1
|
Li Z, Zahra A, Wang Z, Wu J. Physiological implications of the Slack channel in the central and peripheral nervous systems. Eur J Pharmacol 2025; 997:177482. [PMID: 40058757 DOI: 10.1016/j.ejphar.2025.177482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 02/27/2025] [Accepted: 03/06/2025] [Indexed: 03/17/2025]
Abstract
The Slack channels, encoded by KCNT1, are found in neurons across the central nervous system (CNS) and peripheral nervous system (PNS), generating a delayed outward current known as sodium-activated potassium current (IKNa). IKNa plays a crucial role in shaping neuronal excitability and facilitating adaptation in response to sustained stimulation. Slack channels are crucial for neural plasticity and cognitive function; however, mutations in these channels cause profound learning and developmental abnormalities in humans. The physiological significance of Slack conductance in both systems is still unclear. This review provides a comprehensive overview of the role of Slack channels in both CNS and PNS, along with their associated modulators. It offers a summary of diseases that can result from abnormal expression of the Slack channel in both systems. Furthermore, identifying potent blockers and activators of Slack channels could greatly improve our understanding of Slack functions.
Collapse
Affiliation(s)
- Zhen Li
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, 430070, China
| | - Aqeela Zahra
- Department of Pharmacology, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 440070, China
| | - Zhongyu Wang
- School of Basic Medical Sciences, Guangxi Medical University, Nanning, 530021, China
| | - Jianping Wu
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, 430070, China; Department of Pharmacology, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 440070, China; Beijing Tiantan Hospital, Capital Medical University, Beijing, 10070, China; China National Clinical Research Center for Neurological Diseases, Beijing, 10070, China; Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 10070, China.
| |
Collapse
|
2
|
Nguyen T, Lin Z, Dhanesha N, Patel RB, Lane M, Walters GC, Shutov LP, Strack S, Chauhan AK, Usachev YM. Mitochondrial Ca 2+ uniporter b (MCUb) regulates neuronal Ca 2+ dynamics and resistance to ischemic stroke. Cell Calcium 2025; 128:103013. [PMID: 40058292 DOI: 10.1016/j.ceca.2025.103013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 02/24/2025] [Accepted: 02/26/2025] [Indexed: 03/28/2025]
Abstract
Mitochondrial Ca2+ transport regulates many neuronal functions including synaptic transmission, ATP production, gene expression and neuronal survival. The mitochondrial Ca2+ uniporter (MCU) is the core molecular component of the mitochondrial Ca2+ uptake complex in the inner mitochondrial membrane. MCUb is a paralog of MCU that negatively regulates mitochondrial Ca2+ uptake in the heart and the cells of the immune system. However, the function of MCUb in the brain is largely unknown. Here, we report that MCUb knockout (KO) led to enhanced mitochondrial Ca2+ uptake in cortical neurons. By simultaneously monitoring changes in cytosolic and mitochondrial Ca2+ concentrations, [Ca2+]cyt and [Ca2+]mt, respectively, we also found that MCUb KO reduced the [Ca2+]cyt threshold required to induce mitochondrial uptake in cortical neurons during electrical stimulation. Exposure of cortical neurons to toxic concentrations of glutamate led to a collapse of mitochondrial membrane potential (ΔΨmt) and [Ca2+]cyt deregulation, and MCUb deletion accelerated the development of both events. Furthermore, using the middle cerebral artery occlusion (MCAO) as a model of transient ischemic stroke in mice, we found that MCUb KO significantly increased MCAO-induced brain damage in male, but not female mice. These results suggest that MCUb regulates neuronal Ca2+ dynamics and excitotoxicity and reveal a sex-dependent role of MCUb in controlling resistance to brain damage following ischemic stroke.
Collapse
Affiliation(s)
- Tam Nguyen
- Department of Neuroscience and Pharmacology and Iowa Neuroscience Institute, University of Iowa College of Medicine, Iowa City, IA 52242, USA
| | - Zhihong Lin
- Department of Neuroscience and Pharmacology and Iowa Neuroscience Institute, University of Iowa College of Medicine, Iowa City, IA 52242, USA
| | - Nirav Dhanesha
- Department of Internal Medicine, University of Iowa College of Medicine, Iowa City, IA 52242, USA
| | - Rakesh B Patel
- Department of Internal Medicine, University of Iowa College of Medicine, Iowa City, IA 52242, USA
| | - Mallorie Lane
- Department of Neuroscience and Pharmacology and Iowa Neuroscience Institute, University of Iowa College of Medicine, Iowa City, IA 52242, USA
| | - Grant C Walters
- Department of Neuroscience and Pharmacology and Iowa Neuroscience Institute, University of Iowa College of Medicine, Iowa City, IA 52242, USA
| | - Leonid P Shutov
- Department of Neuroscience and Pharmacology and Iowa Neuroscience Institute, University of Iowa College of Medicine, Iowa City, IA 52242, USA
| | - Stefan Strack
- Department of Neuroscience and Pharmacology and Iowa Neuroscience Institute, University of Iowa College of Medicine, Iowa City, IA 52242, USA
| | - Anil K Chauhan
- Department of Internal Medicine, University of Iowa College of Medicine, Iowa City, IA 52242, USA
| | - Yuriy M Usachev
- Department of Neuroscience and Pharmacology and Iowa Neuroscience Institute, University of Iowa College of Medicine, Iowa City, IA 52242, USA.
| |
Collapse
|
3
|
Li Y, Li C, Zhu H, Chu Y. TRPV1 in Dorsal Root Ganglion Contributed to Chronic Pancreatitis Pain. J Pain Palliat Care Pharmacother 2025:1-9. [PMID: 40371900 DOI: 10.1080/15360288.2025.2500984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 02/24/2025] [Accepted: 04/27/2025] [Indexed: 05/16/2025]
Abstract
Chronic pancreatitis presents a formidable challenge in pain management, often leading to significant suffering and reduced quality of life for affected individuals. The intricate interplay of factors contributing to this pain, including inflammation and neural sensitization, has garnered increasing attention in recent research. Among the key players in this scenario are the transient receptor potential vanilloid 1(TRPV1) channels located in dorsal root ganglion (DRG) neurons. These channels, known for their role in pain perception, exhibit heightened sensitivity and altered expression patterns in the context of chronic pancreatitis. Sensitization of TRPV1 channels amplifies their response to various pain triggers, exacerbating the perception of discomfort. Furthermore, dysregulated expression of TRPV1 within DRG neurons contributes to the chronic pain phenotype associated with pancreatitis. Understanding the nuanced mechanisms governing TRPV1 modulation in DRG neurons promises to unlock novel therapeutic avenues for managing chronic pancreatitis pain. By targeting TRPV1 channels specifically in DRG neurons, researchers aim to develop treatments that alleviate pain while minimizing adverse effects, ultimately offering hope for improved outcomes and enhanced well-being for individuals grappling with this debilitating condition.
Collapse
Affiliation(s)
- Yali Li
- Department of Intensive Care Unit, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, China
| | - Chenshuai Li
- Department of Pediatrics, Tianjin Beichen Hospital of Traditional Chinese Medicine, Tianjin, China
| | - Haiyun Zhu
- Department of Intensive Care Unit, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, China
| | - Yuru Chu
- Department of Intensive Care Unit, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, China
| |
Collapse
|
4
|
Shen Y, Lin P. The Role of Cytokines in Postherpetic Neuralgia. J Integr Neurosci 2025; 24:25829. [PMID: 40302252 DOI: 10.31083/jin25829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/02/2024] [Accepted: 10/23/2024] [Indexed: 05/02/2025] Open
Abstract
Nerve injury is a significant cause of postherpetic neuralgia (PHN). It is marked by upregulated expression of cytokines secreted by immune cells such as tumor necrosis factor alpha, interleukin 1 beta (IL-1β), IL-6, IL-18, and IL-10. In neuropathic pain (NP) due to nerve injury, cytokines are important for the induction of neuroinflammation, activation of glial cells, and expression of cation channels. The release of chemokines due to nerve injury promotes immune cell infiltration, recruiting inflammatory cytokines and further amplifying the inflammatory response. The resulting disequilibrium in neuroimmune response and neuroinflammation leads to a reduction of nerve fibers, altered nerve excitability, and neuralgia. PHN is a typical NP and cytokines may induce PHN by promoting central and peripheral sensitization. Currently, treating PHN is challenging and research on the role of cytokine signaling pathways in PHN is lacking. This review summarizes the potential mechanisms of cytokine-mediated PHN and discusses the cytokine signaling pathways associated with the central and peripheral sensitization of PHN. By elucidating the mechanisms of cytokines, the cells and molecules that regulate cytokines, and their signaling systems in PHN, this review reveals important research developments regarding cytokines and their signaling pathways mediating PHN, highlighting new targets of action for the development of analgesic drugs.
Collapse
Affiliation(s)
- Yunyan Shen
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, 310053 Hangzhou, Zhejiang, China
| | - Ping Lin
- Department of Geriatrics, Hangzhou Third People's Hospital, 310009 Hangzhou, Zhejiang, China
| |
Collapse
|
5
|
Chu CC, Hu YH, Li GZ, Chen J, Zhang NN, Gu YX, Wu SY, Zhang HF, Xu YY, Guo HL, Tian X, Chen F. Unveiling the significance of AKAP79/150 in the nervous system disorders: An emerging opportunity for future therapies? Neurobiol Dis 2025; 206:106812. [PMID: 39864527 DOI: 10.1016/j.nbd.2025.106812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/19/2024] [Accepted: 01/22/2025] [Indexed: 01/28/2025] Open
Abstract
A-kinase anchoring protein 79/150 (AKAP79/150) is a crucial scaffolding protein that positions various proteins at specific synaptic sites to modulate excitatory synaptic intensity. As our understanding of AKAP79/150's biology deepens, along with its significant role in the pathophysiology of various human disorders, there is growing evidence that reveals new opportunities for therapeutic interventions. In this review, we examine the fundamental structure and primary functions of AKAP79/150, emphasizing its pathophysiological mechanisms in different nervous system disorders, particularly inflammatory pain, epilepsy, depression, and Alzheimer's disease. We also discuss its potential therapeutic implications for patients suffering from these conditions.
