1
|
Liu F, Liao H, Fang Z, Tang Q, Liu Y, Li C, Zhou C, Zhang Y, Shen J. MicroRNA-6954-3p Downregulation Contributes to Orofacial Neuropathic Pain in Mice Via Targeting Voltage-Gated Sodium Channel β2 Subunit Protein. THE JOURNAL OF PAIN 2024; 25:104598. [PMID: 38866121 DOI: 10.1016/j.jpain.2024.104598] [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: 12/16/2023] [Revised: 05/30/2024] [Accepted: 06/03/2024] [Indexed: 06/14/2024]
Abstract
The voltage-gated sodium channel β2 subunit protein (SCN2B) plays a crucial role in neuropathic pain. However, the role and mechanisms of SCN2B in orofacial neuropathic pain are still unclear. This study aimed to investigate the upstream regulatory mechanisms of SCN2B in the trigeminal ganglion (TG) underlying orofacial neuropathic pain. Chronic constriction injury of the infraorbital nerve (CCI-ION) of mice was performed to establish the model of orofacial neuropathic pain. Von Frey filament test was performed to detect the head withdrawal threshold (HWT) of mice. Quantitative reverse transcription-polymerase chain, western blotting (WB), fluorescence in situ hybridization, and immunofluorescence (IF) staining were used to detect the expression and distribution of SCN2B and miR-6954-3p in the TG of mice. A luciferase activity assay was carried out to prove the binding between SCN2B messenger ribonucleic acid (mRNA) and miR-6954-3p. After the CCI-ION surgery, the levels of Scn2b mRNA and protein significantly increased and miR-6954-3p decreased in the TG of mice with decreasing HWT. IF staining revealed that SCN2B was expressed specifically in the TG neurons. Silencing SCN2B in the TG of CCI-ION mice significantly increased the HWT. Importantly, the 3'-untranslated region of Scn2b mRNA was proved to bind with miR-6954-3p. Fluorescence in situ hybridization and IF staining demonstrated that miR-6954-3p was expressed in TG neurons and co-expressed with SCN2B. Furthermore, intraganglionic injection of miR-6954-3p agomir into the TG of CCI-ION mice resulted in the downregulation of SCN2B and increased the HWT. These findings suggest that the downregulation of miR-6954-3p in the TG promotes orofacial neuropathic pain by promoting SCN2B expression following trigeminal nerve injury. PERSPECTIVE: This study points to the important role of SCN2B in orofacial neuropathic pain. Furthermore, miR-6954-3p is proven to regulate the expression of SCN2B by binding to the 3'-untranslated region of Scn2b mRNA. These findings indicate that SCN2B and miR-6954-3p are potential therapeutic targets for the treatment of orofacial neuropathic pain.
Collapse
Affiliation(s)
- Fei Liu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Honglin Liao
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Zhonghan Fang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Qingfeng Tang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yajing Liu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Chunjie Li
- Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Chen Zhou
- Laboratory of Anesthesia and Critical Care Medicine & Translational Neuroscience Center & West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yanyan Zhang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Jiefei Shen
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
2
|
Talluri B, Addya S, Terashvili M, Medda BK, Banerjee A, Shaker R, Sengupta JN, Banerjee B. Adult zymosan re-exposure exacerbates the molecular alterations in the brainstem rostral ventromedial medulla of rats with early life zymosan-induced cystitis. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2024; 16:100160. [PMID: 39252992 PMCID: PMC11381896 DOI: 10.1016/j.ynpai.2024.100160] [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: 06/06/2024] [Revised: 07/23/2024] [Accepted: 07/23/2024] [Indexed: 09/11/2024]
Abstract
Recent evidence suggests that the descending modulatory pathways from the brainstem rostral ventromedial medulla (RVM) are important for bladder inflammatory pain. This study aimed to identify the long-term molecular changes in RVM neurons due to early life cystitis during neuronal development and the effect of reexposure later in adulthood. RVM tissues from two treatment protocols were used: (1) neonatal zymosan exposures with acute adult rechallenge (RC) and (2) only neonatal zymosan exposures (NRC). RNAseq analysis showed upregulation of several genes associated with synaptic plasticity (Grin1, Grip2, Notch1, Arc, and Scn2b) in the cystitis groups compared to controls in both protocols. The RC protocol exhibited a stronger treatment effect with significantly higher fold differences between the groups compared to the NRC protocol (p < 0.001, fold differences RC vs NRC). In microarrays, miR-34a-5p showed cystitis-induced downregulation in both protocols. Bioinformatics analysis identified multiple 3'UTRs complementary binding sites for miR-34a-5p on Grin2b, Notch1, Grip2, Scn2b, and Arc genes. The enhanced response in the RC protocol indicates a possible priming effect of early life cystitis on rechallenge in adulthood. These long-term molecular alterations may play a critical role in the development of chronic bladder pain conditions as seen in patients with Interstitial Cystitis/Bladder pain syndrome.
Collapse
Affiliation(s)
- Bhavana Talluri
- Gastroenterology & Hepatology Division, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Sankar Addya
- Sydney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Maia Terashvili
- Gastroenterology & Hepatology Division, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Bidyut K Medda
- Gastroenterology & Hepatology Division, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Anjishnu Banerjee
- Division of Biostatistics, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Reza Shaker
- Gastroenterology & Hepatology Division, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Jyoti N Sengupta
- Gastroenterology & Hepatology Division, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Banani Banerjee
- Gastroenterology & Hepatology Division, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
3
|
Sotelo-Hitschfeld P, Bernal L, Nazeri M, Renthal W, Brauchi S, Roza C, Zimmermann K. Comparative Gene Signature of Nociceptors Innervating Mouse Molar Teeth, Cranial Meninges, and Cornea. Anesth Analg 2024; 139:226-234. [PMID: 38236765 DOI: 10.1213/ane.0000000000006816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/19/2024]
Abstract
BACKGROUND The trigeminal ganglion (TG) collects afferent sensory information from various tissues. Recent large-scale RNA sequencing of neurons of the TG and dorsal root ganglion has revealed a variety of functionally distinct neuronal subpopulations, but organ-specific information is lacking. METHODS To link transcriptomic and tissue-specific information, we labeled small-diameter neurons of 3 specific subpopulations of the TG by local application of lipophilic carbocyanine dyes to their innervation site in the dental pulp, cornea, and meninges (dura mater). We then collected mRNA-sequencing data from fluorescent neurons. Differentially expressed genes (DEGs) were analyzed and subjected to downstream gene set enrichment analysis (GSEA), and ion channel profiling was performed. RESULTS A total of 10,903 genes were mapped to the mouse genome (>500 reads). DEG analysis revealed 18 and 81 genes with differential expression (log 2 fold change > 2, Padj < .05) in primary afferent neurons innervating the dental pulp (dental primary afferent neurons [DPAN]) compared to those innervating the meninges (meningeal primary afferent neurons [MPAN]) and the cornea (corneal primary afferent neurons [CPAN]). We found 250 and 292 genes differentially expressed in MPAN as compared to DPAN and to CPAN, and 21 and 12 in CPAN as compared to DPAN and MPAN. Scn2b had the highest log 2 fold change when comparing DPAN versus MPAN and Mmp12 was the most prominent DEG when comparing DPAN versus CPAN and, CPAN versus MPAN. GSEA revealed genes of the immune and mitochondrial oxidative phosphorylation system for the DPAN versus MPAN comparison, cilium- and ribosome-related genes for the CPAN versus DPAN comparison, and respirasome, immune cell- and ribosome-related gene sets for the CPAN versus MPAN comparison. DEG analysis for ion channels revealed no significant differences between the neurons set except for the sodium voltage-gated channel beta subunit 2, Scn2b . However, in each tissue a few ion channels turned up with robust number of reads. In DPAN, these were Cacna1b , Trpv2 , Cnga4 , Hcn1 , and Hcn3 , in CPAN Trpa1 , Trpv1 , Cacna1a , and Kcnk13 and in MPAN Trpv2 and Scn11a . CONCLUSIONS Our study uncovers previously unknown differences in gene expression between sensory neuron subpopulations from the dental pulp, cornea, and dura mater and provides the basis for functional studies, including the investigation of ion channel function and their suitability as targets for tissue-specific analgesia.
Collapse
Affiliation(s)
- Pamela Sotelo-Hitschfeld
- From the Department of Anesthesiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Institute of Physiology and Millennium Nucleus of Ion Channel-Associated Diseases, Universidad Austral de Chile, Valdivia, Chile
| | - Laura Bernal
- From the Department of Anesthesiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Departamento de Biología de Sistemas, Facultad de Medicina, Universidad de Alcalá, Madrid, Spain
| | - Masoud Nazeri
- From the Department of Anesthesiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - William Renthal
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Sebastian Brauchi
- Institute of Physiology and Millennium Nucleus of Ion Channel-Associated Diseases, Universidad Austral de Chile, Valdivia, Chile
| | - Carolina Roza
- Departamento de Biología de Sistemas, Facultad de Medicina, Universidad de Alcalá, Madrid, Spain
| | - Katharina Zimmermann
- From the Department of Anesthesiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
4
|
van der Heijden RA, Biswal S. Up-and-coming Radiotracers for Imaging Pain Generators. Semin Musculoskelet Radiol 2023; 27:661-675. [PMID: 37935213 PMCID: PMC10629993 DOI: 10.1055/s-0043-1775745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2023]
Abstract
Chronic musculoskeletal pain is among the most highly prevalent diseases worldwide. Managing patients with chronic pain remains very challenging because current imaging techniques focus on morphological causes of pain that can be inaccurate and misleading. Moving away from anatomical constructs of disease, molecular imaging has emerged as a method to identify diseases according to their molecular, physiologic, or cellular signatures that can be applied to the variety of biomolecular changes that occur in nociception and pain processing and therefore have tremendous potential for precisely pinpointing the source of a patient's pain. Several molecular imaging approaches to image the painful process are now available, including imaging of voltage-gated sodium channels, calcium channels, hypermetabolic processes, the substance P receptor, the sigma-1 receptor, and imaging of macrophage trafficking. This article provides an overview of promising molecular imaging approaches for the imaging of musculoskeletal pain with a focus on preclinical methods.
Collapse
Affiliation(s)
- Rianne A. van der Heijden
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Sandip Biswal
- Department of Radiology, University of Wisconsin-Madison, Madison, Wisconsin
| |
Collapse
|
5
|
Tonello R, Silveira Prudente A, Hoon Lee S, Faith Cohen C, Xie W, Paranjpe A, Roh J, Park CK, Chung G, Strong JA, Zhang JM, Berta T. Single-cell analysis of dorsal root ganglia reveals metalloproteinase signaling in satellite glial cells and pain. Brain Behav Immun 2023; 113:401-414. [PMID: 37557960 PMCID: PMC10530626 DOI: 10.1016/j.bbi.2023.08.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 07/14/2023] [Accepted: 08/06/2023] [Indexed: 08/11/2023] Open
Abstract
Satellite glial cells (SGCs) are among the most abundant non-neuronal cells in dorsal root ganglia (DRGs) and closely envelop sensory neurons that detect painful stimuli. However, little is still known about their homeostatic activities and their contribution to pain. Using single-cell RNA sequencing (scRNA-seq), we were able to obtain a unique transcriptional profile for SGCs. We found enriched expression of the tissue inhibitor metalloproteinase 3 (TIMP3) and other metalloproteinases in SGCs. Small interfering RNA and neutralizing antibody experiments revealed that TIMP3 modulates somatosensory stimuli. TIMP3 expression decreased after paclitaxel treatment, and its rescue by delivery of a recombinant TIMP3 protein reversed and prevented paclitaxel-induced pain. We also established that paclitaxel directly impacts metalloproteinase signaling in cultured SGCs, which may be used to identify potential new treatments for pain. Therefore, our results reveal a metalloproteinase signaling pathway in SGCs for proper processing of somatosensory stimuli and potential discovery of novel pain treatments.
Collapse
Affiliation(s)
- Raquel Tonello
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH, USA
| | - Arthur Silveira Prudente
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH, USA
| | - Sang Hoon Lee
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH, USA
| | - Cinder Faith Cohen
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH, USA
| | - Wenrui Xie
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH, USA
| | - Aditi Paranjpe
- Bioinformatics Collaborative Services, Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Jueun Roh
- Department of Physiology, Gachon Pain Center, College of Medicine, Gachon University, Incheon 21936, Republic of Korea
| | - Chul-Kyu Park
- Department of Physiology, Gachon Pain Center, College of Medicine, Gachon University, Incheon 21936, Republic of Korea
| | - Gehoon Chung
- Department of Oral Physiology, School of Dentistry, Seoul National University, Seoul, Republic of Korea
| | - Judith A Strong
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH, USA
| | - Jun-Ming Zhang
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH, USA
| | - Temugin Berta
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH, USA.
| |
Collapse
|
6
|
Bigsby S, Neapetung J, Campanucci VA. Voltage-gated sodium channels in diabetic sensory neuropathy: Function, modulation, and therapeutic potential. Front Cell Neurosci 2022; 16:994585. [PMID: 36467605 PMCID: PMC9713017 DOI: 10.3389/fncel.2022.994585] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 10/11/2022] [Indexed: 10/29/2023] Open
Abstract
Voltage-gated sodium channels (Na V ) are the main contributors to action potential generation and essential players in establishing neuronal excitability. Na V channels have been widely studied in pain pathologies, including those that develop during diabetes. Diabetic sensory neuropathy (DSN) is one of the most common complications of the disease. DSN is the result of sensory nerve damage by the hyperglycemic state, resulting in a number of debilitating symptoms that have a significant negative impact in the quality of life of diabetic patients. Among those symptoms are tingling and numbness of hands and feet, as well as exacerbated pain responses to noxious and non-noxious stimuli. DSN is also a major contributor to the development of diabetic foot, which may lead to lower limb amputations in long-term diabetic patients. Unfortunately, current treatments fail to reverse or successfully manage DSN. In the current review we provide an updated report on Na V channels including structure/function and contribution to DSN. Furthermore, we summarize current research on the therapeutic potential of targeting Na V channels in pain pathologies, including DSN.
