931, Germany
| | - Reena Chawla
- BIO5 Institute, 1657 East Helen Street, Tucson, AZ 85721, USA
| | - Kimberly Gomez
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Shizhen Luo
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Yuan Zhou
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Aude Chefdeville
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Cynthia Madura
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Samantha Perez-Miller
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
- Center for Innovation in Brain Sciences, University of Arizona, Tucson, AZ 85721, USA
| | - Shreya Sai Bellampalli
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Angie Dorame
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - David D. Scott
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Liberty François-Moutal
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Zhiming Shan
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
| | - Taylor Woodward
- Department of Psychological and Brain Sciences, Program in Neuroscience and Gill Center for Biomolecular Science, Indiana University, Bloomington, IN 47405-2204, USA
| | - Vijay Gokhale
- BIO5 Institute, 1657 East Helen Street, Tucson, AZ 85721, USA
- College of Pharmacy, University of Arizona, 1703 East Mabel Street, Tucson, AZ 85721, USA
| | - Andrea G. Hohmann
- Department of Psychological and Brain Sciences, Program in Neuroscience and Gill Center for Biomolecular Science, Indiana University, Bloomington, IN 47405-2204, USA
| | - Todd W. Vanderah
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
- Comprehensive Pain and Addiction Center, The University of Arizona, Tucson, AZ 85724, USA
| | - Marcel Patek
- Regulonix LLC, 1555 E. Entrada Segunda, Tucson, AZ 85718, USA
- Bright Rock Path LLC, Tucson, AZ 85724, USA
| | - May Khanna
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
- BIO5 Institute, 1657 East Helen Street, Tucson, AZ 85721, USA
- Center for Innovation in Brain Sciences, University of Arizona, Tucson, AZ 85721, USA
- Regulonix LLC, 1555 E. Entrada Segunda, Tucson, AZ 85718, USA
| | - Tim Hucho
- Department of Anesthesiology and Intensive Care Medicine, Translational Pain Research, University Hospital of Cologne, University Cologne, Joseph-Stelzmann-Str 9, Cologne D-50931, Germany
| | - Rajesh Khanna
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ 85724, USA
- BIO5 Institute, 1657 East Helen Street, Tucson, AZ 85721, USA
- Center for Innovation in Brain Sciences, University of Arizona, Tucson, AZ 85721, USA
- Comprehensive Pain and Addiction Center, The University of Arizona, Tucson, AZ 85724, USA
- Regulonix LLC, 1555 E. Entrada Segunda, Tucson, AZ 85718, USA
| |
Collapse
|
41
|
Jayakar S, Shim J, Jo S, Bean BP, Singeç I, Woolf CJ. Developing nociceptor-selective treatments for acute and chronic pain. Sci Transl Med 2021; 13:eabj9837. [PMID: 34757806 PMCID: PMC9964063 DOI: 10.1126/scitranslmed.abj9837] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Despite substantial efforts dedicated to the development of new, nonaddictive analgesics, success in treating pain has been limited. Clinically available analgesic agents generally lack efficacy and may have undesirable side effects. Traditional target-based drug discovery efforts that generate compounds with selectivity for single targets have a high rate of attrition because of their poor clinical efficacy. Here, we examine the challenges associated with the current analgesic drug discovery model and review evidence in favor of stem cell–derived neuronal-based screening approaches for the identification of analgesic targets and compounds for treating diverse forms of acute and chronic pain.