Collapse
Affiliation(s)
- Chen-Chao Chu
- Department of Pharmacy, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Ya-Hui Hu
- Department of Pharmacy, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Gui-Zhou Li
- Department of Pharmacy, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Jiang Chen
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Ning-Ning Zhang
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Yi-Xue Gu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Shi-Yu Wu
- Department of Pharmacy, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Hai-Feng Zhang
- Department of Pharmacy, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Yang-Yang Xu
- Department of Pharmacy, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Hong-Li Guo
- Department of Pharmacy, Children's Hospital of Nanjing Medical University, Nanjing, China.
| | - Xin Tian
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China; Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Chongqing Medical University, Chongqing, China.
| | - Feng Chen
- Department of Pharmacy, Children's Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
6
|
Parthasarathy A, Anishkin A, Xie Y, Drachuk K, Nishijma Y, Fang J, Koukouritaki SB, Wilcox DA, Zhang DX. Phosphorylation of distal C-terminal residues promotes TRPV4 channel activation in response to arachidonic acid. J Biol Chem 2025; 301:108260. [PMID: 39909371 PMCID: PMC11903807 DOI: 10.1016/j.jbc.2025.108260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 01/22/2025] [Accepted: 01/27/2025] [Indexed: 02/07/2025] Open
Abstract
Transient receptor potential vanilloid 4 (TRPV4) is a Ca2+-permeable channel activated by diverse physical and chemical stimuli, including mechanical stress and endogenous lipid arachidonic acid (AA) and its metabolites. Phosphorylation of TRPV4 by protein kinase A (PKA) and protein kinase C (PKC) is a predominant mechanism for channel regulation, especially in the cytoplasmic domains due to their importance in protein assembly, and channelopathies. However, studies corresponding to phosphorylation sites for these kinases remain incomplete. We investigated the role of Ser-823 residue as a potential phosphorylation site in regulating TRPV4 activity and chemical agonist-induced channel activation. Using mass spectrometry, we identified a new phosphorylation site Ser-823 residue and confirmed the previously known phosphorylation site Ser-824 in the C-terminal tail. The low level of phosphorylation at Ser-823 was stimulated by PKC and to a lesser extent by PKA in human coronary artery endothelial cells (HCAECs) and human embryonic kidney 293 (HEK 293) cells. AA-induced TRPV4 activation was enhanced in the phosphomimetic S823E but was blunted in the S823A/S824A mutants, whereas the channel activation by the synthetic agonist GSK1016790A was unaffected. Further, TRPV4 activation by AA but not GSK1016790A was blunted or abolished by PKA inhibitor alone or in combination with PKC inhibitor, respectively. Using computational modeling, we refined a previously proposed structural model for TRPV4 regulation by Ser-823 and Ser-824 phosphorylation. Together, these results provide insight into how stimuli-specific TRPV4 activation is regulated by the phosphorylation of discrete residues (e.g., Ser-823 and Ser-824) in the C-terminal domains of the TRPV4 channel.
Collapse
Affiliation(s)
- Aravind Parthasarathy
- Department of Medicine, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Andriy Anishkin
- Department of Biology, University of Maryland, College Park, Maryland, USA
| | - Yangjing Xie
- Department of Medicine, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Kostiantyn Drachuk
- Department of Medicine, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Yoshinori Nishijma
- Department of Medicine, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Juan Fang
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA; Children's Research Institute, Children's Wisconsin, Milwaukee, Wisconsin, USA
| | - Sevasti B Koukouritaki
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA; Children's Research Institute, Children's Wisconsin, Milwaukee, Wisconsin, USA
| | - David A Wilcox
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA; Children's Research Institute, Children's Wisconsin, Milwaukee, Wisconsin, USA
| | - David X Zhang
- Department of Medicine, Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA; Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
| |
Collapse
|
7
|
Moriyama S, Tatematsu K, Hinuma S, Kuroda S. Role of phosphorylation and vanilloid ligand structure in ligand-dependent differential activations of transient receptor potential vanilloid 1. Biosci Biotechnol Biochem 2024; 88:1316-1325. [PMID: 39217101 DOI: 10.1093/bbb/zbae119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Vanilloid analogs, which can activate transient receptor potential vanilloid 1 (TRPV1), have been classified into two types based on susceptibility to forskolin (FSK). Treatment of cells expressing TRPV1 with FSK enhances TRPV1 responses to capsaicin-type ligands while diminishing the responses to eugenol-type ligands. In this study, we determined the effect of FSK on the activation of TRPV1 stimulated with vanilloid ligands, through the influx of Ca2+ in HEK293T cells expressing TRPV1. Our findings suggest that the effects of FSK can be attributed to the phosphorylation of TRPV1, as evidenced by using a protein kinase A inhibitor and TRPV1 mutants at potential phosphorylation sites. Furthermore, we examined the structure-activity relationship of 13 vanilloid analogs. Our results indicated that vanilloid compounds could be classified into three types, that is the previously reported two types and a novel type of 10-shogaol, by which TRPV1 activation was insusceptible to the FSK treatment.
Collapse
Affiliation(s)
- Sakura Moriyama
- SANKEN, Osaka University, Ibaraki, Osaka, Japan
- Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, Japan
| | | | | | - Shun'ichi Kuroda
- SANKEN, Osaka University, Ibaraki, Osaka, Japan
- Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
8
|
do Nascimento AM, Marques RB, Roldão AP, Rodrigues AM, Eslava RM, Dale CS, Reis EM, Schechtman D. Exploring protein-protein interactions for the development of new analgesics. Sci Signal 2024; 17:eadn4694. [PMID: 39378285 DOI: 10.1126/scisignal.adn4694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 06/13/2024] [Accepted: 09/16/2024] [Indexed: 10/10/2024]
Abstract
The development of new analgesics has been challenging. Candidate drugs often have limited clinical utility due to side effects that arise because many drug targets are involved in signaling pathways other than pain transduction. Here, we explored the potential of targeting protein-protein interactions (PPIs) that mediate pain signaling as an approach to developing drugs to treat chronic pain. We reviewed the approaches used to identify small molecules and peptide modulators of PPIs and their ability to decrease pain-like behaviors in rodent animal models. We analyzed data from rodent and human sensory nerve tissues to build associated signaling networks and assessed both validated and potential interactions and the structures of the interacting domains that could inform the design of synthetic peptides and small molecules. This resource identifies PPIs that could be explored for the development of new analgesics, particularly between scaffolding proteins and receptors for various growth factors and neurotransmitters, as well as ion channels and other enzymes. Targeting the adaptor function of CBL by blocking interactions between its proline-rich carboxyl-terminal domain and its SH3-domain-containing protein partners, such as GRB2, could disrupt endosomal signaling induced by pain-associated growth factors. This approach would leave intact its E3-ligase functions, which are mediated by other domains and are critical for other cellular functions. This potential of PPI modulators to be more selective may mitigate side effects and improve the clinical management of pain.
Collapse
Affiliation(s)
- Alexandre Martins do Nascimento
- Department of Biochemistry, Chemistry Institute, University of São Paulo, SP 05508-000, Brazil
- Laboratory of Neuromodulation of Experimental Pain (LaNed), Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, SP 05508-000, Brazil
| | - Rauni Borges Marques
- Department of Biochemistry, Chemistry Institute, University of São Paulo, SP 05508-000, Brazil
- Interunit Graduate Program in Bioinformatics, University of São Paulo, SP 05508-000, Brazil
| | - Allan Pradelli Roldão
- Department of Biochemistry, Chemistry Institute, University of São Paulo, SP 05508-000, Brazil
| | - Ana Maria Rodrigues
- Department of Biochemistry, Chemistry Institute, University of São Paulo, SP 05508-000, Brazil
| | - Rodrigo Mendes Eslava
- Department of Biochemistry, Chemistry Institute, University of São Paulo, SP 05508-000, Brazil
| | - Camila Squarzoni Dale
- Laboratory of Neuromodulation of Experimental Pain (LaNed), Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, SP 05508-000, Brazil
| | - Eduardo Moraes Reis
- Department of Biochemistry, Chemistry Institute, University of São Paulo, SP 05508-000, Brazil
| | - Deborah Schechtman
- Department of Biochemistry, Chemistry Institute, University of São Paulo, SP 05508-000, Brazil
| |
Collapse
|
9
|
Guilleminault L, Grassin-Delyle S, Mazzone SB. Drugs Targeting Cough Receptors: New Therapeutic Options in Refractory or Unexplained Chronic Cough. Drugs 2024; 84:763-777. [PMID: 38904926 DOI: 10.1007/s40265-024-02047-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/06/2024] [Indexed: 06/22/2024]
Abstract
Refractory chronic cough is a disabling disease with very limited therapeutic options. A better understanding of cough pathophysiology has led to the development of emerging drugs targeting cough receptors. Recent strides have illuminated novel therapeutic avenues, notably centred on modulating transient receptor potential (TRP) channels, purinergic receptors, and neurokinin receptors. By modulating these receptors, the goal is to intervene in the sensory pathways that trigger cough reflexes, thereby providing relief without compromising vital protective mechanisms. These innovative pharmacotherapies hold promise for improvement of refractory chronic cough by offering improved efficacy and potentially mitigating adverse effects associated with current recommended treatments. A deeper comprehension of their precise mechanisms of action and clinical viability is imperative for optimising therapeutic interventions and elevating patient care standards in respiratory health. This review delineates the evolving landscape of drug development in this domain, emphasising the significance of these advancements in reshaping the paradigm of cough management.
Collapse
Affiliation(s)
- Laurent Guilleminault
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291, CNRS UMR5051, University Toulouse III, 31024, Toulouse, France.
- Department of Respiratory Medicine, Faculty of Medicine, Hôpital Larrey, Toulouse University Hospital, 24 chemin de Pouvourville, 31059, Toulouse, France.
| | - Stanislas Grassin-Delyle
- Exhalomics®, Hôpital Foch, Suresnes, France
- Département de Biotechnologie de la Santé, Université Paris-Saclay, UVSQ, INSERM, Infection et inflammation (2I), U1173, Montigny le Bretonneux, France
| | - Stuart B Mazzone
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
10
|
Simmons SC, Flerlage WJ, Langlois LD, Shepard RD, Bouslog C, Thomas EH, Gouty KM, Sanderson JL, Gouty S, Cox BM, Dell'Acqua ML, Nugent FS. AKAP150-anchored PKA regulates synaptic transmission and plasticity, neuronal excitability and CRF neuromodulation in the mouse lateral habenula. Commun Biol 2024; 7:345. [PMID: 38509283 PMCID: PMC10954712 DOI: 10.1038/s42003-024-06041-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 03/08/2024] [Indexed: 03/22/2024] Open
Abstract
The scaffolding A-kinase anchoring protein 150 (AKAP150) is critically involved in kinase and phosphatase regulation of synaptic transmission/plasticity, and neuronal excitability. Emerging evidence also suggests that AKAP150 signaling may play a key role in brain's processing of rewarding/aversive experiences, however its role in the lateral habenula (LHb, as an important brain reward circuitry) is completely unknown. Using whole cell patch clamp recordings in LHb of male wildtype and ΔPKA knockin mice (with deficiency in AKAP-anchoring of PKA), here we show that the genetic disruption of PKA anchoring to AKAP150 significantly reduces AMPA receptor-mediated glutamatergic transmission and prevents the induction of presynaptic endocannabinoid-mediated long-term depression in LHb neurons. Moreover, ΔPKA mutation potentiates GABAA receptor-mediated inhibitory transmission while increasing LHb intrinsic excitability through suppression of medium afterhyperpolarizations. ΔPKA mutation-induced suppression of medium afterhyperpolarizations also blunts the synaptic and neuroexcitatory actions of the stress neuromodulator, corticotropin releasing factor (CRF), in mouse LHb. Altogether, our data suggest that AKAP150 complex signaling plays a critical role in regulation of AMPA and GABAA receptor synaptic strength, glutamatergic plasticity and CRF neuromodulation possibly through AMPA receptor and potassium channel trafficking and endocannabinoid signaling within the LHb.