Collapse
Affiliation(s)
| | | | - Verónica A. Campanucci
- Department of Anatomy, Physiology and Pharmacology (APP), College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
7
|
Reduced Expression of Voltage-Gated Sodium Channel Beta 2 Restores Neuronal Injury and Improves Cognitive Dysfunction Induced by A β1-42. Neural Plast 2022; 2022:3995227. [PMID: 36406589 PMCID: PMC9671742 DOI: 10.1155/2022/3995227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 10/05/2022] [Accepted: 10/18/2022] [Indexed: 11/12/2022] Open
Abstract
Voltage-gated sodium channel beta 2 (Nav2.2 or Navβ2, coded by SCN2B mRNA), a gene involved in maintaining normal physiological functions of the prefrontal cortex and hippocampus, might be associated with prefrontal cortex aging and memory decline. This study investigated the effects of Navβ2 in amyloid-β 1-42- (Aβ1-42-) induced neural injury model and the potential underlying molecular mechanism. The results showed that Navβ2 knockdown restored neuronal viability of Aβ1-42-induced injury in neurons; increased the contents of brain-derived neurotrophic factor (BDNF), enzyme neprilysin (NEP) protein, and NEP enzyme activity; and effectively altered the proportions of the amyloid precursor protein (APP) metabolites including Aβ42, sAPPα, and sAPPβ, thus ameliorating cognitive dysfunction. This may be achieved through regulating NEP transcription and APP metabolism, accelerating Aβ degradation, alleviating neuronal impairment, and regulating BDNF-related signal pathways to repair neuronal synaptic efficiency. This study provides novel evidence indicating that Navβ2 plays crucial roles in the repair of neuronal injury induced by Aβ1-42 both in vivo and in vitro.
Collapse
|
8
|
Liu XG. Normalization of Neuroinflammation: A New Strategy for Treatment of Persistent Pain and Memory/Emotional Deficits in Chronic Pain. J Inflamm Res 2022; 15:5201-5233. [PMID: 36110505 PMCID: PMC9469940 DOI: 10.2147/jir.s379093] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 08/18/2022] [Indexed: 12/12/2022] Open
Abstract
Chronic pain, which affects around 1/3 of the world population and is often comorbid with memory deficit and mood depression, is a leading source of suffering and disability. Studies in past decades have shown that hyperexcitability of primary sensory neurons resulting from abnormal expression of ion channels and central sensitization mediated pathological synaptic plasticity, such as long-term potentiation in spinal dorsal horn, underlie the persistent pain. The memory/emotional deficits are associated with impaired synaptic connectivity in hippocampus. Dysregulation of numerous endogenous proteins including receptors and intracellular signaling molecules is involved in the pathological processes. However, increasing knowledge contributes little to clinical treatment. Emerging evidence has demonstrated that the neuroinflammation, characterized by overproduction of pro-inflammatory cytokines and glial activation, is reliably detected in humans and animals with chronic pain, and is sufficient to induce persistent pain and memory/emotional deficits. The abnormal expression of ion channels and pathological synaptic plasticity in spinal dorsal horn and in hippocampus are resulting from neuroinflammation. The neuroinflammation is initiated and maintained by the interactions of circulating monocytes, glial cells and neurons. Obviously, unlike infectious diseases and cancer, which are caused by pathogens or malignant cells, chronic pain is resulting from alterations of cells and molecules which have numerous physiological functions. Therefore, normalization (counterbalance) but not simple inhibition of the neuroinflammation is the right strategy for treating neuronal disorders. Currently, no such agent is available in clinic. While experimental studies have demonstrated that intracellular Mg2+ deficiency is a common feature of chronic pain in animal models and supplement Mg2+ are capable of normalizing the neuroinflammation, activation of upregulated proteins that promote recovery, such as translocator protein (18k Da) or liver X receptors, has a similar effect. In this article, relevant experimental and clinical evidence is reviewed and discussed.
Collapse
Affiliation(s)
- Xian-Guo Liu
- Pain Research Center and Department of Physiology, Zhongshan School of Medicine of Sun Yat-sen University, Guangzhou, People's Republic of China
| |
Collapse
|
9
|
Zhang C, Hu MW, Wang XW, Cui X, Liu J, Huang Q, Cao X, Zhou FQ, Qian J, He SQ, Guan Y. scRNA-sequencing reveals subtype-specific transcriptomic perturbations in DRG neurons of PirtEGFPf mice in neuropathic pain condition. eLife 2022; 11:76063. [PMID: 36264609 PMCID: PMC9584610 DOI: 10.7554/elife.76063] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 10/03/2022] [Indexed: 01/22/2023] Open
Abstract
Functionally distinct subtypes/clusters of dorsal root ganglion (DRG) neurons may play different roles in nerve regeneration and pain. However, details about their transcriptomic changes under neuropathic pain conditions remain unclear. Chronic constriction injury (CCI) of the sciatic nerve represents a well-established model of neuropathic pain, and we conducted single-cell RNA-sequencing (scRNA-seq) to characterize subtype-specific perturbations of transcriptomes in lumbar DRG neurons on day 7 post-CCI. By using PirtEGFPf mice that selectively express an enhanced green fluorescent protein in DRG neurons, we established a highly efficient purification process to enrich neurons for scRNA-seq. We observed the emergence of four prominent CCI-induced clusters and a loss of marker genes in injured neurons. Importantly, a portion of injured neurons from several clusters were spared from injury-induced identity loss, suggesting subtype-specific transcriptomic changes in injured neurons. Moreover, uninjured neurons, which are necessary for mediating the evoked pain, also demonstrated cell-type-specific transcriptomic perturbations in these clusters, but not in others. Notably, male and female mice showed differential transcriptomic changes in multiple neuronal clusters after CCI, suggesting transcriptomic sexual dimorphism in DRG neurons after nerve injury. Using Fgf3 as a proof-of-principle, RNAscope study provided further evidence of increased Fgf3 in injured neurons after CCI, supporting scRNA-seq analysis, and calcium imaging study unraveled a functional role of Fgf3 in neuronal excitability. These findings may contribute to the identification of new target genes and the development of DRG neuron cell-type-specific therapies for optimizing neuropathic pain treatment and nerve regeneration.
Collapse
Affiliation(s)
- Chi Zhang
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Ming-Wen Hu
- Department of Ophthalmology, The Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Xue-Wei Wang
- Department of Orthopaedic Surgery, The Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Xiang Cui
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Jing Liu
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Qian Huang
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Xu Cao
- Department of Orthopaedic Surgery, The Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Feng-Quan Zhou
- Department of Orthopaedic Surgery, The Johns Hopkins University School of MedicineBaltimoreUnited States,The Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Jiang Qian
- Department of Ophthalmology, The Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Shao-Qiu He
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Yun Guan
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of MedicineBaltimoreUnited States,Department of Neurological Surgery, The Johns Hopkins University School of MedicineBaltimoreUnited States
| |
Collapse
|
10
|
Kouraki A, Doherty M, Fernandes GS, Zhang W, Walsh DA, Kelly A, Valdes AM. Different genes may be involved in distal and local sensitisation: a genome-wide gene-based association study and meta-analysis. Eur J Pain 2021; 26:740-753. [PMID: 34958702 PMCID: PMC9303629 DOI: 10.1002/ejp.1902] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 11/11/2021] [Accepted: 12/25/2021] [Indexed: 11/22/2022]
Abstract
Background Neuropathic pain symptoms and signs of increased pain sensitization in osteoarthritis (OA) patients may explain persistent pain after total joint replacement (TJR). Therefore, identifying genetic markers associated with pain sensitization and neuropathic‐like pain phenotypes could be clinically important in identifying targets for early intervention. Methods We performed a genome‐wide gene‐based association study (GWGAS) using pressure pain detection thresholds (PPTs) from distal pain‐free sites (anterior tibia), a measure of distal sensitization, and from proximal pain‐affected sites (lateral joint line), a measure of local sensitization, in 320 knee OA participants from the Knee Pain and related health in the Community (KPIC) cohort. We next performed gene‐based fixed‐effects meta‐analysis of PPTs and a neuropathic‐like pain phenotype using genome‐wide association study (GWAS) data from KPIC and from an independent cohort of 613 post‐TJR participants, respectively. Results The most significant genes associated with distal and local sensitization were OR5B3 and BRDT, respectively. We also found previously identified neuropathic pain‐associated genes—KCNA1, MTOR, ADORA1 and SCN3B—associated with PPT at the anterior tibia and an inflammatory pain gene—PTAFR—associated with PPT at the lateral joint line. Meta‐analysis results of anterior tibia and neuropathic‐like pain phenotypes revealed genes associated with bone morphogenesis, neuro‐inflammation, obesity, type 2 diabetes, cardiovascular disease and cognitive function. Conclusions Overall, our results suggest that different biological processes might be involved in distal and local sensitization, and common genetic mechanisms might be implicated in distal sensitization and neuropathic‐like pain. Future studies are needed to replicate these findings. Significance To the best of our knowledge, this is the first GWAS for pain sensitization and the first gene‐based meta‐analysis of pain sensitization and neuropathic‐like pain. Higher pain sensitization and neuropathic pain symptoms are associated with persistent pain after surgery hence, identifying genetic biomarkers and molecular pathways associated with these traits is clinically relevant.
Collapse
Affiliation(s)
- A Kouraki
- Academic Rheumatology, School of Medicine, University of Nottingham, Nottingham City Hospital, Nottingham, NG5 1PB, United Kingdom.,NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, NG5 1PB, United Kingdom
| | - M Doherty
- Academic Rheumatology, School of Medicine, University of Nottingham, Nottingham City Hospital, Nottingham, NG5 1PB, United Kingdom.,Pain Centre Versus Arthritis, University of Nottingham, Nottingham, NG5 1PB, United Kingdom.,Versus Arthritis Centre for Sports, Exercise and Osteoarthritis, University of Nottingham, Nottingham, NG7 2UH, United Kingdom.,NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, NG5 1PB, United Kingdom
| | - G S Fernandes
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS1 6EH, United Kingdom
| | - W Zhang
- Academic Rheumatology, School of Medicine, University of Nottingham, Nottingham City Hospital, Nottingham, NG5 1PB, United Kingdom.,Pain Centre Versus Arthritis, University of Nottingham, Nottingham, NG5 1PB, United Kingdom.,Versus Arthritis Centre for Sports, Exercise and Osteoarthritis, University of Nottingham, Nottingham, NG7 2UH, United Kingdom.,NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, NG5 1PB, United Kingdom
| | - D A Walsh
- Academic Rheumatology, School of Medicine, University of Nottingham, Nottingham City Hospital, Nottingham, NG5 1PB, United Kingdom.,Pain Centre Versus Arthritis, University of Nottingham, Nottingham, NG5 1PB, United Kingdom.,Versus Arthritis Centre for Sports, Exercise and Osteoarthritis, University of Nottingham, Nottingham, NG7 2UH, United Kingdom.,NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, NG5 1PB, United Kingdom
| | - A Kelly
- Academic Rheumatology, School of Medicine, University of Nottingham, Nottingham City Hospital, Nottingham, NG5 1PB, United Kingdom.,NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, NG5 1PB, United Kingdom
| | - A M Valdes
- Academic Rheumatology, School of Medicine, University of Nottingham, Nottingham City Hospital, Nottingham, NG5 1PB, United Kingdom.,Pain Centre Versus Arthritis, University of Nottingham, Nottingham, NG5 1PB, United Kingdom.,NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, NG5 1PB, United Kingdom
| |
Collapse
|
11
|
MicroRNA-133b-3p targets purinergic P2X4 receptor to regulate central poststroke pain in rats. Neuroscience 2021; 481:60-72. [PMID: 34688806 DOI: 10.1016/j.neuroscience.2021.10.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 10/12/2021] [Accepted: 10/13/2021] [Indexed: 01/10/2023]
Abstract
Central poststroke pain (CPSP) is a neuropathic pain syndrome that usually occurs after cerebrovascular accidents. Currently, the pathogenesis of CPSP is not fully understood. Purinergic P2X4 receptor (P2X4R) is implicated in neuropathic pain including CPSP. Herein, we demonstrated that the levels of microRNA-133b-3p (miR-133b-3p), which targets P2X4R transcripts, were significantly downregulated in the ventral posterolateral nucleus of the thalamus (VPL), cerebrospinal fluid (CSF), and plasma of CPSP rats. The expression levels of miR-133b-3p negatively correlated with the severity of allodynia. Genetic knockdown of P2X4R in the VPL protected CPSP rats against allodynia. Similarly, genetic overexpression of miR-133b-3p in the VPL reversed the allodynia established in CPSP rats via downregulation of P2X4R expression. Treatment using gabapentin in CPSP rats significantly restored the decreased miR-133b-3p expression in the VPL, CSF, and plasma and blocked allodynia in CPSP rats. The administration of an miR-133b-3p inhibitor into the VPL abolished the antiallodynic activity of gabapentin. This mechanism was associated with P2X4R expression and involved the endogenous opioid system. Human patients with CPSP showed decreased plasma levels of miR-133b-3p compared with those of control participants. Logistic regression analysis of our patient cohort showed that determining plasma levels of miR-133b-3p may be useful for CPSP diagnosis and treatment.