Collapse
Affiliation(s)
- Selwyn Jayakar
- F.M. Kirby Neurobiology, Boston Children’s Hospital, and Department of Neurology, Harvard Medical School; Boston, MA 02115, USA
| | - Jaehoon Shim
- F.M. Kirby Neurobiology, Boston Children’s Hospital, and Department of Neurology, Harvard Medical School; Boston, MA 02115, USA
| | - Sooyeon Jo
- Department of Neurobiology, Harvard Medical School; Boston, MA 02115, USA
| | - Bruce P Bean
- Department of Neurobiology, Harvard Medical School; Boston, MA 02115, USA
| | - Ilyas Singeç
- National Center for Advancing Translational Sciences (NCATS), Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH); Bethesda, MD 20892, USA
| | - Clifford J Woolf
- F.M. Kirby Neurobiology, Boston Children’s Hospital, and Department of Neurology, Harvard Medical School; Boston, MA 02115, USA
| |
Collapse
|
42
|
Malheiro A, Harichandan A, Bernardi J, Seijas-Gamardo A, Konings GF, Volders PGA, Romano A, Mota C, Wieringa P, Moroni L. 3D culture platform of human iPSCs-derived nociceptors for peripheral nerve modelling and tissue innervation. Biofabrication 2021; 14. [PMID: 34736244 DOI: 10.1088/1758-5090/ac36bf] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 11/04/2021] [Indexed: 11/11/2022]
Abstract
Functional humanized in vitro nerve models are coveted as an alternative to animal models due to their ease of access, lower cost, clinical relevance and no need for recurrent animal sacrifice. To this end, we developed a sensory nerve model using induced pluripotent stem cells (iPSCs)-derived nociceptors that are electrically active and exhibit a functional response to noxious stimuli. The differentiated neurons were co-cultured with primary Schwann cells on an aligned microfibrous scaffold to produce biomimetic peripheral nerve tissue. Compared to glass coverslips, our scaffold enhances tissue development and stabilization. Using this model, we demonstrate that myelin damage can be induced from hyperglycemia exposure (glucose at 45 mM) and mitigated by epalrestat (1µM) supplementation. Through fibrin embedding of the platform, we were able to create 3D anisotropic myelinated tissue, reaching over 6.5 mm in length. Finally, as a proof-of-concept, we incorporated pancreatic pseudoislets and endometrial organoids into our nerve platform, to demonstrate the potential in generating nociceptor innervation models. In summary, we propose here an improved tool for neurobiology research with potential applications in pathology modelling, drug screening and target tissue innervation.
Collapse
Affiliation(s)
- Afonso Malheiro
- Complex Tissue Regeneration department, Maastricht University, Universiteitssingel 40, Maastricht, 6200 MD, NETHERLANDS
| | - Abhishek Harichandan
- Complex Tissue Regeneration department, Maastricht University, Universiteitssingel, 40, Maastricht, 6200 MD, NETHERLANDS
| | - Joyce Bernardi
- Department of Cardiology, Maastricht University, Universiteitssingel 50, Maastricht, 6200 MD, NETHERLANDS
| | - Adrián Seijas-Gamardo
- Complex Tissue Regeneration department, Maastricht University, Universiteitssingel 40, Maastricht, 6200 MD, NETHERLANDS
| | - Gonda F Konings
- Department of Gynaecology, Maastricht University, Universiteitssingel 50, Maastricht, 6200 MD, NETHERLANDS
| | - Paul G A Volders
- Department of Cardiology, Maastricht University, Universiteitssingel 50, Maastricht, 6200 MD, NETHERLANDS
| | - Andrea Romano
- Department of Gynaecology, Maastricht University, Universiteitssingel 50, Maastricht, 6200 MD, NETHERLANDS
| | - Carlos Mota
- Department of Complex Tissue Regeneration (CTR), Maastricht University, Universiteitssingel, 40, office 3.541A, Maastricht, 6229 ER, NETHERLANDS
| | - Paul Wieringa
- Complex Tissue Regeneration, Maastricht University MERLN Institute for Technology-Inspired Regenerative Medicine, Universiteitssingel 40, Maastricht, 6229ER, NETHERLANDS
| | - Lorenzo Moroni
- Complex Tissue Regeneration, Maastricht University, Universiteitsingel, 40, Maastricht, 6200 MD, NETHERLANDS
| |
Collapse
|
43
|
Zeidler M, Kummer KK, Schöpf CL, Kalpachidou T, Kern G, Cader MZ, Kress M. NOCICEPTRA: Gene and microRNA Signatures and Their Trajectories Characterizing Human iPSC-Derived Nociceptor Maturation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2102354. [PMID: 34486248 PMCID: PMC8564443 DOI: 10.1002/advs.202102354] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Indexed: 05/07/2023]
Abstract
Nociceptors are primary afferent neurons serving the reception of acute pain but also the transit into maladaptive pain disorders. Since native human nociceptors are hardly available for mechanistic functional research, and rodent models do not necessarily mirror human pathologies in all aspects, human induced pluripotent stem cell-derived nociceptors (iDN) offer superior advantages as a human model system. Unbiased mRNA::microRNA co-sequencing, immunofluorescence staining, and qPCR validations, reveal expression trajectories as well as miRNA target spaces throughout the transition of pluripotent cells into iDNs. mRNA and miRNA candidates emerge as regulatory hubs for neurite outgrowth, synapse development, and ion channel expression. The exploratory data analysis tool NOCICEPTRA is provided as a containerized platform to retrieve experimentally determined expression trajectories, and to query custom gene sets for pathway and disease enrichments. Querying NOCICEPTRA for marker genes of cortical neurogenesis reveals distinct similarities and differences for cortical and peripheral neurons. The platform provides a public domain neuroresource to exploit the entire data sets and explore miRNA and mRNA as hubs regulating human nociceptor differentiation and function.