Collapse
Affiliation(s)
- Sarah C Simmons
- Uniformed Services University of the Health Sciences, Department of Pharmacology and Molecular Therapeutics, Bethesda, MD, 20814, USA
| | - William J Flerlage
- Uniformed Services University of the Health Sciences, Department of Pharmacology and Molecular Therapeutics, Bethesda, MD, 20814, USA
| | - Ludovic D Langlois
- Uniformed Services University of the Health Sciences, Department of Pharmacology and Molecular Therapeutics, Bethesda, MD, 20814, USA
| | - Ryan D Shepard
- Uniformed Services University of the Health Sciences, Department of Pharmacology and Molecular Therapeutics, Bethesda, MD, 20814, USA
| | - Christopher Bouslog
- Uniformed Services University of the Health Sciences, Department of Pharmacology and Molecular Therapeutics, Bethesda, MD, 20814, USA
| | - Emily H Thomas
- Uniformed Services University of the Health Sciences, Department of Pharmacology and Molecular Therapeutics, Bethesda, MD, 20814, USA
| | - Kaitlyn M Gouty
- Uniformed Services University of the Health Sciences, Department of Pharmacology and Molecular Therapeutics, Bethesda, MD, 20814, USA
| | - Jennifer L Sanderson
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Shawn Gouty
- Uniformed Services University of the Health Sciences, Department of Pharmacology and Molecular Therapeutics, Bethesda, MD, 20814, USA
| | - Brian M Cox
- Uniformed Services University of the Health Sciences, Department of Pharmacology and Molecular Therapeutics, Bethesda, MD, 20814, USA
| | - Mark L Dell'Acqua
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, 80045, USA.
| | - Fereshteh S Nugent
- Uniformed Services University of the Health Sciences, Department of Pharmacology and Molecular Therapeutics, Bethesda, MD, 20814, USA.
| |
Collapse
|
11
|
Jiang C, Zhao J, Zhang Y, Zhu X. Role of EPAC1 in chronic pain. Biochem Biophys Rep 2024; 37:101645. [PMID: 38304575 PMCID: PMC10832381 DOI: 10.1016/j.bbrep.2024.101645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 01/08/2024] [Accepted: 01/10/2024] [Indexed: 02/03/2024] Open
Abstract
Chronic pain usually lasts over three months and commonly occurs in chronic diseases (cancer, arthritis, and diabetes), injuries (herniated discs, torn ligaments), and many major pain disorders (neuropathic pain, fibromyalgia, chronic headaches). Unfortunately, there is currently a lack of effective treatments to help people with chronic pain to achieve complete relief. Therefore,it is particularly important to understand the mechanism of chronic pain and find new therapeutic targets. The exchange protein directly activated by cyclic adenosine monophosphate(cAMP) (EPAC) has been recognized for its functions in nerve regeneration, stimulating insulin release, controlling vascular pressure, and controlling other metabolic activities. In recent years, many studies have found that the subtype of EPAC, EPAC1 is involved in the regulation of neuroinflammation and plays a crucial role in the regulation of pain, which is expected to become a new therapeutic target for chronic pain. This article reviews the major contributions of EPAC1 in chronic pain.
Collapse
Affiliation(s)
- Chenlu Jiang
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
- Medical School of Nantong University, Nantong, 226001, China
| | - Jiacheng Zhao
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
- Medical School of Nantong University, Nantong, 226001, China
| | - Yihang Zhang
- Medical School of Nantong University, Nantong, 226001, China
| | - Xiang Zhu
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, China
| |
Collapse
|
12
|
Simmons S, Flerlage W, Langlois L, Shepard R, Bouslog C, Thomas E, Gouty K, Sanderson J, Gouty S, Cox B, Dell’Acqua M, Nugent F. AKAP150-anchored PKA regulation of synaptic transmission and plasticity, neuronal excitability and CRF neuromodulation in the lateral habenula. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.06.570160. [PMID: 38106086 PMCID: PMC10723374 DOI: 10.1101/2023.12.06.570160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Numerous studies of hippocampal synaptic function in learning and memory have established the functional significance of the scaffolding A-kinase anchoring protein 150 (AKAP150) in kinase and phosphatase regulation of synaptic receptor and ion channel trafficking/function and hence synaptic transmission/plasticity, and neuronal excitability. Emerging evidence also suggests that AKAP150 signaling may play a critical role in brain's processing of rewarding/aversive experiences. Here we focused on an unexplored role of AKAP150 in the lateral habenula (LHb), a diencephalic brain region that integrates and relays negative reward signals from forebrain striatal and limbic structures to midbrain monoaminergic centers. LHb aberrant activity (specifically hyperactivity) is also linked to depression. Using whole cell patch clamp recordings in LHb of male wildtype (WT) and ΔPKA knockin mice (with deficiency in AKAP-anchoring of PKA), we found that the genetic disruption of PKA anchoring to AKAP150 significantly reduced AMPA receptor (AMPAR)-mediated glutamatergic transmission and prevented the induction of presynaptic endocannabinoid (eCB)-mediated long-term depression (LTD) in LHb neurons. Moreover, ΔPKA mutation potentiated GABAA receptor (GABAAR)-mediated inhibitory transmission postsynaptically while increasing LHb intrinsic neuronal excitability through suppression of medium afterhyperpolarizations (mAHPs). Given that LHb is a highly stress-responsive brain region, we further tested the effects of corticotropin releasing factor (CRF) stress neuromodulator on synaptic transmission and intrinsic excitability of LHb neurons in WT and ΔPKA mice. As in our earlier study in rat LHb, CRF significantly suppressed GABAergic transmission onto LHb neurons and increased intrinsic excitability by diminishing small-conductance potassium (SK) channel-mediated mAHPs. ΔPKA mutation-induced suppression of mAHPs also blunted the synaptic and neuroexcitatory actions of CRF in mouse LHb. Altogether, our data suggest that AKAP150 complex signaling plays a critical role in regulation of AMPAR and GABAAR synaptic strength, glutamatergic plasticity and CRF neuromodulation possibly through AMPAR and potassium channel trafficking and eCB signaling within the LHb.
Collapse
Affiliation(s)
- S.C. Simmons
- Uniformed Services University of the Health Sciences, Department of Pharmacology, Bethesda, Maryland 20814, USA
| | - W.J. Flerlage
- Uniformed Services University of the Health Sciences, Department of Pharmacology, Bethesda, Maryland 20814, USA
| | - L.D. Langlois
- Uniformed Services University of the Health Sciences, Department of Pharmacology, Bethesda, Maryland 20814, USA
| | - R.D. Shepard
- Uniformed Services University of the Health Sciences, Department of Pharmacology, Bethesda, Maryland 20814, USA
| | - C. Bouslog
- Uniformed Services University of the Health Sciences, Department of Pharmacology, Bethesda, Maryland 20814, USA
| | - E.H. Thomas
- Uniformed Services University of the Health Sciences, Department of Pharmacology, Bethesda, Maryland 20814, USA
| | - K.M. Gouty
- Uniformed Services University of the Health Sciences, Department of Pharmacology, Bethesda, Maryland 20814, USA
| | - J.L. Sanderson
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - S. Gouty
- Uniformed Services University of the Health Sciences, Department of Pharmacology, Bethesda, Maryland 20814, USA
| | - B.M. Cox
- Uniformed Services University of the Health Sciences, Department of Pharmacology, Bethesda, Maryland 20814, USA
| | - M.L. Dell’Acqua
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - F.S. Nugent
- Uniformed Services University of the Health Sciences, Department of Pharmacology, Bethesda, Maryland 20814, USA
| |
Collapse
|
13
|
Röderer P, Belu A, Heidrich L, Siobal M, Isensee J, Prolingheuer J, Janocha E, Valdor M, Hagendorf S, Bahrenberg G, Opitz T, Segschneider M, Haupt S, Nitzsche A, Brüstle O, Hucho T. Emergence of nociceptive functionality and opioid signaling in human induced pluripotent stem cell-derived sensory neurons. Pain 2023; 164:1718-1733. [PMID: 36727909 PMCID: PMC10348637 DOI: 10.1097/j.pain.0000000000002860] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 10/31/2022] [Accepted: 11/15/2022] [Indexed: 02/03/2023]
Abstract
ABSTRACT Induced pluripotent stem cells (iPSCs) have enabled the generation of various difficult-to-access cell types such as human nociceptors. A key challenge associated with human iPSC-derived nociceptors (hiPSCdNs) is their prolonged functional maturation. While numerous studies have addressed the expression of classic neuronal markers and ion channels in hiPSCdNs, the temporal development of key signaling cascades regulating nociceptor activity has remained largely unexplored. In this study, we used an immunocytochemical high-content imaging approach alongside electrophysiological staging to assess metabotropic and ionotropic signaling of large scale-generated hiPSCdNs across 70 days of in vitro differentiation. During this period, the resting membrane potential became more hyperpolarized, while rheobase, action potential peak amplitude, and membrane capacitance increased. After 70 days, hiPSCdNs exhibited robust physiological responses induced by GABA, pH shift, ATP, and capsaicin. Direct activation of protein kinase A type II (PKA-II) through adenylyl cyclase stimulation with forskolin resulted in PKA-II activation at all time points. Depolarization-induced activation of PKA-II emerged after 35 days of differentiation. However, effective inhibition of forskolin-induced PKA-II activation by opioid receptor agonists required 70 days of in vitro differentiation. Our results identify a pronounced time difference between early expression of functionally important ion channels and emergence of regulatory metabotropic sensitizing and desensitizing signaling only at advanced stages of in vitro cultivation, suggesting an independent regulation of ionotropic and metabotropic signaling. These data are relevant for devising future studies into the development and regulation of human nociceptor function and for defining time windows suitable for hiPSCdN-based drug discovery.