Collapse
|
12
|
Alsaloum M, Labau JIR, Sosniak D, Zhao P, Almomani R, Gerrits M, Hoeijmakers JGJ, Lauria G, Faber CG, Waxman SG, Dib-Hajj S. A novel gain-of-function sodium channel β2 subunit mutation in idiopathic small fiber neuropathy. J Neurophysiol 2021; 126:827-839. [PMID: 34320850 DOI: 10.1152/jn.00184.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Small fiber neuropathy (SFN) is a common condition affecting thinly myelinated Aδ and unmyelinated C fibers, often resulting in excruciating pain and dysautonomia. SFN has been associated with several conditions, but a significant number of cases have no discernible cause. Recent genetic studies have identified potentially pathogenic gain-of-function mutations in several the pore-forming voltage-gated sodium channel α subunits (NaVs) in a subset of patients with SFN, but the auxiliary sodium channel β subunits have been less implicated in the development of the disease. β subunits modulate NaV trafficking and gating, and several mutations have been linked to epilepsy and cardiac dysfunction. Recently, we provided the first evidence for the contribution of a mutation in the β2-subunit to pain in human painful diabetic neuropathy. Here, we provide the first evidence for the involvement of a sodium channel β subunit mutation in the pathogenesis of SFN with no other known causes. We show, through current-clamp analysis, that the newly-identified Y69H variant of the β2 subunit induces neuronal hyperexcitability in dorsal root ganglion neurons, lowering the threshold for action potential firing and allowing for increased repetitive action potential spiking. Underlying the hyperexcitability induced by the β2-Y69H variant, we demonstrate an upregulation in tetrodotoxin-sensitive, but not tetrodotoxin-resistant sodium currents. This provides the first evidence for the involvement of β2 subunits in SFN and strengthens the link between sodium channel β subunits and the development of neuropathic pain in humans.
Collapse
Affiliation(s)
- Matthew Alsaloum
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States.,Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT, United States.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, United States.,Yale Medical Scientist Training Program, Yale School of Medicine, New Haven, CT, United States.,Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT, United States
| | - Julie I R Labau
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States.,Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT, United States.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, United States.,Department of Genetics and Cell Biology, Clinical Genomics Unit, Maastricht University, Maastricht, the Netherlands.,Department of Neurology, Maastricht University Medical Centre+, Maastricht, the Netherlands
| | - Daniel Sosniak
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States.,Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT, United States.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, United States
| | - Peng Zhao
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States.,Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT, United States.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, United States
| | - Rowida Almomani
- Department of Genetics and Cell Biology, Clinical Genomics Unit, Maastricht University, Maastricht, the Netherlands.,Department of Medical Laboratory Sciences, Jordan University of Science and Technology, Irbid, Jordan
| | - Monique Gerrits
- Department of Clinical Genetics, Maastricht University Medical Centre+, Maastricht, the Netherlands
| | | | - Giuseppe Lauria
- Neuroalgology Unit, IRCCS Foundation "Carlo Besta" Neurological Institute, Milan, Italy.,Department of Biomedical and Clinical Sciences "Luigi Sacco," University of Milan, Milan, Italy
| | - Catherina G Faber
- Department of Neurology, Maastricht University Medical Centre+, Maastricht, the Netherlands
| | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States.,Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT, United States.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, United States
| | - Sulayman Dib-Hajj
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States.,Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT, United States.,Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, United States
| |
Collapse
|
13
|
Sánchez-Salcedo JA, Cabrera MME, Molina-Jiménez T, Cortes-Altamirano JL, Alfaro-Rodríguez A, Bonilla-Jaime H. Depression and Pain: use of antidepressant. Curr Neuropharmacol 2021; 20:384-402. [PMID: 34151765 PMCID: PMC9413796 DOI: 10.2174/1570159x19666210609161447] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 04/03/2021] [Accepted: 04/03/2021] [Indexed: 11/24/2022] Open
Abstract
Background: Emotional disorders are common comorbid affectations that exacerbate the severity and persistence of chronic pain. Specifically, depressive symptoms can lead to an excessive duration and intensity of pain. Clinical and preclinical studies have been focused on the underlying mechanisms of chronic pain and depression comorbidity and the use of antidepressants to reduce pain. Aim: This review provides an overview of the comorbid relationship of chronic pain and depression, the clinical and pre-clinical studies performed on the neurobiological aspects of pain and depression, and the use of antidepressants as analgesics. Methods: A systematic search of literature databases was conducted according to pre-defined criteria. The authors independently conducted a focused analysis of the full-text articles. Results: Studies suggest that pain and depression are highly intertwined and may co-exacerbate physical and psychological symptoms. One important biochemical basis for pain and depression focuses on the serotonergic and norepinephrine system, which have been shown to play an important role in this comorbidity. Brain structures that codify pain are also involved in mood. It is evident that using serotonergic and norepinephrine antidepressants are strategies commonly employed to mitigate pain Conclusion: Literature indicates that pain and depression impact each other and play a prominent role in the development and maintenance of other chronic symptoms. Antidepressants continue to be a major therapeutic tool for managing chronic pain. Tricyclic antidepressants (TCAs) are more effective in reducing pain than Selective Serotonin Reuptake Inhibitors (SSRIs) and Serotonin-Noradrenaline Reuptake Inhibitors (SNRIs).
Collapse
Affiliation(s)
- José Armando Sánchez-Salcedo
- Doctorado en Ciencias Biológicas y de la Salud. Universidad Autónoma Metropolitana-Iztapalapa, UAM-I, Apartado Postal 55 535, C.P. 09340, Ciudad de México, Mexico
| | - Maribel Maetizi Estevez Cabrera
- Doctorado en Ciencias Biológicas y de la Salud. Universidad Autónoma Metropolitana-Iztapalapa, UAM-I, Apartado Postal 55 535, C.P. 09340, Ciudad de México, Mexico
| | - Tania Molina-Jiménez
- Facultad de Química Farmacéutica Biológica, Universidad Veracruzana. Circuito Gonzálo Aguirre Beltrán Sn, Zona Universitaria. C.P. 91090 Xalapa-Enríquez
| | - José Luis Cortes-Altamirano
- División de Neurociencias, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", Secretaría de Salud, Ciudad de México, Mexico
| | - Alfonso Alfaro-Rodríguez
- División de Neurociencias, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", Secretaría de Salud, Ciudad de México, Mexico
| | - Herlinda Bonilla-Jaime
- Departamento de Biología de la Reproducción, Universidad Autónoma Metropolitana-Iztapalapa. Apartado Postal 55 535, C.P. 09340, Ciudad de México, Mexico
| |
Collapse
|
14
|
Mujadzic T, Friedman HI, Mujadzic MM, Gober C, Chen E, Atwez A, Durkin M, Mujadzic MM. Modified Carpal Tunnel Release: A New Approach to Minimizing Pillar Pain. Ann Plast Surg 2021; 86:S503-S509. [PMID: 34100807 DOI: 10.1097/sap.0000000000002885] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Pillar pain is a frequent postoperative complication of carpal tunnel release (CTR). The precise definition of pillar pain is lacking, but most authors describe it as diffuse aching pain and tenderness in the thenar and hypothenar area. The etiology of pillar pain is unclear. However, the most prevalent theory is the neurogenic theory, which attributes the pain to the damage of small nerve branches of palmar cutaneous branches of median nerve after surgical incision, with resulting entrapment of the nerves in the scar tissue at the incision site. We postulated that a main source of pillar pain is sensory neuromas along the incision site.In this article, we describe a simple modification of the standard CTR technique with intent to decrease neuroma formation and thus minimizing pillar pain. MATERIALS AND METHODS This is a retrospective study comparing the incidence and duration of pillar pain between patients who underwent standard CTR (SCTR, n = 53) versus the minimizing pillar pain CTR technique (n = 55). Based on duration of pillar pain, the groups were placed into 3 subgroups (<3, 3-6, and >6 months). Presence and duration of pillar pain in each group were recorded along with return to work (RTW), complications, and patient satisfaction. RESULTS The SCTR group had a total of 17 patients with pillar pain (32.1%), 5 of which resolved within 3 months, 7 within 3 to 6 months, and 5 in more than 6 months. The group that underwent the minimizing pillar pain technique had a total of 4 patients with pillar pain (7.2%). Three resolved within 3 months, 1 resolved within 3 to 6 months, and there were no patients with pillar pain lasting more than 6 months. Average RTW time for minimization of pillar pain CTR (MPPCTR) was 34.9 days. Average RTW time for SCTR was 54.8 days. Satisfaction was higher among patients who underwent surgery with MPPCTR. CONCLUSIONS Based on these results, we concluded that MPPCTR compared with SCTR had equal complication rate, however, significantly lower incidence and duration of pillar pain, higher rate of satisfaction, and earlier RTW.
Collapse
Affiliation(s)
- Tarik Mujadzic
- From the Prisma Health/University of South Carolina School of Medicine, Columbia SC
| | - Harold I Friedman
- From the Prisma Health/University of South Carolina School of Medicine, Columbia SC
| | | | | | - Elliot Chen
- From the Prisma Health/University of South Carolina School of Medicine, Columbia SC
| | - Abdelaziz Atwez
- From the Prisma Health/University of South Carolina School of Medicine, Columbia SC
| | - Martin Durkin
- From the Prisma Health/University of South Carolina School of Medicine, Columbia SC
| | - Mirsad M Mujadzic
- From the Prisma Health/University of South Carolina School of Medicine, Columbia SC
| |
Collapse
|
15
|
Alsaloum M, Estacion M, Almomani R, Gerrits MM, Bönhof GJ, Ziegler D, Malik R, Ferdousi M, Lauria G, Merkies IS, Faber CG, Dib-Hajj S, Waxman SG. A gain-of-function sodium channel β2-subunit mutation in painful diabetic neuropathy. Mol Pain 2020; 15:1744806919849802. [PMID: 31041876 PMCID: PMC6510061 DOI: 10.1177/1744806919849802] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Diabetes mellitus is a global challenge with many diverse health sequelae, of which diabetic peripheral neuropathy is one of the most common. A substantial number of patients with diabetic peripheral neuropathy develop chronic pain, but the genetic and epigenetic factors that predispose diabetic peripheral neuropathy patients to develop neuropathic pain are poorly understood. Recent targeted genetic studies have identified mutations in α-subunits of voltage-gated sodium channels (Navs) in patients with painful diabetic peripheral neuropathy. Mutations in proteins that regulate trafficking or functional properties of Navs could expand the spectrum of patients with Nav-related peripheral neuropathies. The auxiliary sodium channel β-subunits (β1–4) have been reported to increase current density, alter inactivation kinetics, and modulate subcellular localization of Nav. Mutations in β-subunits have been associated with several diseases, including epilepsy, cancer, and diseases of the cardiac conducting system. However, mutations in β-subunits have never been shown previously to contribute to neuropathic pain. We report here a patient with painful diabetic peripheral neuropathy and negative genetic screening for mutations in SCN9A, SCN10A, and SCN11A—genes encoding sodium channel α-subunit that have been previously linked to the development of neuropathic pain. Genetic analysis revealed an aspartic acid to asparagine mutation, D109N, in the β2-subunit. Functional analysis using current-clamp revealed that the β2-D109N rendered dorsal root ganglion neurons hyperexcitable, especially in response to repetitive stimulation. Underlying the hyperexcitability induced by the β2-subunit mutation, as evidenced by voltage-clamp analysis, we found a depolarizing shift in the voltage dependence of Nav1.7 fast inactivation and reduced use-dependent inhibition of the Nav1.7 channel.
Collapse
Affiliation(s)
- Matthew Alsaloum
- 1 Department of Neurology, Yale University School of Medicine, New Haven, CT, USA.,2 Center for Neuroscience and Regeneration Research, Veterans Affairs Medical Center, West Haven, CT, USA.,3 Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, CT, USA
| | - Mark Estacion
- 1 Department of Neurology, Yale University School of Medicine, New Haven, CT, USA.,2 Center for Neuroscience and Regeneration Research, Veterans Affairs Medical Center, West Haven, CT, USA
| | - Rowida Almomani
- 4 Department of Clinical Genomics, University Medical Center Maastricht, Maastricht, the Netherlands.,5 Department of Medical Laboratory Sciences, Jordan University of Science and Technology, Irbid, Jordan
| | - Monique M Gerrits
- 4 Department of Clinical Genomics, University Medical Center Maastricht, Maastricht, the Netherlands.,6 Department of Neurology, University Medical Centre Maastricht, Maastricht, the Netherlands
| | - Gidon J Bönhof
- 7 Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
| | - Dan Ziegler
- 7 Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany.,1 8German Center for Diabetes Research, München-Neuherberg, Germany.,9 Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Rayaz Malik
- 10 Weill Cornell Medicine-Qatar, Doha, Qatar.,11 Division of Diabetes, Endocrinology and Gastroenterology, Institute of Human Development, University of Manchester, Manchester, UK
| | - Maryam Ferdousi
- 11 Division of Diabetes, Endocrinology and Gastroenterology, Institute of Human Development, University of Manchester, Manchester, UK
| | - Giuseppe Lauria
- 12 Neuroalgology Unit, IRCCS Foundation "Carlo Besta" Neurological Institute, Milan, Italy.,13 Department of Biomedical and Clinical Sciences "Luigi Sacco," University of Milan, Milan, Italy
| | - Ingemar Sj Merkies
- 6 Department of Neurology, University Medical Centre Maastricht, Maastricht, the Netherlands.,14 Department of Neurology, St. Elisabeth Hospital, Willemstad, Curaçao
| | - Catharina G Faber
- 6 Department of Neurology, University Medical Centre Maastricht, Maastricht, the Netherlands
| | - Sulayman Dib-Hajj
- 1 Department of Neurology, Yale University School of Medicine, New Haven, CT, USA.,2 Center for Neuroscience and Regeneration Research, Veterans Affairs Medical Center, West Haven, CT, USA
| | - Stephen G Waxman
- 1 Department of Neurology, Yale University School of Medicine, New Haven, CT, USA.,2 Center for Neuroscience and Regeneration Research, Veterans Affairs Medical Center, West Haven, CT, USA
| | | |
Collapse
|
16
|
Tan YX, Hong Y, Jiang S, Lu MN, Li S, Chen B, Zhang L, Hu T, Mao R, Mei R, Xiyang YB. MicroRNA‑449a regulates the progression of brain aging by targeting SCN2B in SAMP8 mice. Int J Mol Med 2020; 45:1091-1102. [PMID: 32124967 PMCID: PMC7053848 DOI: 10.3892/ijmm.2020.4502] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 01/29/2020] [Indexed: 12/23/2022] Open
Abstract
Our previous study demonstrated that the expression of sodium channel voltage-gated beta 2 (SCN2B) increased with aging in senescence-accelerated mouse prone 8 (SAMP8) mice, and was identified to be associated with a decline in learning and memory, while the underlying mechanism is unclear. In the present study, multiple differentially expressed miRNAs, which may be involved in the process of aging by regulating target genes, were identified in the prefrontal cortex and hippocampus of SAMP8 mice though miRNA microarray analysis. Using bioinformatics prediction, SCN2B was identified to be one of the potential target genes of miR-449a, which was downregulated in the hippocampus. Previous studies demonstrated that miR-449a is involved in the occurrence and progression of aging by regulating a variety of target genes. Therefore, it was hypothesized that miR-449a may be involved in the process of brain aging by targeting SCN2B. To verify this hypothesis, the following experiments were conducted: A reverse transcription-quantitative polymerase chain reaction assay revealed that the expression level of miR-449a was significantly decreased in the prefrontal cortex and hippocampus of 12-month old SAMP8 mice; a dual-luciferase reporter assay verified that miR-449a regulated SCN2B expression by binding to the 3′-UTR 'seed region'; an anti-Ago co-immunoprecipitation combined with Affymetrix micro-array analyses demonstrated that the target mRNA highly enriched with Ago-miRNPs was confirmed to be SCN2B. Finally, overexpression of miR-449a or inhibition of SCN2B promoted the extension of hippocampal neurons in vitro. The results of the present study suggested that miR-449a was downregulated in the prefrontal cortex and hippocampus of SAMP8 mice and may regulate the process of brain aging by targeting SCN2B.