Collapse
Affiliation(s)
- Maximilian Zeidler
- Institute of PhysiologyMedical University of InnsbruckInnsbruck6020Austria
| | - Kai K. Kummer
- Institute of PhysiologyMedical University of InnsbruckInnsbruck6020Austria
| | - Clemens L. Schöpf
- Institute of PhysiologyMedical University of InnsbruckInnsbruck6020Austria
| | | | - Georg Kern
- Institute of PhysiologyMedical University of InnsbruckInnsbruck6020Austria
| | - M. Zameel Cader
- Weatherall Institute of Molecular MedicineUniversity of OxfordOxfordOX3 9DSUK
| | - Michaela Kress
- Institute of PhysiologyMedical University of InnsbruckInnsbruck6020Austria
| |
Collapse
|
44
|
The insecticide deltamethrin enhances sodium channel slow inactivation of human Nav1.9, Nav1.8 and Nav1.7. Toxicol Appl Pharmacol 2021; 428:115676. [PMID: 34389319 DOI: 10.1016/j.taap.2021.115676] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 07/13/2021] [Accepted: 08/08/2021] [Indexed: 01/05/2023]
Abstract
The insecticide deltamethrin of the pyrethroid class mainly targets voltage-gated sodium channels (Navs). Deltamethrin prolongs the opening of Navs by slowing down fast inactivation and deactivation. Pyrethroids are supposedly safe for humans, however, they have also been linked to the gulf-war syndrome, a neuropathic pain condition that can develop following exposure to certain chemicals. Inherited neuropathic pain conditions have been linked to mutations in the Nav subtypes Nav1.7, Nav1.8, and Nav1.9. Here, we examined the effect of deltamethrin on the human isoforms Nav1.7, Nav1.8, and Nav1.9_C4 (chimera containing the C-terminus of rat Nav1.4) heterologously expressed in HEK293T and ND7/23 cells using whole-cell patch-clamp electrophysiology. For all three Nav subtypes, we observed increased persistent and tail currents that are typical for Nav channels modified by deltamethrin. The most surprising finding was an enhanced slow inactivation induced by deltamethrin in all three Nav subtypes. An enhanced slow inactivation is contrary to the prolonged opening caused by pyrethroids and has not been described for deltamethrin or any other pyrethroid before. Furthermore, we found that the fraction of deltamethrin-modified channels increased use-dependently. However, for Nav1.8, the use-dependent potentiation occurred only when the holding potential was increased to -90 mV, a potential at which the tail currents decay more slowly. This indicates that use-dependent modification is due to an accumulation of tail currents. In summary, our findings support a novel mechanism whereby deltamethrin enhances slow inactivation of voltage-gated sodium channels, which may, depending on the cellular resting membrane potential, reduce neuronal excitability and counteract the well-described pyrethroid effects of prolonging channel opening.
Collapse
|
45
|
Chrysostomidou L, Cooper AH, Weir GA. Cellular models of pain: New technologies and their potential to progress preclinical research. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2021; 10:100063. [PMID: 34977426 PMCID: PMC8683679 DOI: 10.1016/j.ynpai.2021.100063] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 05/12/2021] [Accepted: 05/15/2021] [Indexed: 01/16/2023]
Abstract
Human sensory neurons can reduce the translational gap in analgesic development. Access to dorsal root ganglion (hDRG) neurons is increasing. Diverse sensory neuron subtypes can now be generated via stem cell technology. Advances of these technologies will improve our understanding of human nociception.