Collapse
Affiliation(s)
- Pascal Röderer
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty and University Hospital Bonn, Bonn
- LIFE & BRAIN GmbH, Cellomics Unit, Bonn, Germany, Germany
| | - Andreea Belu
- Translational Pain Research, Department of Anaesthesiology and Intensive Care Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Luzia Heidrich
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty and University Hospital Bonn, Bonn
- LIFE & BRAIN GmbH, Cellomics Unit, Bonn, Germany, Germany
| | - Maike Siobal
- Translational Pain Research, Department of Anaesthesiology and Intensive Care Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Jörg Isensee
- Translational Pain Research, Department of Anaesthesiology and Intensive Care Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Jonathan Prolingheuer
- Translational Pain Research, Department of Anaesthesiology and Intensive Care Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | | | | | | | | | - Thoralf Opitz
- Institute of Experimental Epileptology and Cognition Research, University of Bonn, Bonn, Germany
| | - Michaela Segschneider
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty and University Hospital Bonn, Bonn
| | - Simone Haupt
- LIFE & BRAIN GmbH, Cellomics Unit, Bonn, Germany, Germany
| | - Anja Nitzsche
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty and University Hospital Bonn, Bonn
- LIFE & BRAIN GmbH, Cellomics Unit, Bonn, Germany, Germany
| | - Oliver Brüstle
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty and University Hospital Bonn, Bonn
- LIFE & BRAIN GmbH, Cellomics Unit, Bonn, Germany, Germany
| | - Tim Hucho
- Translational Pain Research, Department of Anaesthesiology and Intensive Care Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
14
|
Zhang Y, Jeske NA. A-kinase anchoring protein 79/150 coordinates α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor sensitization in sensory neurons. Mol Pain 2023; 19:17448069231222406. [PMID: 38073552 PMCID: PMC10722943 DOI: 10.1177/17448069231222406] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/30/2023] [Accepted: 11/30/2023] [Indexed: 12/17/2023] Open
Abstract
Changes in sensory afferent activity contribute to the transition from acute to chronic pain. However, it is unlikely that a single sensory receptor is entirely responsible for persistent pain. It is more probable that extended changes to multiple receptor proteins expressed by afferent neurons support persistent pain. A-Kinase Anchoring Protein 79/150 (AKAP) is an intracellular scaffolding protein expressed in sensory neurons that spatially and temporally coordinates signaling events. Since AKAP scaffolds biochemical modifications of multiple TRP receptors linked to pain phenotypes, we probed for other ionotropic receptors that may be mediated by AKAP and contribute to persistent pain. Here, we identify a role for AKAP modulation of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid Receptor (AMPA-R) functionality in sensory neurons. Pharmacological manipulation of distinct AMPA-R subunits significantly reduces persistent mechanical hypersensitivity observed during hyperalgesic priming. Stimulation of both protein kinases C and A (PKC, PKA, respectively) modulate AMPA-R subunit GluR1 and GluR2 phosphorylation and surface expression in an AKAP-dependent manner in primary cultures of DRG neurons. Furthermore, AKAP knock out reduces sensitized AMPA-R responsivity in DRG neurons. Collectively, these data indicate that AKAP scaffolds AMPA-R subunit organization in DRG neurons that may contribute to the transition from acute-to-chronic pain.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Oral and Maxillofacial Surgery, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Nathaniel A Jeske
- Department of Oral and Maxillofacial Surgery, University of Texas Health San Antonio, San Antonio, TX, USA
- Department of Pharmacology, University of Texas Health San Antonio, San Antonio, TX, USA
- Department of Physiology, University of Texas Health San Antonio, San Antonio, TX, USA
| |
Collapse
|
15
|
Pellegrino A, Mükusch S, Seitz V, Stein C, Herberg FW, Seitz H. Transient Receptor Potential Vanilloid 1 Signaling Is Independent on Protein Kinase A Phosphorylation of Ankyrin-Rich Membrane Spanning Protein. MEDICAL SCIENCES (BASEL, SWITZERLAND) 2022; 10:medsci10040063. [PMID: 36412904 PMCID: PMC9680306 DOI: 10.3390/medsci10040063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/11/2022] [Accepted: 11/14/2022] [Indexed: 11/18/2022]
Abstract
The sensory ion channel transient receptor potential vanilloid 1 (TRPV1) is mainly expressed in small to medium sized dorsal root ganglion neurons, which are involved in the transfer of acute noxious thermal and chemical stimuli. The Ankyrin-rich membrane spanning protein (ARMS) interaction with TRPV1 is modulated by protein kinase A (PKA) mediating sensitization. Here, we hypothesize that PKA phosphorylation sites of ARMS are crucial for the modulation of TRPV1 function, and that the phosphorylation of ARMS is facilitated by the A-kinase anchoring protein 79 (AKAP79). We used transfected HEK293 cells, immunoprecipitation, calcium flux, and patch clamp experiments to investigate potential PKA phosphorylation sites in ARMS and in ARMS-related peptides. Additionally, experiments were done to discriminate between PKA and protein kinase D (PKD) phosphorylation. We found different interaction ratios for TRPV1 and ARMS mutants lacking PKA phosphorylation sites. The degree of TRPV1 sensitization by ARMS mutants is independent on PKA phosphorylation. AKAP79 was also involved in the TRPV1/ARMS/PKA signaling complex. These data show that ARMS is a PKA substrate via AKAP79 in the TRPV1 signaling complex and that all four proteins interact physically, regulating TRPV1 sensitization in transfected HEK293 cells. To assess the physiological and/or therapeutic significance of these findings, similar investigations need to be performed in native neurons and/or in vivo.
Collapse
Affiliation(s)
- Antonio Pellegrino
- Fraunhofer Institute for Cell Therapy and Immunology, 14476 Potsdam, Germany
| | - Sandra Mükusch
- Fraunhofer Institute for Cell Therapy and Immunology, 14476 Potsdam, Germany
| | - Viola Seitz
- Institute of Experimental Anaesthesiology, Charité—Universitätsmedizin Berlin, 12203 Berlin, Germany
- Brandenburg Medical School Theodor Fontane, Fehrbelliner Str. 38, 16816 Neuruppin, Germany
| | - Christoph Stein
- Institute of Experimental Anaesthesiology, Charité—Universitätsmedizin Berlin, 12203 Berlin, Germany
| | | | - Harald Seitz
- Fraunhofer Institute for Cell Therapy and Immunology, 14476 Potsdam, Germany
- Correspondence: ; +49-331-58187-208
| |
Collapse
|
16
|
Robilotto GL, Mohapatra DP, Shepherd AJ, Mickle AD. Role of Src kinase in regulating protein kinase C mediated phosphorylation of TRPV1. Eur J Pain 2022; 26:1967-1978. [PMID: 35900227 PMCID: PMC9483845 DOI: 10.1002/ejp.2017] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 06/19/2022] [Accepted: 07/23/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND Transient receptor potential vanilloid-1 (TRPV1), activated by heat, acidic pH, endogenous vanilloids and capsaicin, is essential for thermal hyperalgesia. Under inflammatory conditions, phosphorylation of TRPV1 by protein kinase C (PKC) can sensitize the channel and decrease the activation threshold. Src kinase also phosphorylates TRPV1, promoting channel trafficking to the plasma membrane. These post-translational modifications are important for several chronic pain conditions. This study presents a previously undescribed relationship between Src and PKC phosphorylation of TRPV1, influencing the thermal hypersensitivity associated with TRPV1 activation. METHODS We assessed TRPV1 channel activity using intracellular calcium imaging and patch-clamp electrophysiology in mouse dorsal root ganglion cultures. Additionally, we used behavioural experiments to evaluate plantar thermal sensitivity following intraplantar injections of activators of known modulators of TRPV1 with and without an Src antagonist. RESULTS Using calcium imaging and patch-clamp techniques, we demonstrated that pharmacological inhibition of Src kinase or mutation of the Src phosphorylation site on TRPV1 prevented PKC but not PKA-mediated sensitization of TRPV1 in vitro. We found that intraplantar injection of the PKC activator phorbol 12-myristate 13-acetate (PMA) or bradykinin produces thermal hypersensitivity that can be attenuated by pharmacological inhibition of Src. Additionally, complete Freund's Adjuvant (CFA)-induced inflammatory hypersensitivity could also be attenuated by local Src kinase inhibition. CONCLUSIONS Our data demonstrate that Src phosphorylation is critical for PKC-mediated sensitization of TRPV1. Further, in a model of inflammatory pain, CFA, Src kinase inhibition could reduce thermal hypersensitivity. Targeting of Src kinase may have analgesic benefits in inflammatory pain conditions. SIGNIFICANCE Src kinase-mediated phosphorylation of TRPV1 is a critical regulator of the PKC-induced sensitization induced by multiple inflammatory mediators. This suggest a new regulatory mechanism governing TRPV1 function and a potential therapeutic target for inflammatory type pain, including cancer pain where Src antagonists are currently utilized.
Collapse
Affiliation(s)
- Gabriella L. Robilotto
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida
| | - Durga P. Mohapatra
- Department of Pharmacology, The University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA, 52242
- Anesthesia, The University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA, 52242
| | - Andrew J. Shepherd
- Department of Pharmacology, The University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA, 52242
- Department of Symptom Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Aaron D. Mickle
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida
- Department of Pharmacology, The University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA, 52242
- J. Crayton Pruitt Family Department of Biomedical Engineering, College of Engineering, University of Florida
- Department of Neuroscience, College of Medicine, University of Florida
| |
Collapse
|
17
|
Inflammatory-associated apoptotic markers: are they the culprit to rheumatoid arthritis pain? Mol Biol Rep 2022; 49:10077-10090. [PMID: 35699858 DOI: 10.1007/s11033-022-07591-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 05/11/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a prolonged inflammatory disease resulting from autoimmune reactions that leads to local and systemic bone erosion, joint defects and functional impairment. Although the inflammation is subsided through the prescription of anti-inflammatory therapeutics, the patients persistently complained of sleepless nights due to flare pain. This indicates the possible contribution of other pathways besides inflammation in leading to RA pain. This review aims to uncover the roles and involvement of several inflammatory-associated apoptotic markers in facilitating pain transmission and processing during the pathogenesis of RA. MATERIALS AND METHODS This narrative review focused on the reports from the previous literature based on the search string of "apoptotic marker AND inflammation AND 'chronic pain' OR 'neuropathic pain' and apoptosis AND 'rheumatoid arthritis' OR arthritis from the databases including Science Direct and Scopus, considering the exclusion criteria of the published abstracts, proceedings or articles on other neuropathic pain types such as painful bowel syndrom, insterstitial cystitis, fibrosis and so on. RESULTS Several studies in the literature demonstrate a close association between imbalanced apoptotic regulations and an increased number of synovial fibroblasts and inflammatory cells in RA. Cell death or specific cell survival has been linked with increased central hypersensitivity in various types of chronic and neuropathic pain. CONCLUSION The RA-related flare pain is possibly contributed by the abnormal regulation of apoptosis through several inflammatory-related pathways, and further studies need to modulate these pathways for the putative anti-nociceptive benefits.
Collapse
|
18
|
Hernández-Cruz EY, Silva-Islas CA, Maldonado PD, Pedraza-Chaverri J, Carballo-Villalobos AI. The antinociceptive effect of garlic, garlic preparations, and derivative compounds. Eur J Pain 2022; 26:947-964. [PMID: 35263014 DOI: 10.1002/ejp.1935] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 02/02/2022] [Accepted: 03/06/2022] [Indexed: 11/08/2022]
Abstract
The antinociceptive effects of garlic have shown promise in treating different chronic diseases in humans, such as knee osteoarthritis, rheumatoid arthritis, and peripheral arterial occlusive disease stage II. The most common garlic products are garlic powder (dried garlic), steam distilled garlic oils, garlic oil macerate, and aged garlic extract. These commercial products contain organosulfur compounds (OSC) that have been extensively evaluated in preclinical models and some clinical assays to treat different diseases against pain. In this review, we describe the importance of some bioactive compounds found in garlic and its role in treating pain. A systematic search of the literature in Dimensions, PubMed, Scopus, Web of Science was performed. Terms and preselected keywords relating to garlic, its derivates and organusulfur compunds in pain, were used to perform a systematic literature search. Two independent reviewers screened papers for inclusion and assessed the methodological quality. The antinociceptive activity of garlic and its OSC is related to its antioxidant and anti-inflammatory properties, which may be explained by the ability to block the synthesis of PGs, pro-inflammatory cytokines and interferon-γ, by the reduction COX- 2 activity and by increases the levels of anti-inflammatory cytokines. Besides, garlic extract is an activator of TRPA1 and TRPV1, where the principal responsible for this activation are OSC. The relationship between these pathways allows a better understanding how garlic and its derivates could be carrying out its pharmacological action over the management of acute and chronic pain and provide a base by further investigations.