Collapse
Affiliation(s)
- Ya-Xin Tan
- Institute of Neuroscience, Basic Medical College, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Ying Hong
- Department of Laboratory Medicine, The Third People's Hospital of Yunnan Province, Kunming, Yunnan 650011, P.R. China
| | - Shui Jiang
- Department of Laboratory Medicine, The Third People's Hospital of Yunnan Province, Kunming, Yunnan 650011, P.R. China
| | - Min-Nan Lu
- Science and Technology Achievement Incubation Center, Kunming, Yunnan 650500, P.R. China
| | - Shan Li
- Institute of Neuroscience, Basic Medical College, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Bo Chen
- Science and Technology Achievement Incubation Center, Kunming, Yunnan 650500, P.R. China
| | - Li Zhang
- Editorial Department of Journal of Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Tao Hu
- Department of Laboratory Medicine, The Third People's Hospital of Yunnan Province, Kunming, Yunnan 650011, P.R. China
| | - Rui Mao
- School of Stomatology, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Rong Mei
- Department of Neurology, The First People's Hospital of Yunnan Province, Kunming, Yunnan 650032, P.R. China
| | - Yan-Bin Xiyang
- Institute of Neuroscience, Basic Medical College, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| |
Collapse
|
17
|
Brandenburger T, Johannsen L, Prassek V, Kuebart A, Raile J, Wohlfromm S, Köhrer K, Huhn R, Hollmann MW, Hermanns H. MiR-34a is differentially expressed in dorsal root ganglia in a rat model of chronic neuropathic pain. Neurosci Lett 2019; 708:134365. [DOI: 10.1016/j.neulet.2019.134365] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 06/24/2019] [Accepted: 07/02/2019] [Indexed: 11/25/2022]
|
18
|
Intrathecal Injection of miR-133b-3p or miR-143-3p Prevents the Development of Persistent Cold and Mechanical Allodynia Following a Peripheral Nerve Injury in Rats. Neuroscience 2018; 386:223-239. [PMID: 30018017 DOI: 10.1016/j.neuroscience.2018.06.040] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 06/01/2018] [Accepted: 06/24/2018] [Indexed: 12/24/2022]
Abstract
In DRG an increase in miR-133b-3p, miR-143-3p, and miR-1-3p correlates with the lack of development of neuropathic pain following a peripheral nerve injury. Using lentiviral (LV) vectors we found that a single injection of LV-miR-133b-3p or LV-miR-143-3p immediately after a peripheral nerve injury prevented the development of sustained mechanical and cold allodynia. Injection of LV-miR-133b-3p or LV-miR-143-3p by themselves or in combination, on day 3 post-injury produced a partial and transient reduction in mechanical allodynia and a sustained decrease in cold allodynia. Injection of LV-miR-1-3p has no effect. Co-injection of LV-miR-1a with miR-133b-3p or miR-143-3p on day 3 post-injury produced a sustained decrease in mechanical and cold allodynia. In DRG cultures, miR-133b-3p and miR-143-3p but not miR-1-3p, enhanced the depolarization-evoked cytoplasmic calcium increase. Using 3'UTR target clones containing a Gaussian luciferase reporter gene we found that with the 3'UTR-Scn2b, miR-133-3p and miR-143-3p reduced the expression while miR-1-3p enhanced the expression of the reporter gene. With the 3'UTR-TRPM8, miR-133-3p and miR-143-3p reduced the expression and miR-1-3p had no effect. With the 3'UTR-Piezo2, miR-133-3p increased the expression while miR-143-3p and miR-1-3p had no effect. LV-miR133b-3p, LV-miR-143-3p and LV-miR1a-3p reduced Scn2b-mRNA and Piezo2-mRNA. LV-miR133b-3p and LV-miR-143-3p reduced TRPM8-mRNA. LV-miR-133b-3p and LV-miR-143-3p prevent the development of chronic pain when injected immediately after the injury, but are only partially effective when injected at later times. LV-miR-1a-3p had no effect on pain, but complemented the actions of LV-miR-133b-3p or LV-miR-143-3p resulting in a sustained reversal of pain when co-injected 3 days following nerve injury.
Collapse
|
19
|
Salinas-Abarca AB, Velazquez-Lagunas I, Franco-Enzástiga Ú, Torres-López JE, Rocha-González HI, Granados-Soto V. ATF2, but not ATF3, participates in the maintenance of nerve injury-induced tactile allodynia and thermal hyperalgesia. Mol Pain 2018; 14:1744806918787427. [PMID: 29921170 PMCID: PMC6050803 DOI: 10.1177/1744806918787427] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Transcription factors are proteins that modulate the transcriptional rate of target genes in the nucleus in response to extracellular or cytoplasmic signals. Activating transcription factors 2 (ATF2) and 3 (ATF3) respond to environmental signals and maintain cellular homeostasis. There is evidence that inflammation and nerve injury modulate ATF2 and ATF3 expression. However, the function of these transcription factors in pain is unknown. The purpose of this study was to investigate the contribution of ATF2 and ATF3 to nerve injury-induced neuropathic pain. L5/6 spinal nerve ligation induced tactile allodynia and thermal hyperalgesia. Moreover, nerve damage enhanced ATF2 and ATF3 protein expression in injured L5/6 dorsal root ganglia and spinal cord but not in uninjured L4 dorsal root ganglia. Nerve damage also enhanced ATF2 immunoreactivity in dorsal root ganglia and spinal cord 7 to 21 days post-injury. Repeated intrathecal post-treatment with a small-interfering RNA targeted against ATF2 (ATF2 siRNA) or anti-ATF2 antibody partially reversed tactile allodynia and thermal hyperalgesia. In contrast, ATF3 siRNA or anti-ATF3 antibody did not modify nociceptive behaviors. ATF2 immunoreactivity was found in dorsal root ganglia and spinal cord co-labeling with NeuN mainly in non-peptidergic (IB4+) but also in peptidergic (CGRP+) neurons. ATF2 was found mainly in small- and medium-sized neurons. These results suggest that ATF2, but not ATF3, is found in strategic sites related to spinal nociceptive processing and participates in the maintenance of neuropathic pain in rats.
Collapse
Affiliation(s)
- Ana B Salinas-Abarca
- 1 Neurobiology of Pain Laboratory, Departamento de Farmacobiología, Cinvestav, Mexico
| | | | | | - Jorge E Torres-López
- 2 Laboratorio Mecanismos del Dolor, Centro de Investigación, División Académica de Ciencias de la Salud, Universidad Juárez Autónoma de Tabasco, Mexico.,3 Hospital Regional de Alta Especialidad Dr. Juan Graham Casasús, Mexico
| | - Héctor I Rocha-González
- 4 Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico
| | - Vinicio Granados-Soto
- 1 Neurobiology of Pain Laboratory, Departamento de Farmacobiología, Cinvestav, Mexico
| |
Collapse
|
20
|
Co-expression of β Subunits with the Voltage-Gated Sodium Channel Na V1.7: the Importance of Subunit Association and Phosphorylation and Their Effects on Channel Pharmacology and Biophysics. J Mol Neurosci 2018; 65:154-166. [PMID: 29744740 DOI: 10.1007/s12031-018-1082-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 05/01/2018] [Indexed: 12/19/2022]
Abstract
The voltage-gated sodium ion channel NaV1.7 is crucial in pain signaling. We examined how auxiliary β2 and β3 subunits and the phosphorylation state of the channel influence its biophysical properties and pharmacology. The human NaV1.7α subunit was co-expressed with either β2 or β3 subunits in HEK-293 cells. The β2 subunits and the NaV1.7α, however, were barely associated as evidenced by immunoprecipitation. Therefore, the β2 subunits did not change the biophysical properties of the channel. In contrast, β3 subunit was clearly associated with NaV1.7α. This subunit had a significant degree of glycosylation, and only the fully glycosylated β3 subunit was associated with the NaV1.7α. Electrophysiological characterisation revealed that the β3 subunit had small but consistent effects: a right-hand shift of the steady-state inactivation and faster recovery from inactivation. Furthermore, the β3 subunit reduced the susceptibility of NaV1.7α to several sodium channel blockers. In addition, we assessed the functional effect of NaV1.7α phosphorylation. Inhibition of kinase activity increased channel inactivation, while the blocking phosphatases produced the opposite effect. In conclusion, co-expression of β subunits with NaV1.7α, to better mimic the native channel properties, may be ineffective in cases when subunits are not associated, as shown in our experiments with β2. The β3 subunit significantly influences the function of NaV1.7α and, together with the phosphorylation of the channel, regulates its biophysical and pharmacological properties. These are important findings to take into account when considering the role of NaV1.7 channel in pain signaling.
Collapse
|
21
|
Palmisano M, Caputi FF, Mercatelli D, Romualdi P, Candeletti S. Dynorphinergic system alterations in the corticostriatal circuitry of neuropathic mice support its role in the negative affective component of pain. GENES BRAIN AND BEHAVIOR 2018; 18:e12467. [PMID: 29430855 PMCID: PMC7379183 DOI: 10.1111/gbb.12467] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 01/19/2018] [Accepted: 02/07/2018] [Indexed: 01/01/2023]
Abstract
The dynorphinergic system is involved in pain transmission at spinal level, where dynorphin exerts antinociceptive or pronociceptive effects, based on its opioid or non‐opioid actions. Surprisingly, little evidence is currently available concerning the supraspinal role of the dynorphinergic system in pain conditions. The present study aimed to investigate whether neuropathic pain is accompanied by prodynorphin (Pdyn) and κ‐opioid receptor (Oprk1) gene expression alterations in selected mouse brain areas. To this end, mice were subjected to chronic constriction injury of the right sciatic nerve and neuropathic pain behavioral signs were ascertained after 14 days. At this interval, a marked increase in Pdyn mRNA in the anterior cingulate cortex (ACC) and prefrontal cortex (PFC) was observed. Oprk1 gene expression was increased in the PFC, and decreased in the ACC and nucleus accumbens (NAc). No changes were observed in the other investigated regions. Because of the relationship between dynorphin and the brain‐derived neurotrophic factor, and the role of this neurotrophin in chronic pain‐related neuroplasticity, we investigated brain‐derived neurotrophic factor gene (Bdnf) expression in the areas showing Pdyn or Oprk1 mRNAs changes. Bdnf mRNA levels were increased in both the ACC and PFC, whereas no changes were assessed in the NAc. Present data indicate that the dynorphinergic system undergoes quite selective alterations involving the corticostriatal circuitry during neuropathic pain, suggesting a contribution to the negative affective component of pain. Moreover, parallel increases in Pdyn and Bdnf mRNA at cortical level suggest the occurrence of likely interactions between these systems in neuropathic pain maladaptive neuroplasticity.
Collapse
Affiliation(s)
- M Palmisano
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - F F Caputi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - D Mercatelli
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - P Romualdi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - S Candeletti
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| |
Collapse
|
22
|
Bang S, Yoo J, Gong X, Liu D, Han Q, Luo X, Chang W, Chen G, Im ST, Kim YH, Strong JA, Zhang MZ, Zhang JM, Lee SY, Ji RR. Differential Inhibition of Nav1.7 and Neuropathic Pain by Hybridoma-Produced and Recombinant Monoclonal Antibodies that Target Nav1.7 : Differential activities of Nav1.7-targeting monoclonal antibodies. Neurosci Bull 2018; 34:22-41. [PMID: 29333591 PMCID: PMC5799132 DOI: 10.1007/s12264-018-0203-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Accepted: 12/18/2017] [Indexed: 12/21/2022] Open
Abstract
The voltage-gated Na+ channel subtype Nav1.7 is important for pain and itch in rodents and humans. We previously showed that a Nav1.7-targeting monoclonal antibody (SVmab) reduces Na+ currents and pain and itch responses in mice. Here, we investigated whether recombinant SVmab (rSVmab) binds to and blocks Nav1.7 similar to SVmab. ELISA tests revealed that SVmab was capable of binding to Nav1.7-expressing HEK293 cells, mouse DRG neurons, human nerve tissue, and the voltage-sensor domain II of Nav1.7. In contrast, rSVmab showed no or weak binding to Nav1.7 in these tests. Patch-clamp recordings showed that SVmab, but not rSVmab, markedly inhibited Na+ currents in Nav1.7-expressing HEK293 cells. Notably, electrical field stimulation increased the blocking activity of SVmab and rSVmab in Nav1.7-expressing HEK293 cells. SVmab was more effective than rSVmab in inhibiting paclitaxel-induced mechanical allodynia. SVmab also bound to human DRG neurons and inhibited their Na+ currents. Finally, potential reasons for the differential efficacy of SVmab and rSVmab and future directions are discussed.