In vitro models fill a vital niche in preclinical pain research, allowing detailed study of molecular pathways, and in the case of humanised systems, providing a translational bridge between in vivo animal models and human patients. Significant advances in cellular technology available to basic pain researchers have occurred in the last decade, including developing protocols to differentiate sensory neuron-like cells from stem cells and greater access to human dorsal root ganglion tissue. In this review, we discuss the use of both models in preclinical pain research: What can a human sensory neuron in a dish tell us that rodent in vivo models cannot? How similar are these models to their endogenous counterparts, and how should we judge them? What limitations do we need to consider? How can we leverage cell models to improve translational success? In vitro human sensory neuron models equip pain researchers with a valuable tool to investigate human nociception. With continual development, consideration for their advantages and limitations, and effective integration with other experimental strategies, they could become a driving force for the pain field's advancement.
Collapse
Affiliation(s)
- Lina Chrysostomidou
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Andrew H Cooper
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Greg A Weir
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
46
|
Regulation and drug modulation of a voltage-gated sodium channel: Pivotal role of the S4-S5 linker in activation and slow inactivation. Proc Natl Acad Sci U S A 2021; 118:2102285118. [PMID: 34260401 PMCID: PMC8285963 DOI: 10.1073/pnas.2102285118] [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: 12/05/2022] Open
Abstract
Voltage-gated sodium channels initiate electric signals in cell communications. The S4–S5 linker between the voltage-sensing and pore modules transmits depolarization signals to trigger channel activation. The mechanisms of this action, however, remain elusive. By combining biophysical and computational approaches, we identify a critical residue, T140, in the S4–S5 linker of the bacterial sodium channel NaChBac, which plays a pivotal role in channel activation and drug modulation of slow inactivation. Specifically, we discovered conformation-dependent drug binding at this site and unveiled a toggling mode of action by T140, which switches interaction partners with different S6 residues to regulate channel activation and slow inactivation. These observations suggest the possibility of conformation-specific drugs targeting the gating machinery of voltage-gated ion channels. Voltage-gated sodium (NaV) channels control excitable cell functions. While structural investigations have revealed conformation details of different functional states, the mechanisms of both activation and slow inactivation remain unclear. Here, we identify residue T140 in the S4–S5 linker of the bacterial voltage-gated sodium channel NaChBac as critical for channel activation and drug effects on inactivation. Mutations at T140 either attenuate activation or render the channel nonfunctional. Propofol, a clinical anesthetic known to inhibit NaChBac by promoting slow inactivation, binds to a pocket between the S4–S5 linker and S6 helix in a conformation-dependent manner. Using 19F-NMR to quantify site-specific binding by saturation transfer differences (STDs), we found strong STDs in inactivated, but not activated, NaChBac. Molecular dynamics simulations show a highly dynamic pocket in the activated conformation, limiting STD buildup. In contrast, drug binding to this pocket promotes and stabilizes the inactivated states. Our results provide direct experimental evidence showing distinctly different associations between the S4–S5 linker and S6 helix in activated and inactivated states. Specifically, an exchange occurs between interaction partners T140 and N234 of the same subunit in activation, and T140 and N225 of the domain-swapped subunit in slow inactivation. The drug action on slow inactivation of prokaryotic NaV channels seems to have a mechanism similar to the recently proposed “door-wedge” action of the isoleucine-phenylalanine-methionine (IFM) motif on the fast inactivation of eukaryotic NaV channels. Elucidating this gating mechanism points to a possible direction for conformation-dependent drug development.