Collapse
Affiliation(s)
- Estefani Yaquelin Hernández-Cruz
- Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México (UNAM), CDMX, 04510, México.,Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México (UNAM), Ciudad Universitaria, CDMX, 04510, México
| | - Carlos Alfredo Silva-Islas
- Laboratorio de Patología Vascular Cerebral, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, CDMX, 14269, México
| | - Perla D Maldonado
- Laboratorio de Patología Vascular Cerebral, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, CDMX, 14269, México
| | - José Pedraza-Chaverri
- Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México (UNAM), CDMX, 04510, México
| | - Azucena Ibeth Carballo-Villalobos
- Departamento de Química Inorgánica y Nuclear, Facultad de Química, Universidad Nacional Autónoma de México (UNAM), CDMX, 04510, México
| |
Collapse
|
19
|
Inferiority complex: why do sensory ion channels multimerize? Biochem Soc Trans 2022; 50:213-222. [PMID: 35166323 PMCID: PMC9022975 DOI: 10.1042/bst20211002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 11/17/2022]
Abstract
Peripheral somatosensory nerves are equipped with versatile molecular sensors which respond to acute changes in the physical environment. Most of these sensors are ion channels that, when activated, depolarize the sensory nerve terminal causing it to generate action potentials, which is the first step in generation of most somatic sensations, including pain. The activation and inactivation of sensory ion channels is tightly regulated and modulated by a variety of mechanisms. Amongst such mechanisms is the regulation of sensory ion channel activity via direct molecular interactions with other proteins in multi-protein complexes at the plasma membrane of sensory nerve terminals. In this brief review, we will consider several examples of such complexes formed around a prototypic sensory receptor, transient receptor potential vanilloid type 1 (TRPV1). We will also discuss some inherent conceptual difficulties arising from the multitude of reported complexes.
Collapse
|
20
|
Yang Y, Wang Y, Zhang Z, Wang S, Li Z. WSF-CT-11, a Sesquiterpene Derivative, Activates AMP-Activated Protein Kinase with Anti-diabetic Effects in 3T3-L1 Adipocytes. ACS OMEGA 2021; 6:31272-31281. [PMID: 34841171 PMCID: PMC8613856 DOI: 10.1021/acsomega.1c05061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 10/18/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND AMP-activated protein kinase (AMPK) is a therapeutic target against type II diabetes (T2D). Synthetic sesquiterpene derivatives were investigated to identify novel AMPK activators as anti-diabetic drugs because the leading drugs like metformin and thiazolidinediones (TZDs) activate AMPK by inhibiting the synthesis of adenosine 5'-triphosphate and thus are associated with some side effects. RESULTS We identified WSF-CT-11 as an AMPK activator in HEK293T cells and found that WSF-CT-11 activates AMPK by the activation of transient receptor potential vanilloid type 1 (TRPV1), a Ca2+-permeable cation channel. The increased Ca2+ influx then activates phosphoinositide 3-kinase (PI3K), protein kinase B (PKB/Akt), and Ca2+/calmodulin-dependent protein kinase II (CaMKII), which in turn phosphorylates TRPV1 and facilitates the formation of a TRPV1/Akt/CaMKII/AMPK complex. This complex might be important for the regulation of AMPK activity. In 3T3-L1 adipocytes, WSF-CT-11-induced AMPK activation has three anti-diabetic effects. It promotes the assembly of high-molecular-weight adiponectin, which has stronger insulin-sensitizing activity than other multimers. It improves translocation of the glucose transporter type 4, increases glucose uptake, and induces the inhibitory phosphorylation of peroxisome proliferator-activated receptor γ and thus suppresses adipogenesis. CONCLUSION WSF-CT-11 is a novel AMPK activator and potential drug against T2D without the side effects of metformin and TZDs.
Collapse
Affiliation(s)
- Yang Yang
- MOE
Key Laboratory of Bioinoformatics, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yunyun Wang
- College
of Chemical Engineering, Nanjing Forestry
University, Jiangsu 210037, China
| | - Zhijie Zhang
- Institute
of Chinese Materia Medica, China Academy
of Chinese Medical Sciences, Beijing 100700, China
| | - Shifa Wang
- College
of Chemical Engineering, Nanjing Forestry
University, Jiangsu 210037, China
| | - Zhen Li
- MOE
Key Laboratory of Bioinoformatics, School of Life Sciences, Tsinghua University, Beijing 100084, China
| |
Collapse
|
21
|
Shibata M, Kayama Y, Takizawa T, Ibata K, Shimizu T, Yuzaki M, Suzuki N, Nakahara J. Resilience to capsaicin-induced mitochondrial damage in trigeminal ganglion neurons. Mol Pain 2021; 16:1744806920960856. [PMID: 32985330 PMCID: PMC7536481 DOI: 10.1177/1744806920960856] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Capsaicin is an agonist of transient receptor potential cation channel subfamily V member 1 (TRPV1). Strong TRPV1 stimulation with capsaicin causes mitochondrial damage in primary sensory neurons. However, the effect of repetitive and moderate exposure to capsaicin on the integrity of neuronal mitochondria remains largely unknown. Our electron microscopic analysis revealed that repetitive stimulation of the facial skin of mice with 10 mM capsaicin induced short-term damage to the mitochondria in small-sized trigeminal ganglion neurons. Further, capsaicin-treated mice exhibited decreased sensitivity to noxious heat stimulation, indicating TRPV1 dysfunction, in parallel with the mitochondrial damage in the trigeminal ganglion neurons. To analyze the capsaicin-induced mitochondrial damage and its relevant cellular events in detail, we performed cell-based assays using TRPV1-expressing PC12 cells. Dose-dependent capsaicin-mediated mitochondrial toxicity was observed. High doses of capsaicin caused rapid destruction of mitochondrial internal structure, while low doses induced mitochondrial swelling. Further, capsaicin induced a dose-dependent loss of mitochondria and autophagy-mediated degradation of mitochondria (mitophagy). Concomitantly, transcriptional upregulation of mitochondrial proteins, cytochrome c oxidase subunit IV, Mic60/Mitofilin, and voltage-dependent anion channel 1 was observed, which implied induction of mitochondrial biogenesis to compensate for the loss of mitochondria. Collectively, although trigeminal ganglion neurons transiently exhibit mitochondrial damage and TRPV1 dysfunction following moderate capsaicin exposure, they appear to be resilient to such a challenge. Our in vitro data show a dose-response relationship in capsaicin-mediated mitochondrial toxicity. We postulate that induction of mitophagy and mitochondrial biogenesis in response to capsaicin stimulation play important roles in repairing the damaged mitochondrial system.
Collapse
Affiliation(s)
- Mamoru Shibata
- Department of Neurology, Keio University School of Medicine, Japan
| | - Yohei Kayama
- Department of Neurology, Keio University School of Medicine, Japan
| | - Tsubasa Takizawa
- Department of Neurology, Keio University School of Medicine, Japan
| | - Keiji Ibata
- Department of Physiology, Keio University School of Medicine, Japan.,Department of Physiology, St. Marianna Medical University, Japan
| | | | - Michisuke Yuzaki
- Department of Physiology, Keio University School of Medicine, Japan
| | - Norihiro Suzuki
- Department of Neurology, Keio University School of Medicine, Japan
| | - Jin Nakahara
- Department of Neurology, Keio University School of Medicine, Japan
| |
Collapse
|
22
|
Barragan-Iglesias P, Kunder N, Wanghzou A, Black B, Ray PR, Lou TF, de la Peña JB, Atmaramani R, Shukla T, Pancrazio JJ, Price TJ, Campbell ZT. A peptide encoded within a 5' untranslated region promotes pain sensitization in mice. Pain 2021; 162:1864-1875. [PMID: 33449506 PMCID: PMC8119312 DOI: 10.1097/j.pain.0000000000002191] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 01/04/2021] [Indexed: 12/23/2022]
Abstract
ABSTRACT Translational regulation permeates neuronal function. Nociceptors are sensory neurons responsible for the detection of harmful stimuli. Changes in their activity, termed plasticity, are intimately linked to the persistence of pain. Although inhibitors of protein synthesis robustly attenuate pain-associated behavior, the underlying targets that support plasticity are largely unknown. Here, we examine the contribution of protein synthesis in regions of RNA annotated as noncoding. Based on analyses of previously reported ribosome profiling data, we provide evidence for widespread translation in noncoding transcripts and regulatory regions of mRNAs. We identify an increase in ribosome occupancy in the 5' untranslated regions of the calcitonin gene-related peptide (CGRP/Calca). We validate the existence of an upstream open reading frame (uORF) using a series of reporter assays. Fusion of the uORF to a luciferase reporter revealed active translation in dorsal root ganglion neurons after nucleofection. Injection of the peptide corresponding to the calcitonin gene-related peptide-encoded uORF resulted in pain-associated behavioral responses in vivo and nociceptor sensitization in vitro. An inhibitor of heterotrimeric G protein signaling blocks both effects. Collectively, the data suggest pervasive translation in regions of the transcriptome annotated as noncoding in dorsal root ganglion neurons and identify a specific uORF-encoded peptide that promotes pain sensitization through GPCR signaling.
Collapse
Affiliation(s)
- Paulino Barragan-Iglesias
- University of Texas at Dallas, School of Behavioral and
Brain Sciences, Richardson, TX, 75080, USA
- Department of Physiology and Pharmacology, Center for Basic
Sciences, Autonomous University of Aguascalientes, Aguascalientes, 20130,
Mexico
| | - Nikesh Kunder
- University of Texas at Dallas, Department of Biological
Sciences, Richardson, TX, 75080, USA
| | - Andi Wanghzou
- University of Texas at Dallas, School of Behavioral and
Brain Sciences, Richardson, TX, 75080, USA
| | - Bryan Black
- University of Texas at Dallas, Department of
Bioengineering, Richardson, TX, 75080, USA
| | - Pradipta R. Ray
- University of Texas at Dallas, School of Behavioral and
Brain Sciences, Richardson, TX, 75080, USA
| | - Tzu-Fang Lou
- University of Texas at Dallas, Department of Biological
Sciences, Richardson, TX, 75080, USA
| | - June Bryan de la Peña
- University of Texas at Dallas, Department of Biological
Sciences, Richardson, TX, 75080, USA
| | - Rahul Atmaramani
- University of Texas at Dallas, Department of
Bioengineering, Richardson, TX, 75080, USA
| | - Tarjani Shukla
- University of Texas at Dallas, Department of Biological
Sciences, Richardson, TX, 75080, USA
| | - Joseph J. Pancrazio
- University of Texas at Dallas, Department of
Bioengineering, Richardson, TX, 75080, USA
- Center for Advanced Pain Studies, University of Texas at
Dallas, Richardson, TX, 75080, USA
| | - Theodore J. Price
- University of Texas at Dallas, School of Behavioral and
Brain Sciences, Richardson, TX, 75080, USA
- Center for Advanced Pain Studies, University of Texas at
Dallas, Richardson, TX, 75080, USA
| | - Zachary T. Campbell
- University of Texas at Dallas, Department of Biological
Sciences, Richardson, TX, 75080, USA
- Center for Advanced Pain Studies, University of Texas at
Dallas, Richardson, TX, 75080, USA
| |
Collapse
|
23
|
Shibata M, Tang C. Implications of Transient Receptor Potential Cation Channels in Migraine Pathophysiology. Neurosci Bull 2021; 37:103-116. [PMID: 32870468 PMCID: PMC7811976 DOI: 10.1007/s12264-020-00569-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 05/11/2020] [Indexed: 12/19/2022] Open
Abstract
Migraine is a common and debilitating headache disorder. Although its pathogenesis remains elusive, abnormal trigeminal and central nervous system activity is likely to play an important role. Transient receptor potential (TRP) channels, which transduce noxious stimuli into pain signals, are expressed in trigeminal ganglion neurons and brain regions closely associated with the pathophysiology of migraine. In the trigeminal ganglion, TRP channels co-localize with calcitonin gene-related peptide, a neuropeptide crucially implicated in migraine pathophysiology. Many preclinical and clinical data support the roles of TRP channels in migraine. In particular, activation of TRP cation channel V1 has been shown to regulate calcitonin gene-related peptide release from trigeminal nerves. Intriguingly, several effective anti-migraine therapies, including botulinum neurotoxin type A, affect the functions of TRP cation channels. Here, we discuss currently available data regarding the roles of major TRP cation channels in the pathophysiology of migraine and the therapeutic applicability thereof.