Collapse
Affiliation(s)
- Sangsu Bang
- Department of Anesthesiology, Duke University Medical Center, 595 LaSalle St, Durham, NC, 27710, USA
| | - Jiho Yoo
- Department of Biochemistry, Duke University Medical Center, 303 Research Drive, Durham, NC, 27710, USA
| | - Xingrui Gong
- Pain Research Center, Department of Anesthesiology, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH, 45267-0531, USA
- Department of Anesthesiology, Shanghai Children's Medical Center, Shanghai, 200127, China
| | - Di Liu
- Department of Anesthesiology, Duke University Medical Center, 595 LaSalle St, Durham, NC, 27710, USA
| | - Qingjian Han
- Department of Anesthesiology, Duke University Medical Center, 595 LaSalle St, Durham, NC, 27710, USA
| | - Xin Luo
- Department of Anesthesiology, Duke University Medical Center, 595 LaSalle St, Durham, NC, 27710, USA
| | - Wonseok Chang
- Department of Anesthesiology, Duke University Medical Center, 595 LaSalle St, Durham, NC, 27710, USA
- Department of Physiology and Biophysics, College of Medicine, Eulji University, 143-5 Yongdu-Dong, Jung-Gu, Daejeon, 34824, Korea
| | - Gang Chen
- Department of Anesthesiology, Duke University Medical Center, 595 LaSalle St, Durham, NC, 27710, USA
| | - Sang-Taek Im
- Department of Physiology, College of Medicine, Gachon University, Incheon, 21999, Korea
| | - Yong Ho Kim
- Department of Anesthesiology, Duke University Medical Center, 595 LaSalle St, Durham, NC, 27710, USA
- Department of Physiology, College of Medicine, Gachon University, Incheon, 21999, Korea
| | - Judith A Strong
- Pain Research Center, Department of Anesthesiology, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH, 45267-0531, USA
| | - Ma-Zhong Zhang
- Department of Anesthesiology, Shanghai Children's Medical Center, Shanghai, 200127, China
| | - Jun-Ming Zhang
- Pain Research Center, Department of Anesthesiology, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH, 45267-0531, USA.
| | - Seok-Yong Lee
- Department of Biochemistry, Duke University Medical Center, 303 Research Drive, Durham, NC, 27710, USA.
| | - Ru-Rong Ji
- Department of Anesthesiology, Duke University Medical Center, 595 LaSalle St, Durham, NC, 27710, USA.
| |
Collapse
|
23
|
Abstract
Voltage-gated sodium channels are protein complexes comprised of one pore forming α subunit and two, non-pore forming, β subunits. The voltage-gated sodium channel β subunits were originally identified to function as auxiliary subunits, which modulate the gating, kinetics, and localization of the ion channel pore. Since that time, the five β subunits have been shown to play crucial roles as multifunctional signaling molecules involved in cell adhesion, cell migration, neuronal pathfinding, fasciculation, and neurite outgrowth. Here, we provide an overview of the evidence implicating the β subunits in their conducting and non-conducting roles. Mutations in the β subunit genes (SCN1B-SCN4B) have been linked to a variety of diseases. These include cancer, epilepsy, cardiac arrhythmias, sudden infant death syndrome/sudden unexpected death in epilepsy, neuropathic pain, and multiple neurodegenerative disorders. β subunits thus provide novel therapeutic targets for future drug discovery.
Collapse
Affiliation(s)
- Alexandra A Bouza
- Department of Pharmacology, University of Michigan Medical School, 2200 MSRBIII, 1150 W. Medical Center Dr., Ann Arbor, MI, 48109-5632, USA
| | - Lori L Isom
- Department of Pharmacology, University of Michigan Medical School, 2301 MSRB III, 1150 W. Medical Center Dr., Ann Arbor, MI, 48109-5632, USA.
| |
Collapse
|
24
|
Hull JM, Isom LL. Voltage-gated sodium channel β subunits: The power outside the pore in brain development and disease. Neuropharmacology 2017; 132:43-57. [PMID: 28927993 DOI: 10.1016/j.neuropharm.2017.09.018] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 07/19/2017] [Accepted: 09/11/2017] [Indexed: 12/19/2022]
Abstract
Voltage gated sodium channels (VGSCs) were first identified in terms of their role in the upstroke of the action potential. The underlying proteins were later identified as saxitoxin and scorpion toxin receptors consisting of α and β subunits. We now know that VGSCs are heterotrimeric complexes consisting of a single pore forming α subunit joined by two β subunits; a noncovalently linked β1 or β3 and a covalently linked β2 or β4 subunit. VGSC α subunits contain all the machinery necessary for channel cell surface expression, ion conduction, voltage sensing, gating, and inactivation, in one central, polytopic, transmembrane protein. VGSC β subunits are more than simple accessories to α subunits. In the more than two decades since the original cloning of β1, our knowledge of their roles in physiology and pathophysiology has expanded immensely. VGSC β subunits are multifunctional. They confer unique gating mechanisms, regulate cellular excitability, affect brain development, confer distinct channel pharmacology, and have functions that are independent of the α subunits. The vast array of functions of these proteins stems from their special station in the channelome: being the only known constituents that are cell adhesion and intra/extracellular signaling molecules in addition to being part of channel complexes. This functional trifecta and how it goes awry demonstrates the power outside the pore in ion channel signaling complexes, broadening the term channelopathy beyond defects in ion conduction. This article is part of the Special Issue entitled 'Channelopathies.'
Collapse
Affiliation(s)
- Jacob M Hull
- Neuroscience Program and Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI 48109, United States
| | - Lori L Isom
- Neuroscience Program and Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI 48109, United States.
| |
Collapse
|
25
|
Berta T, Perrin FE, Pertin M, Tonello R, Liu YC, Chamessian A, Kato AC, Ji RR, Decosterd I. Gene Expression Profiling of Cutaneous Injured and Non-Injured Nociceptors in SNI Animal Model of Neuropathic Pain. Sci Rep 2017; 7:9367. [PMID: 28839165 PMCID: PMC5570923 DOI: 10.1038/s41598-017-08865-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 07/17/2017] [Indexed: 12/12/2022] Open
Abstract
Nociceptors are a particular subtype of dorsal root ganglion (DRG) neurons that detect noxious stimuli and elicit pain. Although recent efforts have been made to reveal the molecular profile of nociceptors in normal conditions, little is known about how this profile changes in pathological conditions. In this study we exploited laser capture microdissection to specifically collect individual injured and non-injured nociceptive DRG neurons and to define their gene profiling in rat spared nerve injury (SNI) model of neuropathic pain. We found minimal transcriptional changes in non-injured neurons at 7 days after SNI. In contrast, several novel transcripts were altered in injured nociceptors, and the global signature of these LCM-captured neurons differed markedly from that the gene expression patterns found previously using whole DRG tissue following SNI. Pathway analysis of the transcriptomic profile of the injured nociceptors revealed oxidative stress as a key biological process. We validated the increase of caspase-6 (CASP6) in small-sized DRG neurons and its functional role in SNI- and paclitaxel-induced neuropathic pain. Our results demonstrate that the identification of gene regulation in a specific population of DRG neurons (e.g., nociceptors) is an effective strategy to reveal new mechanisms and therapeutic targets for neuropathic pain from different origins.
Collapse
Affiliation(s)
- Temugin Berta
- Pain Center, Department of anesthesiology, Lausanne University Hospital (CHUV) and Faculty of biology and medicine (FBM), University of Lausanne (UNIL), 1011, Lausanne, Switzerland.
- Department of Anesthesiology, Duke University Medical Center, 595 LaSalle Street, Durham, NC, 27710, USA.
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, Ohio, 45267, USA.
| | - Florence E Perrin
- Department of Basic Neuroscience, Faculty of Medicine, 1211, Geneva 4, Geneva, Switzerland
- University of Montpellier, Montpellier, F-34095 France, INSERM, U1198, Montpellier, F-34095 France, EPHE, Paris, F-75014, France
| | - Marie Pertin
- Pain Center, Department of anesthesiology, Lausanne University Hospital (CHUV) and Faculty of biology and medicine (FBM), University of Lausanne (UNIL), 1011, Lausanne, Switzerland
- Department of Fundamental Neurosciences, Faculty of biology and medicine (FBM), University of Lausanne (UNIL), 1005, Lausanne, Switzerland
| | - Raquel Tonello
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, Ohio, 45267, USA
| | - Yen-Chin Liu
- Department of Anesthesiology, College of Medicine, National Cheng Kung University, Tainan city, Taiwan
| | - Alexander Chamessian
- Department of Anesthesiology, Duke University Medical Center, 595 LaSalle Street, Durham, NC, 27710, USA
| | - Ann C Kato
- Department of Basic Neuroscience, Faculty of Medicine, 1211, Geneva 4, Geneva, Switzerland
| | - Ru-Rong Ji
- Department of Anesthesiology, Duke University Medical Center, 595 LaSalle Street, Durham, NC, 27710, USA
| | - Isabelle Decosterd
- Pain Center, Department of anesthesiology, Lausanne University Hospital (CHUV) and Faculty of biology and medicine (FBM), University of Lausanne (UNIL), 1011, Lausanne, Switzerland
- Department of Fundamental Neurosciences, Faculty of biology and medicine (FBM), University of Lausanne (UNIL), 1005, Lausanne, Switzerland
| |
Collapse
|
26
|
Bulleyaconitine A preferably reduces tetrodotoxin-sensitive sodium current in uninjured dorsal root ganglion neurons of neuropathic rats probably via inhibition of protein kinase C. Pain 2017; 158:2169-2180. [DOI: 10.1097/j.pain.0000000000001018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
27
|
Ito N, Sakai A, Miyake N, Maruyama M, Iwasaki H, Miyake K, Okada T, Sakamoto A, Suzuki H. miR-15b mediates oxaliplatin-induced chronic neuropathic pain through BACE1 down-regulation. Br J Pharmacol 2017; 174:386-395. [PMID: 28012171 DOI: 10.1111/bph.13698] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 12/11/2016] [Accepted: 12/16/2016] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND AND PURPOSE Although oxaliplatin is an effective anti-cancer platinum compound, it can cause painful chronic neuropathy, and its molecular mechanisms are poorly understood. MicroRNAs (miRNAs) are small non-coding RNAs that negatively regulate gene expression in a sequence-specific manner. Although miRNAs have been increasingly recognized as important modulators in a variety of pain conditions, their involvement in chemotherapy-induced neuropathic pain is unknown. EXPERIMENTAL APPROACH Oxaliplatin-induced chronic neuropathic pain was induced in rats by i.p. injections of oxaliplatin (2 mg·kg-1 ) for five consecutive days. The expression levels of miR-15b and β-site amyloid precursor protein-cleaving enzyme 1 (BACE1 also known as β-secretase 1) were examined in the dorsal root ganglion (DRG). To examine the function of miR-15b, an adeno-associated viral vector encoding miR-15b was injected into the DRG in vivo. KEY RESULTS Among the miRNAs examined in the DRG in the late phase of oxaliplatin-induced neuropathic pain, miR-15b was most robustly increased. Our in vitro assay results determined that BACE1 was a target of miR-15b. BACE1 and miR-15b were co-expressed in putative myelinated and unmyelinated DRG neurons. Overexpression of miR-15b in DRG neurons caused mechanical allodynia in association with reduced expression of BACE1. Consistent with these results, a BACE1 inhibitor dose-dependently induced significant mechanical allodynia. CONCLUSIONS AND IMPLICATIONS These findings suggest that miR-15b contributes to oxaliplatin-induced chronic neuropathic pain at least in part through the down-regulation of BACE1.
Collapse
Affiliation(s)
- Naomi Ito
- Department of Pharmacology, Nippon Medical School, Tokyo, Japan.,Department of Anesthesiology, Nippon Medical School, Tokyo, Japan
| | - Atsushi Sakai
- Department of Pharmacology, Nippon Medical School, Tokyo, Japan
| | - Noriko Miyake
- Department of Biochemistry and Molecular Biology, Nippon Medical School, Tokyo, Japan
| | - Motoyo Maruyama
- Department of Pharmacology, Nippon Medical School, Tokyo, Japan.,Division of Laboratory Animal Science, Nippon Medical School, Tokyo, Japan
| | - Hirotoshi Iwasaki
- Department of Pharmacology, Nippon Medical School, Tokyo, Japan.,Department of Anesthesiology, Nippon Medical School, Tokyo, Japan
| | - Koichi Miyake
- Department of Biochemistry and Molecular Biology, Nippon Medical School, Tokyo, Japan
| | - Takashi Okada
- Department of Biochemistry and Molecular Biology, Nippon Medical School, Tokyo, Japan
| | | | - Hidenori Suzuki
- Department of Pharmacology, Nippon Medical School, Tokyo, Japan
| |
Collapse
|
28
|
Masocha W. Gene expression profile of sodium channel subunits in the anterior cingulate cortex during experimental paclitaxel-induced neuropathic pain in mice. PeerJ 2016; 4:e2702. [PMID: 27896032 PMCID: PMC5119229 DOI: 10.7717/peerj.2702] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 10/19/2016] [Indexed: 12/19/2022] Open
Abstract
Paclitaxel, a chemotherapeutic agent, causes neuropathic pain whose supraspinal pathophysiology is not fully understood. Dysregulation of sodium channel expression, studied mainly in the periphery and spinal cord level, contributes to the pathogenesis of neuropathic pain. We examined gene expression of sodium channel (Nav) subunits by real time polymerase chain reaction (PCR) in the anterior cingulate cortex (ACC) at day 7 post first administration of paclitaxel, when mice had developed paclitaxel-induced thermal hyperalgesia. The ACC was chosen because increased activity in the ACC has been observed during neuropathic pain. In the ACC of vehicle-treated animals the threshold cycle (Ct) values for Nav1.4, Nav1.5, Nav1.7, Nav1.8 and Nav1.9 were above 30 and/or not detectable in some samples. Thus, comparison in mRNA expression between untreated control, vehicle-treated and paclitaxel treated animals was done for Nav1.1, Nav1.2, Nav1.3, Nav1.6, Nax as well as Navβ1–Navβ4. There were no differences in the transcript levels of Nav1.1–Nav1.3, Nav1.6, Nax, Navβ1–Navβ3 between untreated and vehicle-treated mice, however, vehicle treatment increased Navβ4 expression. Paclitaxel treatment significantly increased the mRNA expression of Nav1.1, Nav1.2, Nav1.6 and Nax, but not Nav1.3, sodium channel alpha subunits compared to vehicle-treated animals. Treatment with paclitaxel significantly increased the expression of Navβ1 and Navβ3, but not Navβ2 and Navβ4, sodium channel beta subunits compared to vehicle-treated animals. These findings suggest that during paclitaxel-induced neuropathic pain (PINP) there is differential upregulation of sodium channels in the ACC, which might contribute to the increased neuronal activity observed in the area during neuropathic pain.