Collapse
|
47
|
Alvarez P, Bogen O, Green PG, Levine JD. Nociceptor Overexpression of Na V1.7 Contributes to Chronic Muscle Pain Induced by Early-Life Stress. THE JOURNAL OF PAIN 2021; 22:806-816. [PMID: 33636374 PMCID: PMC8406703 DOI: 10.1016/j.jpain.2021.02.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 01/27/2021] [Accepted: 02/07/2021] [Indexed: 01/06/2023]
Abstract
Adult rats previously submitted to neonatal limited bedding (NLB), a model of early-life stress, display muscle mechanical hyperalgesia and nociceptor hyperexcitability, the underlying mechanism for which is unknown. Since voltage-gated sodium channel subtype 7 (NaV1.7) contributes to mechanical hyperalgesia in several preclinical pain models and is critical for nociceptor excitability, we explored its role in the muscle hyperalgesia exhibited by adult NLB rats. Western blot analyses demonstrated increased NaV1.7 protein expression in L4-L5 dorsal root ganglia (DRG) from adult NLB rats, and antisense oligodeoxynucleotide (AS ODN) targeting NaV1.7 alpha subunit mRNA attenuated the expression of NaV1.7 in DRG extracts. While this AS ODN did not affect nociceptive threshold in normal rats it significantly attenuated hyperalgesia in NLB rats. The selective NaV1.7 activator OD1 produced dose-dependent mechanical hyperalgesia that was enhanced in NLB rats, whereas the NaV1.7 blocker ProTx-II prevented OD1-induced hyperalgesia in control rats and ongoing hyperalgesia in NLB rats. AS ODN knockdown of extracellular signal-regulated kinase 1/2, which enhances NaV1.7 function, also inhibited mechanical hyperalgesia in NLB rats. Our results support the hypothesis that overexpression of NaV1.7 in muscle nociceptors play a role in chronic muscle pain induced by early-life stress, suggesting that NaV1.7 is a target for the treatment of chronic muscle pain. PERSPECTIVE: We demonstrate that early-life adversity, induced by exposure to inconsistent maternal care, produces chronic muscle hyperalgesia, which depends, at least in part, on increased expression of NaV1.7 in nociceptors.
Collapse
Affiliation(s)
- Pedro Alvarez
- Department of Oral and Maxillofacial Surgery, University of California, San Francisco, San Francisco, California
| | - Oliver Bogen
- Department of Oral and Maxillofacial Surgery, University of California, San Francisco, San Francisco, California; UCSF Pain and Addiction Research Center, University of California, San Francisco, San Francisco, California
| | - Paul G Green
- Department of Oral and Maxillofacial Surgery, University of California, San Francisco, San Francisco, California; UCSF Pain and Addiction Research Center, University of California, San Francisco, San Francisco, California; Department of Preventative and Restorative Dental Sciences, University of California, San Francisco, San Francisco, California
| | - Jon D Levine
- Department of Oral and Maxillofacial Surgery, University of California, San Francisco, San Francisco, California; UCSF Pain and Addiction Research Center, University of California, San Francisco, San Francisco, California; Department of Medicine, University of California San Francisco, San Francisco, California.
| |
Collapse
|
48
|
Middleton SJ, Barry AM, Comini M, Li Y, Ray PR, Shiers S, Themistocleous AC, Uhelski ML, Yang X, Dougherty PM, Price TJ, Bennett DL. Studying human nociceptors: from fundamentals to clinic. Brain 2021; 144:1312-1335. [PMID: 34128530 PMCID: PMC8219361 DOI: 10.1093/brain/awab048] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 11/26/2020] [Accepted: 12/08/2020] [Indexed: 12/14/2022] Open
Abstract
Chronic pain affects one in five of the general population and is the third most important cause of disability-adjusted life-years globally. Unfortunately, treatment remains inadequate due to poor efficacy and tolerability. There has been a failure in translating promising preclinical drug targets into clinic use. This reflects challenges across the whole drug development pathway, from preclinical models to trial design. Nociceptors remain an attractive therapeutic target: their sensitization makes an important contribution to many chronic pain states, they are located outside the blood-brain barrier, and they are relatively specific. The past decade has seen significant advances in the techniques available to study human nociceptors, including: the use of corneal confocal microscopy and biopsy samples to observe nociceptor morphology, the culture of human nociceptors (either from surgical or post-mortem tissue or using human induced pluripotent stem cell derived nociceptors), the application of high throughput technologies such as transcriptomics, the in vitro and in vivo electrophysiological characterization through microneurography, and the correlation with pain percepts provided by quantitative sensory testing. Genome editing in human induced pluripotent stem cell-derived nociceptors enables the interrogation of the causal role of genes in the regulation of nociceptor function. Both human and rodent nociceptors are more heterogeneous at a molecular level than previously appreciated, and while we find that there are broad similarities between human and rodent nociceptors there are also important differences involving ion channel function, expression, and cellular excitability. These technological advances have emphasized the maladaptive plastic changes occurring in human nociceptors following injury that contribute to chronic pain. Studying human nociceptors has revealed new therapeutic targets for the suppression of chronic pain and enhanced repair. Cellular models of human nociceptors have enabled the screening of small molecule and gene therapy approaches on nociceptor function, and in some cases have enabled correlation with clinical outcomes. Undoubtedly, challenges remain. Many of these techniques are difficult to implement at scale, current induced pluripotent stem cell differentiation protocols do not generate the full diversity of nociceptor populations, and we still have a relatively poor understanding of inter-individual variation in nociceptors due to factors such as age, sex, or ethnicity. We hope our ability to directly investigate human nociceptors will not only aid our understanding of the fundamental neurobiology underlying acute and chronic pain but also help bridge the translational gap.