Collapse
Affiliation(s)
- Mamoru Shibata
- Department of Neurology, Keio University School of Medicine, Tokyo, 160-8582, Japan.
- Department of Neurology, Tokyo Dental College Ichikawa General Hospital, Chiba, 272-8513, Japan.
| | - Chunhua Tang
- Department of Neurology, Keio University School of Medicine, Tokyo, 160-8582, Japan
- Department of Neurology and Center for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| |
Collapse
|
24
|
Zhao X, Xia B, Cheng J, Zhu MX, Li Y. PKCε SUMOylation Is Required for Mediating the Nociceptive Signaling of Inflammatory Pain. Cell Rep 2020; 33:108191. [DOI: 10.1016/j.celrep.2020.108191] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 07/27/2020] [Accepted: 09/02/2020] [Indexed: 12/20/2022] Open
|
25
|
Deletion of a Neuronal Drp1 Activator Protects against Cerebral Ischemia. J Neurosci 2020; 40:3119-3129. [PMID: 32144179 DOI: 10.1523/jneurosci.1926-19.2020] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 02/25/2020] [Accepted: 02/26/2020] [Indexed: 11/21/2022] Open
Abstract
Mitochondrial fission catalyzed by dynamin-related protein 1 (Drp1) is necessary for mitochondrial biogenesis and maintenance of healthy mitochondria. However, excessive fission has been associated with multiple neurodegenerative disorders, and we recently reported that mice with smaller mitochondria are sensitized to ischemic stroke injury. Although pharmacological Drp1 inhibition has been put forward as neuroprotective, the specificity and mechanism of the inhibitor used is controversial. Here, we provide genetic evidence that Drp1 inhibition is neuroprotective. Drp1 is activated by dephosphorylation of an inhibitory phosphorylation site, Ser637. We identify Bβ2, a mitochondria-localized protein phosphatase 2A (PP2A) regulatory subunit, as a neuron-specific Drp1 activator in vivo Bβ2 KO mice of both sexes display elongated mitochondria in neurons and are protected from cerebral ischemic injury. Functionally, deletion of Bβ2 and maintained Drp1 Ser637 phosphorylation improved mitochondrial respiratory capacity, Ca2+ homeostasis, and attenuated superoxide production in response to ischemia and excitotoxicity in vitro and ex vivo Last, deletion of Bβ2 rescued excessive stroke damage associated with dephosphorylation of Drp1 S637 and mitochondrial fission. These results indicate that the state of mitochondrial connectivity and PP2A/Bβ2-mediated dephosphorylation of Drp1 play a critical role in determining the severity of cerebral ischemic injury. Therefore, Bβ2 may represent a target for prophylactic neuroprotective therapy in populations at high risk of stroke.SIGNIFICANCE STATEMENT With recent advances in clinical practice including mechanical thrombectomy up to 24 h after the ischemic event, there is resurgent interest in neuroprotective stroke therapies. In this study, we demonstrate reduced stroke damage in the brain of mice lacking the Bβ2 regulatory subunit of protein phosphatase 2A, which we have shown previously acts as a positive regulator of the mitochondrial fission enzyme dynamin-related protein 1 (Drp1). Importantly, we provide evidence that deletion of Bβ2 can rescue excessive ischemic damage in mice lacking the mitochondrial PKA scaffold AKAP1, apparently via opposing effects on Drp1 S637 phosphorylation. These results highlight reversible phosphorylation in bidirectional regulation of Drp1 activity and identify Bβ2 as a potential pharmacological target to protect the brain from stroke injury.
Collapse
|
26
|
Gonçalves dos Santos G, Delay L, Yaksh TL, Corr M. Neuraxial Cytokines in Pain States. Front Immunol 2020; 10:3061. [PMID: 32047493 PMCID: PMC6997465 DOI: 10.3389/fimmu.2019.03061] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 12/16/2019] [Indexed: 12/11/2022] Open
Abstract
A high-intensity potentially tissue-injuring stimulus generates a homotopic response to escape the stimulus and is associated with an affective phenotype considered to represent pain. In the face of tissue or nerve injury, the afferent encoding systems display robust changes in the input-output function, leading to an ongoing sensation reported as painful and sensitization of the nociceptors such that an enhanced pain state is reported for a given somatic or visceral stimulus. Our understanding of the mechanisms underlying this non-linear processing of nociceptive stimuli has led to our appreciation of the role played by the functional interactions of neural and immune signaling systems in pain phenotypes. In pathological states, neural systems interact with the immune system through the actions of a variety of soluble mediators, including cytokines. Cytokines are recognized as important mediators of inflammatory and neuropathic pain, supporting system sensitization and the development of a persistent pathologic pain. Cytokines can induce a facilitation of nociceptive processing at all levels of the neuraxis including supraspinal centers where nociceptive input evokes an affective component of the pain state. We review here several key proinflammatory and anti-inflammatory cytokines/chemokines and explore their underlying actions at four levels of neuronal organization: (1) peripheral nociceptor termini; (2) dorsal root ganglia; (3) spinal cord; and (4) supraspinal areas. Thus, current thinking suggests that cytokines by this action throughout the neuraxis play key roles in the induction of pain and the maintenance of the facilitated states of pain behavior generated by tissue injury/inflammation and nerve injury.
Collapse
Affiliation(s)
| | - Lauriane Delay
- Department of Anesthesiology, University of California, San Diego, La Jolla, CA, United States
| | - Tony L. Yaksh
- Department of Anesthesiology, University of California, San Diego, La Jolla, CA, United States
| | - Maripat Corr
- Division of Rheumatology, Allergy and Immunology, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
27
|
Jang Y, Kim M, Hwang SW. Molecular mechanisms underlying the actions of arachidonic acid-derived prostaglandins on peripheral nociception. J Neuroinflammation 2020; 17:30. [PMID: 31969159 PMCID: PMC6975075 DOI: 10.1186/s12974-020-1703-1] [Citation(s) in RCA: 148] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 01/06/2020] [Indexed: 12/30/2022] Open
Abstract
Arachidonic acid-derived prostaglandins not only contribute to the development of inflammation as intercellular pro-inflammatory mediators, but also promote the excitability of the peripheral somatosensory system, contributing to pain exacerbation. Peripheral tissues undergo many forms of diseases that are frequently accompanied by inflammation. The somatosensory nerves innervating the inflamed areas experience heightened excitability and generate and transmit pain signals. Extensive studies have been carried out to elucidate how prostaglandins play their roles for such signaling at the cellular and molecular levels. Here, we briefly summarize the roles of arachidonic acid-derived prostaglandins, focusing on four prostaglandins and one thromboxane, particularly in terms of their actions on afferent nociceptors. We discuss the biosynthesis of the prostaglandins, their specific action sites, the pathological alteration of the expression levels of related proteins, the neuronal outcomes of receptor stimulation, their correlation with behavioral nociception, and the pharmacological efficacy of their regulators. This overview will help to a better understanding of the pathological roles that prostaglandins play in the somatosensory system and to a finding of critical molecular contributors to normalizing pain.
Collapse
Affiliation(s)
- Yongwoo Jang
- Department of Psychiatry and Program in Neuroscience, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA.,Department of Biomedical Engineering, Hanyang University, Seoul, 04763, South Korea
| | - Minseok Kim
- Department of Biomedical Sciences, Korea University, Seoul, 02841, South Korea
| | - Sun Wook Hwang
- Department of Biomedical Sciences, Korea University, Seoul, 02841, South Korea. .,Department of Physiology, College of Medicine, Korea University, Seoul, 02841, South Korea.
| |
Collapse
|
28
|
Christie S, O'Rielly R, Li H, Wittert GA, Page AJ. Biphasic effects of methanandamide on murine gastric vagal afferent mechanosensitivity. J Physiol 2019; 598:139-150. [PMID: 31642519 DOI: 10.1113/jp278696] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 10/20/2019] [Indexed: 12/21/2022] Open
Abstract
KEY POINTS The fine control of food intake is important for the maintenance of a healthy metabolic state. Gastric vagal afferents (GVAs) are involved in the peripheral regulation of food intake via signalling the degree of distension of the stomach which ultimately leads to feelings of fullness and satiety. This study provides evidence that endocannabinoids such as anandamide are capable of regulating GVA sensitivity in a concentration-dependent biphasic manner. This biphasic effect is dependent upon interactions between the CB1, TRPV1 and GHSR receptors. These data have important implications for the peripheral control of food intake. ABSTRACT Gastric vagal afferents (GVAs) signal to the hindbrain resulting in satiety. Endocannabinoids are endogenous ligands of cannabinoid 1 receptor (CB1) and transient receptor potential vanilloid-1 (TRPV1) channels. The endocannabinoid anandamide (AEA) is expressed in the stomach, and its receptor CB1 is expressed in ghrelin-positive gastric mucosal cells. Further, TRPV1, CB1 and growth hormone secretagogue receptor (ghrelin receptor, GHSR) are expressed in subpopulations of GVA neurons. This study aimed to determine the interaction between TRPV1, CB1, GHSR and endocannabinoids in the modulation of GVA signalling. An in vitro electrophysiology preparation was used to assess GVA mechanosensitivity in male C57BL/6 mice. Effects of methanandamide (mAEA; 1-100 nm), on GVA responses to stretch were determined in the absence and presence of antagonists of CB1, TRPV1, GHSR, protein kinase-A (PKA), protein kinase-C (PKC) and G-protein subunits Gαi/o , or Gαq . Low doses (1-10 nm) of mAEA reduced GVA responses to 3 g stretch, whereas high doses (30-100 nm) increased the response. The inhibitory and excitatory effects of mAEA (1-100 nm) were reduced/lost in the presence of a CB1 and TRPV1 antagonist. PKA, Gαi/o or GHSR antagonists prevented the inhibitory effect of mAEA on GVA mechanosensitivity. Conversely, in the presence of a PKC or Gαq antagonist the excitatory effect of mAEA was reduced or lost, respectively. Activation of CB1, by mAEA, can activate or inhibit TRPV1 to increase or decrease GVA responses to stretch, depending on the pathway activated. These interactions could play an important role in the fine control of food intake.