Collapse
Affiliation(s)
- Willias Masocha
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Kuwait University , Safat , Kuwait
| |
Collapse
|
29
|
Shimada Y, Sato T, Yajima T, Fujita M, Hashimoto N, Shoji N, Sasano T, Ichikawa H. SCN2B in the Rat Trigeminal Ganglion and Trigeminal Sensory Nuclei. Cell Mol Neurobiol 2016; 36:1399-1408. [PMID: 26852328 DOI: 10.1007/s10571-016-0340-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 01/28/2016] [Indexed: 12/21/2022]
Abstract
The beta-2 subunit of the mammalian brain voltage-gated sodium channel (SCN2B) was examined in the rat trigeminal ganglion (TG) and trigeminal sensory nuclei. In the TG, 42.6 % of sensory neurons were immunoreactive (IR) for SCN2B. These neurons had various cell body sizes. In facial skins and oral mucosae, corpuscular nerve endings contained SCN2B-immunoreactivity. SCN2B-IR nerve fibers formed nerve plexuses beneath taste buds in the tongue and incisive papilla. However, SCN2B-IR free nerve endings were rare in cutaneous and mucosal epithelia. Tooth pulps, muscle spindles and major salivary glands were also innervated by SCN2B-IR nerve fibers. A double immunofluorescence method revealed that about 40 % of SCN2B-IR neurons exhibited calcitonin gene-related peptide (CGRP)-immunoreactivity. However, distributions of SCN2B- and CGRP-IR nerve fibers were mostly different in facial, oral and cranial structures. By retrograde tracing method, 60.4 and 85.3 % of TG neurons innervating the facial skin and tooth pulp, respectively, showed SCN2B-immunoreactivity. CGRP-immunoreactivity was co-localized by about 40 % of SCN2B-IR cutaneous and tooth pulp TG neurons. In trigeminal sensory nuclei of the brainstem, SCN2B-IR neuronal cell bodies were common in deep laminae of the subnucleus caudalis, and the subnuclei interpolaris and oralis. In the mesencephalic trigeminal tract nucleus, primary sensory neurons also exhibited SCN2B-immunoreactivity. In other regions of trigeminal sensory nuclei, SCN2B-IR cells were very infrequent. SCN2B-IR neuropil was detected in deep laminae of the subnucleus caudalis as well as in the subnuclei interpolaris, oralis and principalis. These findings suggest that SCN2B is expressed by various types of sensory neurons in the TG. There appears to be SCN2B-containing pathway in the TG and trigeminal sensory nuclei.
Collapse
Affiliation(s)
- Yusuke Shimada
- Division of Oral Diagnosis, Graduate School of Dentistry, Tohoku University, Sendai, 980-8575, Japan
| | - Tadasu Sato
- Division of Oral and Craniofacial Anatomy, Graduate School of Dentistry, Tohoku University, 4-1 Seiryo-machi, Sendai, 980-8575, Japan
| | - Takehiro Yajima
- Division of Operative Dentistry, Graduate School of Dentistry, Tohoku University, Sendai, 980-8575, Japan
| | - Masatoshi Fujita
- Division of Dental Anesthesiology and Pain Management, Graduate School of Dentistry, Tohoku University, Sendai, 980-8575, Japan
| | - Naoya Hashimoto
- Division of Oral Diagnosis, Graduate School of Dentistry, Tohoku University, Sendai, 980-8575, Japan
| | - Noriaki Shoji
- Division of Oral Diagnosis, Graduate School of Dentistry, Tohoku University, Sendai, 980-8575, Japan
| | - Takashi Sasano
- Division of Oral Diagnosis, Graduate School of Dentistry, Tohoku University, Sendai, 980-8575, Japan
| | - Hiroyuki Ichikawa
- Division of Oral and Craniofacial Anatomy, Graduate School of Dentistry, Tohoku University, 4-1 Seiryo-machi, Sendai, 980-8575, Japan.
| |
Collapse
|
30
|
Munasinghe NR, Christie MJ. Conotoxins That Could Provide Analgesia through Voltage Gated Sodium Channel Inhibition. Toxins (Basel) 2015; 7:5386-407. [PMID: 26690478 PMCID: PMC4690140 DOI: 10.3390/toxins7124890] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 10/23/2015] [Accepted: 11/19/2015] [Indexed: 12/19/2022] Open
Abstract
Chronic pain creates a large socio-economic burden around the world. It is physically and mentally debilitating, and many sufferers are unresponsive to current therapeutics. Many drugs that provide pain relief have adverse side effects and addiction liabilities. Therefore, a great need has risen for alternative treatment strategies. One rich source of potential analgesic compounds that has emerged over the past few decades are conotoxins. These toxins are extremely diverse and display selective activity at ion channels. Voltage gated sodium (NaV) channels are one such group of ion channels that play a significant role in multiple pain pathways. This review will explore the literature around conotoxins that bind NaV channels and determine their analgesic potential.
Collapse
Affiliation(s)
- Nehan R Munasinghe
- Discipline of Pharmacology, The University of Sydney, Sydney, NSW 2006, Australia.
| | - MacDonald J Christie
- Discipline of Pharmacology, The University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
31
|
Abstract
Voltage-gated sodium channels (VGSCs) are responsible for the initiation and propagation of action potentials in excitable cells. VGSCs in mammalian brain are heterotrimeric complexes of α and β subunits. Although β subunits were originally termed auxiliary, we now know that they are multifunctional signaling molecules that play roles in both excitable and nonexcitable cell types and with or without the pore-forming α subunit present. β subunits function in VGSC and potassium channel modulation, cell adhesion, and gene regulation, with particularly important roles in brain development. Mutations in the genes encoding β subunits are linked to a number of diseases, including epilepsy, sudden death syndromes like SUDEP and SIDS, and cardiac arrhythmia. Although VGSC β subunit-specific drugs have not yet been developed, this protein family is an emerging therapeutic target.
Collapse
Affiliation(s)
- Heather A O'Malley
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan 48109;
| | | |
Collapse
|
32
|
Laedermann CJ, Abriel H, Decosterd I. Post-translational modifications of voltage-gated sodium channels in chronic pain syndromes. Front Pharmacol 2015; 6:263. [PMID: 26594175 PMCID: PMC4633509 DOI: 10.3389/fphar.2015.00263] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 10/23/2015] [Indexed: 02/06/2023] Open
Abstract
In the peripheral sensory nervous system the neuronal expression of voltage-gated sodium channels (Navs) is very important for the transmission of nociceptive information since they give rise to the upstroke of the action potential (AP). Navs are composed of nine different isoforms with distinct biophysical properties. Studying the mutations associated with the increase or absence of pain sensitivity in humans, as well as other expression studies, have highlighted Nav1.7, Nav1.8, and Nav1.9 as being the most important contributors to the control of nociceptive neuronal electrogenesis. Modulating their expression and/or function can impact the shape of the AP and consequently modify nociceptive transmission, a process that is observed in persistent pain conditions. Post-translational modification (PTM) of Navs is a well-known process that modifies their expression and function. In chronic pain syndromes, the release of inflammatory molecules into the direct environment of dorsal root ganglia (DRG) sensory neurons leads to an abnormal activation of enzymes that induce Navs PTM. The addition of small molecules, i.e., peptides, phosphoryl groups, ubiquitin moieties and/or carbohydrates, can modify the function of Navs in two different ways: via direct physical interference with Nav gating, or via the control of Nav trafficking. Both mechanisms have a profound impact on neuronal excitability. In this review we will discuss the role of Protein Kinase A, B, and C, Mitogen Activated Protein Kinases and Ca++/Calmodulin-dependent Kinase II in peripheral chronic pain syndromes. We will also discuss more recent findings that the ubiquitination of Nav1.7 by Nedd4-2 and the effect of methylglyoxal on Nav1.8 are also implicated in the development of experimental neuropathic pain. We will address the potential roles of other PTMs in chronic pain and highlight the need for further investigation of PTMs of Navs in order to develop new pharmacological tools to alleviate pain.
Collapse
Affiliation(s)
- Cedric J. Laedermann
- F.M. Kirby Neurobiology Research Center, Boston Children’s Hospital, Harvard Medical School, BostonMA, USA
| | - Hugues Abriel
- Department of Clinical Research, University of BernBern, Switzerland
| | - Isabelle Decosterd
- Pain Center, Department of Anesthesiology, Lausanne University Hospital (CHUV) and University of LausanneLausanne, Switzerland
- Department of Fundamental Neurosciences, University of LausanneLausanne, Switzerland
| |
Collapse
|
33
|
Xie W, Strong JA, Zhang JM. Local knockdown of the NaV1.6 sodium channel reduces pain behaviors, sensory neuron excitability, and sympathetic sprouting in rat models of neuropathic pain. Neuroscience 2015; 291:317-30. [PMID: 25686526 DOI: 10.1016/j.neuroscience.2015.02.010] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 01/21/2015] [Accepted: 02/05/2015] [Indexed: 11/15/2022]
Abstract
In the spinal nerve ligation (SNL) model of neuropathic pain, as in other pain models, abnormal spontaneous activity of myelinated sensory neurons occurs early and is essential for establishing pain behaviors and other pathologies. Sympathetic sprouting into the dorsal root ganglion (DRG) is observed after SNL, and sympathectomy reduces pain behavior. Sprouting and spontaneous activity may be mutually reinforcing: blocking neuronal activity reduces sympathetic sprouting, and sympathetic spouts functionally increase spontaneous activity in vitro. However, most studies in this field have used nonspecific methods to block spontaneous activity, methods that also block evoked and normal activity. In this study, we injected small inhibitory (si) RNA directed against the NaV1.6 sodium channel isoform into the DRG before SNL. This isoform can mediate high-frequency repetitive firing, like that seen in spontaneously active neurons. Local knockdown of NaV1.6 markedly reduced mechanical pain behaviors induced by SNL, reduced sympathetic sprouting into the ligated sensory ganglion, and blocked abnormal spontaneous activity and other measures of hyperexcitability in myelinated neurons in the ligated sensory ganglion. Immunohistochemical experiments showed that sympathetic sprouting preferentially targeted NaV1.6-positive neurons. Under these experimental conditions, NaV1.6 knockdown did not prevent or strongly alter single evoked action potentials, unlike previous less specific methods used to block spontaneous activity. NaV1.6 knockdown also reduced pain behaviors in another pain model, chronic constriction of the sciatic nerve, provided the model was modified so that the lesion site was relatively close to the siRNA-injected lumbar DRGs. The results highlight the relative importance of abnormal spontaneous activity in establishing both pain behaviors and sympathetic sprouting, and suggest that the NaV1.6 isoform may have value as a therapeutic target.
Collapse
Affiliation(s)
- W Xie
- Pain Research Center, Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0531, USA.
| | - J A Strong
- Pain Research Center, Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0531, USA.
| | - J-M Zhang
- Pain Research Center, Department of Anesthesiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0531, USA.
| |
Collapse
|
34
|
DNA Microarray Analysis on the Genes Differentially Expressed in the Liver of the Pufferfish, Takifugu rubripes, Following an Intramuscular Administration of Tetrodotoxin. MICROARRAYS 2014; 3:226-44. [PMID: 27600346 PMCID: PMC4979056 DOI: 10.3390/microarrays3040226] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 09/28/2014] [Accepted: 10/15/2014] [Indexed: 01/13/2023]
Abstract
Pufferfish accumulate tetrodotoxin (TTX) mainly in the liver and ovary. This study aims at investigating the effect of TTX accumulation in the liver of cultured specimens of torafugu Takifugu rubripes on the hepatic gene expression by microarray analysis on Day 5 after the intramuscular administration of 0.25 mg TTX/kg body weight into the caudal muscle. TTX was detected in the liver, skin and ovary in the TTX-administered individuals. The total amount of TTX accumulated in the body was 67 ± 8% of the administered dose on Day 5. Compared with the buffer-administered control group, a total of 59 genes were significantly upregulated more than two-fold in the TTX-administered group, including those encoding chymotrypsin-like elastase family member 2A, transmembrane protein 168 and Rho GTP-activating protein 29. In contrast, a total of 427 genes were downregulated by TTX administration, including those encoding elongation factor G2, R-spondin-3, nuclear receptor activator 2 and fatty acyl-CoA hydrolase precursor. In conclusion, our results demonstrate that the intramuscular administration of TTX changes the expression of hepatic genes involved in various signaling pathways.