Collapse
Affiliation(s)
- Steven J Middleton
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Allison M Barry
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Maddalena Comini
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Yan Li
- Department of Anesthesia and Pain Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Pradipta R Ray
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Stephanie Shiers
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Andreas C Themistocleous
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK.,Brain Function Research Group, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2000, South Africa
| | - Megan L Uhelski
- Department of Anesthesia and Pain Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xun Yang
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| | - Patrick M Dougherty
- Brain Function Research Group, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2000, South Africa
| | - Theodore J Price
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA
| | - David L Bennett
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK
| |
Collapse
|
49
|
Kankowski S, Grothe C, Haastert-Talini K. Neuropathic pain: Spotlighting anatomy, experimental models, mechanisms, and therapeutic aspects. Eur J Neurosci 2021; 54:4475-4496. [PMID: 33942412 DOI: 10.1111/ejn.15266] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 04/19/2021] [Accepted: 04/28/2021] [Indexed: 12/30/2022]
Abstract
The International Association for the Study of Pain defines neuropathic pain as "pain arising as a direct consequence of a lesion or disease affecting the somatosensory system". The associated changes can be observed in the peripheral as well as the central nervous system. The available literature discusses a wide variety of causes as predisposing for the development and amplification of neuropathic pain. Further, key interactions within sensory pathways have been discovered, but no common molecular mechanism leading to neuropathic pain has been identified until now. In the first part of this review, the pain mediating lateral spinothalamic tract is described. Different in vivo models are presented that allow studying trauma-, chemotherapy-, virus-, and diabetes-induced neuropathic pain in rodents. We furthermore discuss approaches to assess neuropathic pain in these models. Second, the current knowledge about cellular and molecular mechanisms suggested to underlie the development of neuropathic pain is presented and discussed. A summary of established therapies that are already applied in the clinic and novel, promising approaches closes the paper. In conclusion, the established animal models are able to emulate the diversity of neuropathic pain observed in the clinics. However, the assessment of neuropathic pain in the presented in vivo models should be improved. The determination of common molecular markers with suitable in vitro models would simplify the assessment of neuropathic pain in vivo. This would furthermore provide insights into common molecular mechanisms of the disease and establish a basis to search for satisfying therapeutic approaches.
Collapse
Affiliation(s)
- Svenja Kankowski
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School (MHH), Hannover, Germany
| | - Claudia Grothe
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School (MHH), Hannover, Germany.,Center for Systems Neuroscience (ZNS) Hannover, Hannover, Germany
| | - Kirsten Haastert-Talini
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School (MHH), Hannover, Germany.,Center for Systems Neuroscience (ZNS) Hannover, Hannover, Germany
| |
Collapse
|
50
|
Schinke C, Fernandez Vallone V, Ivanov A, Peng Y, Körtvelyessy P, Nolte L, Huehnchen P, Beule D, Stachelscheid H, Boehmerle W, Endres M. Modeling chemotherapy induced neurotoxicity with human induced pluripotent stem cell (iPSC) -derived sensory neurons. Neurobiol Dis 2021; 155:105391. [PMID: 33984509 DOI: 10.1016/j.nbd.2021.105391] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 05/06/2021] [Accepted: 05/07/2021] [Indexed: 12/20/2022] Open