Collapse
Affiliation(s)
- Stewart Christie
- Vagal Afferent Research Group, Centre for Nutrition and Gastrointestinal Disease, Adelaide Medical School, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Rebecca O'Rielly
- Vagal Afferent Research Group, Centre for Nutrition and Gastrointestinal Disease, Adelaide Medical School, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Hui Li
- Vagal Afferent Research Group, Centre for Nutrition and Gastrointestinal Disease, Adelaide Medical School, University of Adelaide, Adelaide, SA, 5005, Australia.,Lifelong Health, South Australian Health and Medical Research Institute, Adelaide, SA, 5000, Australia
| | - Gary A Wittert
- Vagal Afferent Research Group, Centre for Nutrition and Gastrointestinal Disease, Adelaide Medical School, University of Adelaide, Adelaide, SA, 5005, Australia.,Lifelong Health, South Australian Health and Medical Research Institute, Adelaide, SA, 5000, Australia
| | - Amanda J Page
- Vagal Afferent Research Group, Centre for Nutrition and Gastrointestinal Disease, Adelaide Medical School, University of Adelaide, Adelaide, SA, 5005, Australia.,Lifelong Health, South Australian Health and Medical Research Institute, Adelaide, SA, 5000, Australia
| |
Collapse
|
29
|
Zhou YM, Wu L, Wei S, Jin Y, Liu TT, Qiu CY, Hu WP. Enhancement of acid-sensing ion channel activity by prostaglandin E2 in rat dorsal root ganglion neurons. Brain Res 2019; 1724:146442. [PMID: 31513790 DOI: 10.1016/j.brainres.2019.146442] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 07/12/2019] [Accepted: 09/07/2019] [Indexed: 11/16/2022]
Abstract
Prostaglandin E2 (PGE2) and proton are typical inflammatory mediators. They play a major role in pain processing and hypersensitivity through activating their cognate receptors expressed in terminals of nociceptive sensory neurons. However, it remains unclear whether there is an interaction between PGE2 receptors and proton-activated acid-sensing ion channels (ASICs). Herein, we show that PGE2 enhanced the functional activity of ASICs in rat dorsal root ganglion (DRG) neurons through EP1 and EP4 receptors. In the present study, PGE2 concentration-dependently increased ASIC currents in DRG neurons. It shifted the proton concentration-response curve upwards, without change in the apparent affinity of proton for ASICs. Moreover, PGE2 enhancement of ASIC currents was partially blocked by EP1 or EP4 receptor antagonist. PGE2 failed to enhance ASIC currents when simultaneous blockade of both EP1 and EP4 receptors. PGE2 enhancement was partially suppressed after inhibition of intracellular PKC or PKA signaling, and completely disappeared after concurrent blockade of both PKC and PKA signaling. PGE2 increased significantly the expression levels of p-PKCε and p-PKA in DRG cells. PGE2 also enhanced proton-evoked action potentials in rat DRG neurons. Finally, peripherally administration of PGE2 dose-dependently exacerbated acid-induced nocifensive behaviors in rats through EP1 and EP4 receptors. Our results indicate that PGE2 enhanced the electrophysiological activity of ASICs in DRG neurons and contributed to acidosis-evoked pain, which revealed a novel peripheral mechanism underlying PGE2 involvement in hyperalgesia by sensitizing ASICs in primary sensory neurons.
Collapse
Affiliation(s)
- Yi-Mei Zhou
- Research Center of Basic Medical Sciences, School of Basic Medical Sciences, Hubei University of Science and Technology, 88 Xianning Road, Xianning 437100, Hubei, PR China; Department of Pharmacology, Hubei University of Science and Technology, 88 Xianning Road, Xianning 437100, Hubei, PR China
| | - Lei Wu
- Research Center of Basic Medical Sciences, School of Basic Medical Sciences, Hubei University of Science and Technology, 88 Xianning Road, Xianning 437100, Hubei, PR China
| | - Shuang Wei
- Research Center of Basic Medical Sciences, School of Basic Medical Sciences, Hubei University of Science and Technology, 88 Xianning Road, Xianning 437100, Hubei, PR China; Department of Pharmacology, Hubei University of Science and Technology, 88 Xianning Road, Xianning 437100, Hubei, PR China
| | - Ying Jin
- Research Center of Basic Medical Sciences, School of Basic Medical Sciences, Hubei University of Science and Technology, 88 Xianning Road, Xianning 437100, Hubei, PR China; Department of Pharmacology, Hubei University of Science and Technology, 88 Xianning Road, Xianning 437100, Hubei, PR China
| | - Ting-Ting Liu
- Research Center of Basic Medical Sciences, School of Basic Medical Sciences, Hubei University of Science and Technology, 88 Xianning Road, Xianning 437100, Hubei, PR China; Department of Physiology, School of Basic Medical Sciences, Hubei University of Science and Technology, 88 Xianning Road, Xianning 437100, Hubei, PR China
| | - Chun-Yu Qiu
- Department of Pharmacology, Hubei University of Science and Technology, 88 Xianning Road, Xianning 437100, Hubei, PR China
| | - Wang-Ping Hu
- Research Center of Basic Medical Sciences, School of Basic Medical Sciences, Hubei University of Science and Technology, 88 Xianning Road, Xianning 437100, Hubei, PR China; Department of Physiology, School of Basic Medical Sciences, Hubei University of Science and Technology, 88 Xianning Road, Xianning 437100, Hubei, PR China.
| |
Collapse
|
30
|
Fu W, Nelson TS, Santos DF, Doolen S, Gutierrez JJ, Ye N, Zhou J, Taylor B. An NPY Y1 receptor antagonist unmasks latent sensitization and reveals the contribution of protein kinase A and Epac to chronic inflammatory pain. Pain 2019; 160:1754-1765. [PMID: 31335645 PMCID: PMC6903783 DOI: 10.1097/j.pain.0000000000001557] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Peripheral inflammation produces a long-lasting latent sensitization of spinal nociceptive neurons, that is, masked by tonic inhibitory controls. We explored mechanisms of latent sensitization with an established four-step approach: (1) induction of inflammation; (2) allow pain hypersensitivity to resolve; (3) interrogate latent sensitization with a channel blocker, mutant mouse, or receptor antagonist; and (4) disrupt compensatory inhibition with a receptor antagonist so as to reinstate pain hypersensitivity. We found that the neuropeptide Y Y1 receptor antagonist BIBO3304 reinstated pain hypersensitivity, indicative of an unmasking of latent sensitization. BIBO3304-evoked reinstatement was not observed in AC1 knockout mice and was prevented with intrathecal co-administration of a pharmacological blocker to the N-methyl-D-aspartate receptor (NMDAR), adenylyl cyclase type 1 (AC1), protein kinase A (PKA), transient receptor potential cation channel A1 (TRPA1), channel V1 (TRPV1), or exchange protein activated by cAMP (Epac1 or Epac2). A PKA activator evoked both pain reinstatement and touch-evoked pERK expression in dorsal horn; the former was prevented with intrathecal co-administration of a TRPA1 or TRPV1 blocker. An Epac activator also evoked pain reinstatement and pERK expression. We conclude that PKA and Epac are sufficient to maintain long-lasting latent sensitization of dorsal horn neurons that is kept in remission by the NPY-Y1 receptor system. Furthermore, we have identified and characterized 2 novel molecular signaling pathways in the dorsal horn that drive latent sensitization in the setting of chronic inflammatory pain: NMDAR→AC1→PKA→TRPA1/V1 and NMDAR→AC1→Epac1/2. New treatments for chronic inflammatory pain might either increase endogenous NPY analgesia or inhibit AC1, PKA, or Epac.
Collapse
Affiliation(s)
- Weisi Fu
- Department of Physiology, University of Kentucky Medical Center, Lexington KY, USA
| | - Tyler S. Nelson
- Department of Anesthesiology, Pittsburgh Center for Pain Research, and the Opiate Research Center at the University of Pittsburgh, University of Pittsburgh, Pittsburgh, PA USA
- Center for Neuroscience at the University of Pittsburgh, Pittsburgh, PA USA
- Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA USA
| | - Diogo F. Santos
- Department of Anesthesiology, Pittsburgh Center for Pain Research, and the Opiate Research Center at the University of Pittsburgh, University of Pittsburgh, Pittsburgh, PA USA
| | - Suzanne Doolen
- Department of Anesthesiology, Pittsburgh Center for Pain Research, and the Opiate Research Center at the University of Pittsburgh, University of Pittsburgh, Pittsburgh, PA USA
| | - Javier J.P. Gutierrez
- Department of Anesthesiology, Pittsburgh Center for Pain Research, and the Opiate Research Center at the University of Pittsburgh, University of Pittsburgh, Pittsburgh, PA USA
| | - Na Ye
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Jia Zhou
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Bradley Taylor
- Department of Physiology, University of Kentucky Medical Center, Lexington KY, USA
- Department of Anesthesiology, Pittsburgh Center for Pain Research, and the Opiate Research Center at the University of Pittsburgh, University of Pittsburgh, Pittsburgh, PA USA
- Center for Neuroscience at the University of Pittsburgh, Pittsburgh, PA USA
| |
Collapse
|
31
|
Takayama Y, Derouiche S, Maruyama K, Tominaga M. Emerging Perspectives on Pain Management by Modulation of TRP Channels and ANO1. Int J Mol Sci 2019; 20:E3411. [PMID: 31336748 PMCID: PMC6678529 DOI: 10.3390/ijms20143411] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 07/01/2019] [Accepted: 07/09/2019] [Indexed: 12/27/2022] Open
Abstract
Receptor-type ion channels are critical for detection of noxious stimuli in primary sensory neurons. Transient receptor potential (TRP) channels mediate pain sensations and promote a variety of neuronal signals that elicit secondary neural functions (such as calcitonin gene-related peptide [CGRP] secretion), which are important for physiological functions throughout the body. In this review, we focus on the involvement of TRP channels in sensing acute pain, inflammatory pain, headache, migraine, pain due to fungal infections, and osteo-inflammation. Furthermore, action potentials mediated via interactions between TRP channels and the chloride channel, anoctamin 1 (ANO1), can also generate strong pain sensations in primary sensory neurons. Thus, we also discuss mechanisms that enhance neuronal excitation and are dependent on ANO1, and consider modulation of pain sensation from the perspective of both cation and anion dynamics.