Collapse
|
35
|
Gould HJ, Soignier RD, Cho SR, Hernandez C, Diamond I, Taylor BK, Paul D. Ranolazine Attenuates Mechanical Allodynia Associated with Demyelination Injury. PAIN MEDICINE 2014; 15:1771-80. [DOI: 10.1111/pme.12516] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
36
|
Norcini M, Sideris A, Martin Hernandez LA, Zhang J, Blanck TJJ, Recio-Pinto E. An approach to identify microRNAs involved in neuropathic pain following a peripheral nerve injury. Front Neurosci 2014; 8:266. [PMID: 25221468 PMCID: PMC4148822 DOI: 10.3389/fnins.2014.00266] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 08/06/2014] [Indexed: 11/29/2022] Open
Abstract
Peripheral nerve injury alters the expression of hundreds of proteins in dorsal root ganglia (DRG). Targeting some of these proteins has led to successful treatments for acute pain, but not for sustained post-operative neuropathic pain. The latter may require targeting multiple proteins. Since a single microRNA (miR) can affect the expression of multiple proteins, here, we describe an approach to identify chronic neuropathic pain-relevant miRs. We used two variants of the spared nerve injury (SNI): Sural-SNI and Tibial-SNI and found distinct pain phenotypes between the two. Both models induced strong mechanical allodynia, but only Sural-SNI rats maintained strong mechanical and cold allodynia, as previously reported. In contrast, we found that Tibial-SNI rats recovered from mechanical allodynia and never developed cold allodynia. Since both models involve nerve injury, we increased the probability of identifying differentially regulated miRs that correlated with the quality and magnitude of neuropathic pain and decreased the probability of detecting miRs that are solely involved in neuronal regeneration. We found seven such miRs in L3-L5 DRG. The expression of these miRs increased in Tibial-SNI. These miRs displayed a lower level of expression in Sural-SNI, with four having levels lower than those in sham animals. Bioinformatic analysis of how these miRs could affect the expression of some ion channels supports the view that, following a peripheral nerve injury, the increase of the seven miRs may contribute to the recovery from neuropathic pain while the decrease of four of them may contribute to the development of chronic neuropathic pain. The approach used resulted in the identification of a small number of potentially neuropathic pain relevant miRs. Additional studies are required to investigate whether manipulating the expression of the identified miRs in primary sensory neurons can prevent or ameliorate chronic neuropathic pain following peripheral nerve injuries.
Collapse
Affiliation(s)
- Monica Norcini
- Department of Anesthesiology, NYU Langone Medical Center New York, NY, USA
| | - Alexandra Sideris
- Department of Anesthesiology, NYU Langone Medical Center New York, NY, USA
| | | | - Jin Zhang
- Department of Anesthesiology, NYU Langone Medical Center New York, NY, USA
| | - Thomas J J Blanck
- Department of Anesthesiology, NYU Langone Medical Center New York, NY, USA ; Department of Neuroscience and Physiology, NYU Langone Medical Center New York, NY, USA
| | - Esperanza Recio-Pinto
- Department of Anesthesiology, NYU Langone Medical Center New York, NY, USA ; Department of Biochemistry and Molecular Pharmacology, NYU Langone Medical Center New York, NY, USA
| |
Collapse
|
37
|
Raju HB, Englander Z, Capobianco E, Tsinoremas NF, Lerch JK. Identification of potential therapeutic targets in a model of neuropathic pain. Front Genet 2014; 5:131. [PMID: 24904634 PMCID: PMC4033210 DOI: 10.3389/fgene.2014.00131] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 04/24/2014] [Indexed: 01/18/2023] Open
Abstract
Neuropathic pain (NP) is caused by damage to the nervous system, resulting in dysfunction and aberrant pain. The cellular functions (e.g., peripheral neuron spinal cord innervation, neuronal excitability) associated with NP often develop over time and are likely associated with gene expression changes. Gene expression studies on the cells involved in NP (e.g., sensory dorsal root ganglion neurons) are publically available; the mining of these studies may enable the identification of novel targets and the subsequent development of therapies that are essential for improving quality of life for the millions of individuals suffering with NP. Here we analyzed a publically available microarray dataset (GSE30165) in order to identify new RNAs (e.g., messenger RNA (mRNA) isoforms and non-coding RNAs) underlying NP. GSE30165 profiled gene expression in dorsal root ganglion neurons (DRG) and in sciatic nerve (SN) after resection, a NP model. Gene ontological analysis shows enrichment for sensory and neuronal processes. Protein network analysis demonstrates DRG upregulated genes typical to an injury and NP response. Of the top changing genes, 34 and 36% are associated with more than one protein coding isoform in the DRG and SN, respectively. The majority of genes are receptor and enzymes. We identified 15 long non-coding RNAs (lncRNAs) targeting these genes in LNCipedia.org, an online comprehensive lncRNA database. These RNAs represent new therapeutic targets for preventing NP development and this approach demonstrates the feasibility of data reanalysis for their identification.
Collapse
Affiliation(s)
- Hemalatha B Raju
- Center for Computational Science, Department of Medicine, University of Miami Miller School of Medicine Miami, FL, USA ; Human Genetics and Genomics Graduate Program, University of Miami Miller School of Medicine Miami, FL, USA
| | - Zoe Englander
- Department of Biomedical Engineering, Duke University Durham, NC, USA
| | - Enrico Capobianco
- Center for Computational Science, Department of Medicine, University of Miami Miller School of Medicine Miami, FL, USA ; Laboratory of Integrative Systems Medicine, National Research Council (CNR) Pisa, Italy
| | - Nicholas F Tsinoremas
- Center for Computational Science, Department of Medicine, University of Miami Miller School of Medicine Miami, FL, USA
| | - Jessica K Lerch
- Department of Neuroscience, Center for Brain and Spinal Cord Repair, The Ohio State University Columbus, OH, USA
| |
Collapse
|
38
|
Laedermann CJ, Pertin M, Suter MR, Decosterd I. Voltage-gated sodium channel expression in mouse DRG after SNI leads to re-evaluation of projections of injured fibers. Mol Pain 2014; 10:19. [PMID: 24618114 PMCID: PMC4007621 DOI: 10.1186/1744-8069-10-19] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 02/28/2014] [Indexed: 12/19/2022] Open
Abstract
Background Dysregulation of voltage-gated sodium channels (Navs) is believed to play a major role in nerve fiber hyperexcitability associated with neuropathic pain. A complete transcriptional characterization of the different isoforms of Navs under normal and pathological conditions had never been performed on mice, despite their widespread use in pain research. Navs mRNA levels in mouse dorsal root ganglia (DRG) were studied in the spared nerve injury (SNI) and spinal nerve ligation (SNL) models of neuropathic pain. In the SNI model, injured and non-injured neurons were intermingled in lumbar DRG, which were pooled to increase the tissue available for experiments. Results A strong downregulation was observed for every Navs isoform expressed except for Nav1.2; even Nav1.3, known to be upregulated in rat neuropathic pain models, was lower in the SNI mouse model. This suggests differences between these two species. In the SNL model, where the cell bodies of injured and non-injured fibers are anatomically separated between different DRG, most Navs were observed to be downregulated in the L5 DRG receiving axotomized fibers. Transcription was then investigated independently in the L3, L4 and L5 DRG in the SNI model, and an important downregulation of many Navs isoforms was observed in the L3 DRG, suggesting the presence of numerous injured neurons there after SNI. Consequently, the proportion of axotomized neurons in the L3, L4 and L5 DRG after SNI was characterized by studying the expression of activating transcription factor 3 (ATF3). Using this marker of nerve injury confirmed that most injured fibers find their cell bodies in the L3 and L4 DRG after SNI in C57BL/6 J mice; this contrasts with their L4 and L5 DRG localization in rats. The spared sural nerve, through which pain hypersensitivity is measured in behavioral studies, mostly projects into the L4 and L5 DRG. Conclusions The complex regulation of Navs, together with the anatomical rostral shift of the DRG harboring injured fibers in C57BL/6 J mice, emphasize that caution is necessary and preliminary anatomical experiments should be carried out for gene and protein expression studies after SNI in mouse strains.
Collapse
Affiliation(s)
- Cédric J Laedermann
- Pain Center, Department of Anesthesiology, University Hospital Center and University of Lausanne, Lausanne 1011, Switzerland.
| | | | | | | |
Collapse
|
39
|
Calhoun JD, Isom LL. The role of non-pore-forming β subunits in physiology and pathophysiology of voltage-gated sodium channels. Handb Exp Pharmacol 2014; 221:51-89. [PMID: 24737232 DOI: 10.1007/978-3-642-41588-3_4] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Voltage-gated sodium channel β1 and β2 subunits were discovered as auxiliary proteins that co-purify with pore-forming α subunits in brain. The other family members, β1B, β3, and β4, were identified by homology and shown to modulate sodium current in heterologous systems. Work over the past 2 decades, however, has provided strong evidence that these proteins are not simply ancillary ion channel subunits, but are multifunctional signaling proteins in their own right, playing both conducting (channel modulatory) and nonconducting roles in cell signaling. Here, we discuss evidence that sodium channel β subunits not only regulate sodium channel function and localization but also modulate voltage-gated potassium channels. In their nonconducting roles, VGSC β subunits function as immunoglobulin superfamily cell adhesion molecules that modulate brain development by influencing cell proliferation and migration, axon outgrowth, axonal fasciculation, and neuronal pathfinding. Mutations in genes encoding β subunits are linked to paroxysmal diseases including epilepsy, cardiac arrhythmia, and sudden infant death syndrome. Finally, β subunits may be targets for the future development of novel therapeutics.
Collapse
Affiliation(s)
- Jeffrey D Calhoun
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, 48109-5632, USA
| | | |
Collapse
|
40
|
Sakai A, Saitow F, Miyake N, Miyake K, Shimada T, Suzuki H. miR-7a alleviates the maintenance of neuropathic pain through regulation of neuronal excitability. ACTA ACUST UNITED AC 2013; 136:2738-50. [PMID: 23861446 DOI: 10.1093/brain/awt191] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Neuronal damage in the somatosensory system causes intractable chronic neuropathic pain. Plastic changes in sensory neuron excitability are considered the cellular basis of persistent pain. Non-coding microRNAs modulate specific gene translation to impact on diverse cellular functions and their dysregulation causes various diseases. However, their significance in adult neuronal functions and disorders is still poorly understood. Here, we show that miR-7a is a key functional RNA sustaining the late phase of neuropathic pain through regulation of neuronal excitability in rats. In the late phase of neuropathic pain, microarray analysis identified miR-7a as the most robustly decreased microRNA in the injured dorsal root ganglion. Moreover, local induction of miR-7a, using an adeno-associated virus vector, in sensory neurons of injured dorsal root ganglion, suppressed established neuropathic pain. In contrast, miR-7a overexpression had no effect on acute physiological or inflammatory pain. Furthermore, miR-7a downregulation was sufficient to cause pain-related behaviours in intact rats. miR-7a targeted the β2 subunit of the voltage-gated sodium channel, and decreased miR-7a associated with neuropathic pain caused increased β2 subunit protein expression, independent of messenger RNA levels. Consistently, miR-7a overexpression in primary sensory neurons of injured dorsal root ganglion suppressed increased β2 subunit expression and normalized long-lasting hyperexcitability of nociceptive neurons. These findings demonstrate miR-7a downregulation is causally involved in maintenance of neuropathic pain through regulation of neuronal excitability, and miR-7a replenishment offers a novel therapeutic strategy specific for chronic neuropathic pain.
Collapse
Affiliation(s)
- Atsushi Sakai
- Department of Pharmacology, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8602, Japan
| | | | | | | | | | | |
Collapse
|
41
|
Piller N, Decosterd I, Suter MR. Reverse transcription quantitative real-time polymerase chain reaction reference genes in the spared nerve injury model of neuropathic pain: validation and literature search. BMC Res Notes 2013; 6:266. [PMID: 23841944 PMCID: PMC3717014 DOI: 10.1186/1756-0500-6-266] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Accepted: 07/03/2013] [Indexed: 12/22/2022] Open
Abstract
Background The reverse transcription quantitative real-time polymerase chain reaction (RT-qPCR) is a widely used, highly sensitive laboratory technique to rapidly and easily detect, identify and quantify gene expression. Reliable RT-qPCR data necessitates accurate normalization with validated control genes (reference genes) whose expression is constant in all studied conditions. This stability has to be demonstrated. We performed a literature search for studies using quantitative or semi-quantitative PCR in the rat spared nerve injury (SNI) model of neuropathic pain to verify whether any reference genes had previously been validated. We then analyzed the stability over time of 7 commonly used reference genes in the nervous system – specifically in the spinal cord dorsal horn and the dorsal root ganglion (DRG). These were: Actin beta (Actb), Glyceraldehyde-3-phosphate dehydrogenase (GAPDH), ribosomal proteins 18S (18S), L13a (RPL13a) and L29 (RPL29), hypoxanthine phosphoribosyltransferase 1 (HPRT1) and hydroxymethylbilane synthase (HMBS). We compared the candidate genes and established a stability ranking using the geNorm algorithm. Finally, we assessed the number of reference genes necessary for accurate normalization in this neuropathic pain model. Results We found GAPDH, HMBS, Actb, HPRT1 and 18S cited as reference genes in literature on studies using the SNI model. Only HPRT1 and 18S had been once previously demonstrated as stable in RT-qPCR arrays. All the genes tested in this study, using the geNorm algorithm, presented gene stability values (M-value) acceptable enough for them to qualify as potential reference genes in both DRG and spinal cord. Using the coefficient of variation, 18S failed the 50% cut-off with a value of 61% in the DRG. The two most stable genes in the dorsal horn were RPL29 and RPL13a; in the DRG they were HPRT1 and Actb. Using a 0.15 cut-off for pairwise variations we found that any pair of stable reference gene was sufficient for the normalization process. Conclusions In the rat SNI model, we validated and ranked Actb, RPL29, RPL13a, HMBS, GAPDH, HPRT1 and 18S as good reference genes in the spinal cord. In the DRG, 18S did not fulfill stability criteria. The combination of any two stable reference genes was sufficient to provide an accurate normalization.