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a frequent, potentially irreversible adverse effect of cytotoxic chemotherapy often leading to a reduction or discontinuation of treatment which negatively impacts patients' prognosis. To date, however, neither predictive biomarkers nor preventive treatments for CIPN are available, which is partially due to a lack of suitable experimental models. We therefore aimed to evaluate whether sensory neurons derived from induced pluripotent stem cells (iPSC-DSN) can serve as human disease model system for CIPN. Treatment of iPSC-DSN for 24 h with the neurotoxic drugs paclitaxel, bortezomib, vincristine and cisplatin led to axonal blebbing and a dose dependent decline of cell viability in clinically relevant IC50 ranges, which was not observed for the non-neurotoxic compounds doxorubicin and 5-fluorouracil. Paclitaxel treatment effects were less pronounced after 24 h but prominent when treatment was applied for 72 h. Global transcriptome analyses performed at 24 h, i.e. before paclitaxel-induced cell death occurred, revealed the differential expression of genes of neuronal injury, cellular stress response, and sterol pathways. We further evaluated if known neuroprotective strategies can be reproduced in iPSC-DSN and observed protective effects of lithium replicating findings from rodent dorsal root ganglia cells. Comparing sensory neurons derived from two different healthy donors, we found preliminary evidence that these cell lines react differentially to neurotoxic drugs as expected from the variable presentation of CIPN in patients. In conclusion, iPSC-DSN are a promising platform to study the pathogenesis of CIPN and to evaluate neuroprotective treatment strategies. In the future, the application of patient-specific iPSC-DSN could open new avenues for personalized medicine with individual risk prediction, choice of chemotherapeutic compounds and preventive treatments.
Collapse
Affiliation(s)
- Christian Schinke
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Klinik und Hochschulambulanz für Neurologie, Charitéplatz 1, 10117 Berlin, Germany; Berlin Institute of Health at Charité, Universitätsmedizin Berlin, Anna-Louisa-Karsch Straße 2, 10178 Berlin, Germany
| | - Valeria Fernandez Vallone
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Stem Cell Core Facility, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Andranik Ivanov
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Core Unit Bioinformatics, Charitéplatz 1, 10117 Berlin, Germany
| | - Yangfan Peng
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Klinik und Hochschulambulanz für Neurologie, Charitéplatz 1, 10117 Berlin, Germany; Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institut für Neurophysiologie, Charitéplatz 1, 10117 Berlin, Germany
| | - Péter Körtvelyessy
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Klinik und Hochschulambulanz für Neurologie, Charitéplatz 1, 10117 Berlin, Germany; Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Neuropathology, Charitéplatz 1, 10117 Berlin, Germany; German Center for Neurodegenerative Diseases, 39120 Magdeburg, Germany
| | - Luca Nolte
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Klinik und Hochschulambulanz für Neurologie, Charitéplatz 1, 10117 Berlin, Germany
| | - Petra Huehnchen
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Klinik und Hochschulambulanz für Neurologie, Charitéplatz 1, 10117 Berlin, Germany; Berlin Institute of Health at Charité, Universitätsmedizin Berlin, Anna-Louisa-Karsch Straße 2, 10178 Berlin, Germany; Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, NeuroCure Cluster of Excellence, Charitéplatz 1, 10117 Berlin, Germany
| | - Dieter Beule
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Core Unit Bioinformatics, Charitéplatz 1, 10117 Berlin, Germany; Max-Delbrueck Center for Molecular Medicine, 13125 Berlin, Germany
| | - Harald Stachelscheid
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Stem Cell Core Facility, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Wolfgang Boehmerle
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Klinik und Hochschulambulanz für Neurologie, Charitéplatz 1, 10117 Berlin, Germany; Berlin Institute of Health at Charité, Universitätsmedizin Berlin, Anna-Louisa-Karsch Straße 2, 10178 Berlin, Germany; Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, NeuroCure Cluster of Excellence, Charitéplatz 1, 10117 Berlin, Germany.
| | - Matthias Endres
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Klinik und Hochschulambulanz für Neurologie, Charitéplatz 1, 10117 Berlin, Germany; Berlin Institute of Health at Charité, Universitätsmedizin Berlin, Anna-Louisa-Karsch Straße 2, 10178 Berlin, Germany; Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, NeuroCure Cluster of Excellence, Charitéplatz 1, 10117 Berlin, Germany; Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Center for Stroke Research Berlin, Charitéplatz 1, 10117 Berlin, Germany; German Center for Neurodegenerative Diseases (DZNE), partner site Berlin, Germany; German Center for Cardiovascular Research (DZHK), partner site Berlin, Germany
| |
Collapse
|