Collapse
Affiliation(s)
- Nicolas Piller
- Pain Center, Department of Anesthesiology, University Hospital Center and University of Lausanne, Avenue du Bugnon 46, 1011 Lausanne, Switzerland
| | | | | |
Collapse
|
42
|
Dustrude ET, Wilson SM, Ju W, Xiao Y, Khanna R. CRMP2 protein SUMOylation modulates NaV1.7 channel trafficking. J Biol Chem 2013; 288:24316-31. [PMID: 23836888 DOI: 10.1074/jbc.m113.474924] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Voltage-gated sodium channel (NaV) trafficking is incompletely understood. Post-translational modifications of NaVs and/or auxiliary subunits and protein-protein interactions have been posited as NaV-trafficking mechanisms. Here, we tested if modification of the axonal collapsin response mediator protein 2 (CRMP2) by a small ubiquitin-like modifier (SUMO) could affect NaV trafficking; CRMP2 alters the extent of NaV slow inactivation conferred by the anti-epileptic (R)-lacosamide, implying NaV-CRMP2 functional coupling. Expression of a CRMP2 SUMOylation-incompetent mutant (CRMP2-K374A) in neuronal model catecholamine A differentiated (CAD) cells did not alter lacosamide-induced NaV slow inactivation compared with CAD cells expressing wild type CRMP2. Like wild type CRMP2, CRMP2-K374A expressed robustly in CAD cells. Neurite outgrowth, a canonical CRMP2 function, was moderately reduced by the mutation but was still significantly higher than enhanced GFP-transfected cortical neurons. Notably, huwentoxin-IV-sensitive NaV1.7 currents, which predominate in CAD cells, were significantly reduced in CAD cells expressing CRMP2-K374A. Increasing deSUMOylation with sentrin/SUMO-specific protease SENP1 or SENP2 in wild type CRMP2-expressing CAD cells decreased NaV1.7 currents. Consistent with a reduction in current density, biotinylation revealed a significant reduction in surface NaV1.7 levels in CAD cells expressing CRMP2-K374A; surface NaV1.7 expression was also decreased by SENP1 + SENP2 overexpression. Currents in HEK293 cells stably expressing NaV1.7 were reduced by CRMP2-K374A in a manner dependent on the E2-conjugating enzyme Ubc9. No decrement in current density was observed in HEK293 cells co-expressing CRMP2-K374A and NaV1.1 or NaV1.3. Diminution of sodium currents, largely NaV1.7, was recapitulated in sensory neurons expressing CRMP2-K374A. Our study elucidates a novel regulatory mechanism that utilizes CRMP2 SUMOylation to choreograph NaV1.7 trafficking.
Collapse
Affiliation(s)
- Erik T Dustrude
- Paul and Carole Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | | | | | | | | |
Collapse
|
43
|
Abstract
OBJECTIVE A review of the innovative role molecular imaging plays in musculoskeletal radiology is provided. Musculoskeletal molecular imaging is under development in four key areas: imaging the activity of osteoblasts and osteoclasts, imaging of molecular and cellular biomarkers of arthritic joint destruction, cellular imaging of osteomyelitis, and imaging generators of musculoskeletal pain. CONCLUSION Together, these applications suggest that next-generation musculoskeletal radiology will facilitate quantitative visualization of molecular and cellular biomarkers, an advancement that appeared futuristic just a decade ago.
Collapse
|
44
|
Abe T, Shimoda T, Urade M, Hasegawa M, Sugiyo S, Takemura M. c-Fos induction in the brainstem following electrical stimulation of the trigeminal ganglion of chronically mandibular nerve-transected rats. Somatosens Mot Res 2013; 30:175-84. [DOI: 10.3109/08990220.2013.790805] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
45
|
Pitcher GM, Ritchie J, Henry JL. Peripheral neuropathy induces cutaneous hypersensitivity in chronically spinalized rats. PAIN MEDICINE 2013; 14:1057-71. [PMID: 23855791 DOI: 10.1111/pme.12123] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND/OBJECTIVES The present study was aimed at the issue of whether peripheral nerve injury-induced chronic pain is maintained by supraspinal structures governing descending facilitation to the spinal dorsal horn, or whether altered peripheral nociceptive mechanisms sustain central hyperexcitability and, in turn, neuropathic pain. We examined this question by determining the contribution of peripheral/spinal mechanisms, isolated from supraspinal influence(s), in cutaneous hypersensitivity in an animal model of peripheral neuropathy. METHODS Adult rats were spinalized at T8-T9; 8 days later, peripheral neuropathy was induced by implanting a 2-mm polyethylene cuff around the left sciatic nerve. Hind paw withdrawal responses to mechanical or thermal plantar stimulation were evaluated using von Frey filaments or a heat lamp, respectively. RESULTS Spinalized rats without cuff implantation exhibited a moderate decrease in mechanical withdrawal threshold on ~day 10 (P < 0.05) and in thermal withdrawal threshold on ~day 18 (P < 0.05). However, cuff-implanted spinalized rats developed a more rapid and significant decrease in mechanical (~day 4; P < 0.001) and thermal (~day 10; P < 0.05) withdrawal thresholds that remained significantly decreased through the duration of the study. CONCLUSIONS Our findings demonstrate an aberrant peripheral/spinal mechanism that induces and maintains thermal and to a greater degree tactile cutaneous hypersensitivity in the cuff model of neuropathic pain, and raise the prospect that altered peripheral/spinal nociceptive mechanisms in humans with peripheral neuropathy may have a pathologically relevant role in both inducing and sustaining neuropathic pain.
Collapse
Affiliation(s)
- Graham M Pitcher
- Departments of Physiology and Psychiatry, McGill University, Montreal, Quebec, Canada.
| | | | | |
Collapse
|
46
|
Chronic pain treatment: the influence of tricyclic antidepressants on serotonin release and uptake in mast cells. Mediators Inflamm 2013; 2013:340473. [PMID: 23710115 PMCID: PMC3654333 DOI: 10.1155/2013/340473] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Accepted: 04/02/2013] [Indexed: 12/11/2022] Open
Abstract
The involvement of serotonin (5-HT) in chronic pain mechanisms is established. 5-HT inhibits central painful stimuli, but recent data suggests that 5-HT could also enhance pain stimulus from the periphery, where mast cells play an important role. We aimed in our study to clarify the influence of selected tricyclic antidepressants (TCAs) on mast cell function: secretion, uptake, and reuptake of 5-HT, that could interfere with 5-HT levels and in this way contribute to the generation of pain. As an experimental model, we used isolated rat peritoneal mast cells and incubated them with selected TCAs (clomipramine, amitriptyline, doxepin, and imipramine) under different experimental conditions. 5-HT release, uptake, and reuptake were determined spectrofluorometrically. We showed that TCAs were able to inhibit 5-HT secretion from mast cells, as well as uptake of exogenous 5-HT and reuptake of secreted 5-HT back into mast cells. The effects of TCAs were concentration dependent; higher concentrations of TCAs inhibited the secretion of 5-HT induced by compound 48/80, whereas lower concentrations of TCAs inhibited 5-HT uptake. The most effective TCA was halogenated clomipramine. As TCAs are well introduced in chronic pain treatment, the insight into mechanisms of action is important for an understanding of their effect in various pain conditions.
Collapse
|
47
|
Dharmshaktu P, Tayal V, Kalra BS. Efficacy of Antidepressants as Analgesics: A Review. J Clin Pharmacol 2013; 52:6-17. [DOI: 10.1177/0091270010394852] [Citation(s) in RCA: 211] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
48
|
Maschietto M, Girardi S, Dal Maschio M, Scorzeto M, Vassanelli S. Sodium channel β2 subunit promotes filopodia-like processes and expansion of the dendritic tree in developing rat hippocampal neurons. Front Cell Neurosci 2013; 7:2. [PMID: 23355803 PMCID: PMC3555079 DOI: 10.3389/fncel.2013.00002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 01/07/2013] [Indexed: 12/19/2022] Open
Abstract
The β2 auxiliary subunit of voltage-gated sodium channels (VGSCs) appears at early stages of brain development. It is abundantly expressed in the mammalian central nervous system where it forms complexes with different channel isoforms, including Nav1.2. From the structural point of view, β2 is a transmembrane protein: at its extracellular N-terminus an Ig-like type C2 domain mediates the binding to the pore-forming alpha subunit with disulfide bonds and the interactions with the extracellular matrix. Given this structural versatility, β2 has been suggested to play multiple functions ranging from channel targeting to the plasma membrane and gating modulation to control of cell adhesion. We report that, when expressed in Chinese Hamster Ovary cells CHO-K1, the subunit accumulates at the perimetral region of adhesion and particularly in large lamellipodia-like membrane processes where it induces formation of filopodia-like structures. When overexpressed in developing embryonic rat hippocampal neurons in vitro, β2 specifically promotes formation of filopodia-like processes in dendrites leading to expansion of the arborization tree, while axonal branching remains unaltered. In contrast to this striking and highly specific effect on dendritic morphology, the targeting of functional sodium channels to the plasma membrane, including the preferential localization of Nav1.2 at the axon, and their gating properties are only minimally affected. From these and previously reported observations it is suggested that β2, among its multiple functions, may contribute to promote dendritic outgrowth and to regulate neuronal wiring at specific stages of neuronal development.
Collapse
Affiliation(s)
- Marta Maschietto
- Department of Biomedical Sciences, University of Padova Padova, Italy
| | | | | | | | | |
Collapse
|
49
|
Eijkelkamp N, Linley JE, Baker MD, Minett MS, Cregg R, Werdehausen R, Rugiero F, Wood JN. Neurological perspectives on voltage-gated sodium channels. Brain 2012; 135:2585-612. [PMID: 22961543 PMCID: PMC3437034 DOI: 10.1093/brain/aws225] [Citation(s) in RCA: 262] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The activity of voltage-gated sodium channels has long been linked to disorders of neuronal excitability such as epilepsy and chronic pain. Recent genetic studies have now expanded the role of sodium channels in health and disease, to include autism, migraine, multiple sclerosis, cancer as well as muscle and immune system disorders. Transgenic mouse models have proved useful in understanding the physiological role of individual sodium channels, and there has been significant progress in the development of subtype selective inhibitors of sodium channels. This review will outline the functions and roles of specific sodium channels in electrical signalling and disease, focusing on neurological aspects. We also discuss recent advances in the development of selective sodium channel inhibitors.
Collapse
Affiliation(s)
- Niels Eijkelkamp
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, Gower Street, London WC1E 6BT, UK.
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Chen C, Calhoun JD, Zhang Y, Lopez-Santiago L, Zhou N, Davis TH, Salzer JL, Isom LL. Identification of the cysteine residue responsible for disulfide linkage of Na+ channel α and β2 subunits. J Biol Chem 2012; 287:39061-9. [PMID: 22992729 PMCID: PMC3493947 DOI: 10.1074/jbc.m112.397646] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Revised: 09/13/2012] [Indexed: 01/28/2023] Open
Abstract
Voltage-gated Na(+) channels in the brain are composed of a single pore-forming α subunit, one non-covalently linked β subunit (β1 or β3), and one disulfide-linked β subunit (β2 or β4). The final step in Na(+) channel biosynthesis in central neurons is concomitant α-β2 disulfide linkage and insertion into the plasma membrane. Consistent with this, Scn2b (encoding β2) null mice have reduced Na(+) channel cell surface expression in neurons, and action potential conduction is compromised. Here we generated a series of mutant β2 cDNA constructs to investigate the cysteine residue(s) responsible for α-β2 subunit covalent linkage. We demonstrate that a single cysteine-to-alanine substitution at extracellular residue Cys-26, located within the immunoglobulin (Ig) domain, abolishes the covalent linkage between α and β2 subunits. Loss of α-β2 covalent complex formation disrupts the targeting of β2 to nodes of Ranvier in a myelinating co-culture system and to the axon initial segment in primary hippocampal neurons, suggesting that linkage with α is required for normal β2 subcellular localization in vivo. WT β2 subunits are resistant to live cell Triton X-100 detergent extraction from the hippocampal axon initial segment, whereas mutant β2 subunits, which cannot form disulfide bonds with α, are removed by detergent. Taken together, our results demonstrate that α-β2 covalent association via a single, extracellular disulfide bond is required for β2 targeting to specialized neuronal subcellular domains and for β2 association with the neuronal cytoskeleton within those domains.
Collapse
Affiliation(s)
- Chunling Chen
- From the Department of Pharmacology and Cellular and Molecular Biology Program, University of Michigan Medical School, Ann Arbor, Michigan 48109 and
| | - Jeffrey D. Calhoun
- From the Department of Pharmacology and Cellular and Molecular Biology Program, University of Michigan Medical School, Ann Arbor, Michigan 48109 and
| | - Yanqing Zhang
- the Departments of Cell Biology and Neurology and the New York University Neuroscience Institute, New York University School of Medicine, New York, New York 10016
| | - Luis Lopez-Santiago
- From the Department of Pharmacology and Cellular and Molecular Biology Program, University of Michigan Medical School, Ann Arbor, Michigan 48109 and
| | - Ningna Zhou
- From the Department of Pharmacology and Cellular and Molecular Biology Program, University of Michigan Medical School, Ann Arbor, Michigan 48109 and
| | - Tigwa H. Davis
- From the Department of Pharmacology and Cellular and Molecular Biology Program, University of Michigan Medical School, Ann Arbor, Michigan 48109 and
| | - James L. Salzer
- the Departments of Cell Biology and Neurology and the New York University Neuroscience Institute, New York University School of Medicine, New York, New York 10016
| | - Lori L. Isom
- From the Department of Pharmacology and Cellular and Molecular Biology Program, University of Michigan Medical School, Ann Arbor, Michigan 48109 and
| |
Collapse
|