Collapse
Affiliation(s)
- Yasunori Takayama
- Department of Physiology, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa, Tokyo 142-8555, Japan.
| | - Sandra Derouiche
- Thermal Biology group, Exploratory Research Center on Life and Living Systems, National Institutes for Natural Sciences, 5-1 Aza-higashiyama, Myodaiji, Okazaki, Aichi 444-8787, Japan.
| | - Kenta Maruyama
- National Institute for Physiological Sciences, National Institutes for Natural Sciences, 5-1 Aza-higashiyama, Myodaiji, Okazaki, Aichi 444-8787, Japan.
| | - Makoto Tominaga
- Thermal Biology group, Exploratory Research Center on Life and Living Systems, National Institutes for Natural Sciences, 5-1 Aza-higashiyama, Myodaiji, Okazaki, Aichi 444-8787, Japan.
| |
Collapse
|
32
|
Nociceptor Signalling through ion Channel Regulation via GPCRs. Int J Mol Sci 2019; 20:ijms20102488. [PMID: 31137507 PMCID: PMC6566991 DOI: 10.3390/ijms20102488] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 05/08/2019] [Accepted: 05/13/2019] [Indexed: 12/23/2022] Open
Abstract
The prime task of nociceptors is the transformation of noxious stimuli into action potentials that are propagated along the neurites of nociceptive neurons from the periphery to the spinal cord. This function of nociceptors relies on the coordinated operation of a variety of ion channels. In this review, we summarize how members of nine different families of ion channels expressed in sensory neurons contribute to nociception. Furthermore, data on 35 different types of G protein coupled receptors are presented, activation of which controls the gating of the aforementioned ion channels. These receptors are not only targeted by more than 20 separate endogenous modulators, but can also be affected by pharmacotherapeutic agents. Thereby, this review provides information on how ion channel modulation via G protein coupled receptors in nociceptors can be exploited to provide improved analgesic therapy.
Collapse
|
33
|
Jeske NA. Dynamic Opioid Receptor Regulation in the Periphery. Mol Pharmacol 2019; 95:463-467. [PMID: 30723091 PMCID: PMC6442319 DOI: 10.1124/mol.118.114637] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 12/19/2018] [Indexed: 12/26/2022] Open
Abstract
Opioids serve a vital role in the current analgesic array of treatment options. They are useful in acute instances involving severe pain associated with trauma, surgery, and terminal diseases such as cancer. In the past three decades, multiple receptor isoforms and conformations have been reported throughout literature. Most of these studies conducted systemic analyses of opioid receptor function, often generalizing findings from receptor systems in central nervous tissue or exogenously expressing immortalized cell lines as common mechanisms throughout physiology. However, a culmination of innovative experimental data indicates that opioid receptor systems are differentially modulated depending on their anatomic expression profile. Importantly, opioid receptors expressed in the peripheral nervous system undergo regulation uncommon to similar receptors expressed in central nervous system tissues. This distinctive characteristic begs one to question whether peripheral opioid receptors maintain anatomically unique roles, and whether they may serve an analgesic advantage in providing pain relief without promoting addiction.
Collapse
Affiliation(s)
- Nathaniel A Jeske
- Departments of Oral and Maxillofacial Surgery, Pharmacology, and Physiology, Center for Biomedical Neuroscience, University of Texas Health San Antonio, San Antonio, Texas
| |
Collapse
|
34
|
Warwick CA, Shutov LP, Shepherd AJ, Mohapatra DP, Usachev YM. Mechanisms underlying mechanical sensitization induced by complement C5a: the roles of macrophages, TRPV1, and calcitonin gene-related peptide receptors. Pain 2019; 160:702-711. [PMID: 30507785 PMCID: PMC6377341 DOI: 10.1097/j.pain.0000000000001449] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The complement system significantly contributes to the development of inflammatory and neuropathic pain, but the underlying mechanisms are poorly understood. Recently, we identified the signaling pathway responsible for thermal hypersensitivity induced by the complement system component C5a. Here, we examine the mechanisms of another important action of C5a, induction of mechanical hypersensitivity. We found that intraplantar injection of C5a produced a dose-dependent mechanical sensitization and that this effect was blocked by chemogenetic ablation of macrophages in both male and female mice. Knockout of TRPV1 or pretreatment with the TRPV1 antagonists, AMG9810 or 5'-iodoresiniferatoxin (5'-IRTX), significantly reduced C5a-induced mechanical sensitization. Notably, local administration of 5'-IRTX 90 minutes after C5a injection resulted in a slow, but complete, reversal of mechanical sensitization, indicating that TRPV1 activity was required for maintaining C5a-induced mechanical hypersensitivity. This slow reversal suggests that neurogenic inflammation and neuropeptide release may be involved. Indeed, pretreatment with a calcitonin gene-related peptide (CGRP) receptor antagonist (but not an antagonist of the neurokinin 1 receptor) prevented C5a-induced mechanical sensitization. Furthermore, intraplantar injection of CGRP produced significant mechanical sensitization in both wild-type and TRPV1 knockout mice. Taken together, these findings suggest that C5a produces mechanical sensitization by initiating macrophage-to-sensory-neuron signaling cascade that involves activation of TRPV1 and CGRP receptor as critical steps in this process.
Collapse
Affiliation(s)
- Charles A. Warwick
- Department of Pharmacology and Iowa Neuroscience Institute, University of Iowa, Iowa City, IA 52242
| | - Leonid P. Shutov
- Department of Pharmacology and Iowa Neuroscience Institute, University of Iowa, Iowa City, IA 52242
| | - Andrew J. Shepherd
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO 63110
| | - Durga P. Mohapatra
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO 63110
| | - Yuriy M. Usachev
- Department of Pharmacology and Iowa Neuroscience Institute, University of Iowa, Iowa City, IA 52242
| |
Collapse
|
35
|
Inflammation induces Epac-protein kinase C alpha and epsilon signaling in TRPV1-mediated hyperalgesia. Pain 2019; 159:2383-2393. [PMID: 30015706 DOI: 10.1097/j.pain.0000000000001346] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The exchange proteins activated by cAMP (Epacs) have been shown to play important roles in producing inflammation-induced nociception. Transient receptor potential vanilloid type 1 (TRPV1) is a major receptor processing thermal and chemosensitive nociceptive information. The role of Epacs in modulating the activity of TRPV1 has yet to be determined. Studying the effect of complete Freund adjuvant (CFA)-induced inflammation on capsaicin-activated TRPV1 nociceptive responses in dorsal root ganglia (DRG), we found that CFA produced a large increase in capsaicin-induced responses. The increase was inhibited by Epac1 and Epac2 antagonists. Thus, activation of Epacs is critical in producing enhancement in TRPV1-mediated responses under inflammatory conditions. In addition, the inflammation-induced enhancement of TRPV1 responses was blocked by PKCα and PKCε inhibitors, suggesting the essential roles of these PKCs in enhancing TRPV1 responses. To determine the mechanism underlying the Epac actions on TRPV1, we studied the effects of the Epac activator, 8-(4-chlorophenylthio)-2-O-methyl-cAMP (CPT), on capsaicin-induced nociceptive behavioral responses, capsaicin-activated currents, expression and membrane trafficking of PKC and TRPV1 in DRG. CPT was found to enhance capsaicin-induced nociception and ionic currents. The enhancement was inhibited by PKCα and PKCε inhibitors. In addition, CPT increased the expression of phosphorylated PKCα (pPKCα) and membrane TRPV1 expression in DRG. Studying the colocalization of TRPV1 and pPKCα or pPKCε in DRG slices prepared from CFA-treated rats, we found that pPKCα or pPKCε expressed with TRPV1 in different-sized neurons to exert differential influences on TRPV1 activity. Thus, Epac-PKC signaling is critically important in producing inflammation-induced potentiation of TRPV1 functions.
Collapse
|
36
|
Min H, Cho WH, Lee H, Choi B, Kim YJ, Lee HK, Joo Y, Jung SJ, Choi SY, Lee S, Lee SJ. Association of TRPV1 and TLR4 through the TIR domain potentiates TRPV1 activity by blocking activation-induced desensitization. Mol Pain 2018; 14:1744806918812636. [PMID: 30355052 PMCID: PMC6856976 DOI: 10.1177/1744806918812636] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Background We have previously reported that histamine-induced pruritus was attenuated in
toll-like receptor 4 (TLR4) knockout mice due to decreased transient
receptor potential V1 (TRPV1) sensitivity. Our results implied that TLR4
potentiated TRPV1 activation in sensory neurons; however, the molecular
mechanism has yet to be elucidated. In this study, we investigated the
molecular mechanisms of TLR4-mediated TRPV1 potentiation using
TLR4-deficient sensory neurons and a heterologous expression system. Methods Primary sensory neurons were obtained from wild-type or TLR4 knockout mice,
and HEK293T cells expressing TRPV1 and TLR4 were prepared by transient
transfection. TRPV1 activity was analyzed by calcium imaging,
fluorophotometry, and patch-clamp recording. Subcellular protein
distribution was tested by immunocytochemistry and cell surface
biotinylation assay. Protein interaction was assessed by western blot and
immunoprecipitation assay. Results Direct association between TRPV1 and TLR4 was detected in HEK293T cells upon
heterologous TRPV1 and TLR4 expression. In an immunoprecipitation assay
using TLR4-deletion mutants and soluble toll/interleukin-1 receptor (TIR)
protein, the cytoplasmic TIR domain of TLR4 was required for TLR4-TRPV1
association and TRPV1 potentiation. In TLR4-deficient sensory neurons, the
activation-induced desensitization of TRPV1 increased, accompanied by
enhanced TRPV1 clearance from the cell membrane upon activation compared to
wild-type neurons. In addition, heterologous TLR4 expression inhibited
activation-induced TRPV1 endocytosis and lysosomal degradation in HEK293T
cells. Conclusion Our data show that direct association between TRPV1 and TLR4 through the TIR
domain enhances TRPV1 activity by blocking activation-induced TRPV1
desensitization.
Collapse
Affiliation(s)
- Hyunjung Min
- 1 Department of Neuroscience and Physiology, Dental Research Institute, School of Dentistry, Seoul National University, Republic of Korea
| | - Woo-Hyun Cho
- 1 Department of Neuroscience and Physiology, Dental Research Institute, School of Dentistry, Seoul National University, Republic of Korea
| | - Hyunkyoung Lee
- 1 Department of Neuroscience and Physiology, Dental Research Institute, School of Dentistry, Seoul National University, Republic of Korea
| | - Boomin Choi
- 1 Department of Neuroscience and Physiology, Dental Research Institute, School of Dentistry, Seoul National University, Republic of Korea
| | - Yoon-Jung Kim
- 1 Department of Neuroscience and Physiology, Dental Research Institute, School of Dentistry, Seoul National University, Republic of Korea
| | - Han Kyu Lee
- 2 Department of Biomedical Science, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea
| | - Yeonhee Joo
- 3 Department of Microbiology, School of Systems Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Sung Jun Jung
- 2 Department of Biomedical Science, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea
| | - Se-Young Choi
- 1 Department of Neuroscience and Physiology, Dental Research Institute, School of Dentistry, Seoul National University, Republic of Korea
| | - Soojin Lee
- 3 Department of Microbiology, School of Systems Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Sung Joong Lee
- 1 Department of Neuroscience and Physiology, Dental Research Institute, School of Dentistry, Seoul National University, Republic of Korea
| |
Collapse
|
37
|
Abstract
Abstract Primary sensory neurons are responsible for transmitting sensory information from the peripheral to the central nervous system. Their responses to incoming stimulation become greatly enhanced and prolonged following inflammation, giving rise to exaggerated nociceptive responses and chronic pain. The inflammatory mediator, prostaglandin E2 (PGE2), released from the inflamed tissue surrounding the terminals of sensory neurons contributes to the abnormal pain responses. PGE2 acts on G protein-coupled EP receptors to activate adenylyl cyclase, which catalyzes the conversion of adenosine triphosphate to cyclic adenosine 3′,5′-monophosphate (cAMP). Under normal conditions, cAMP activates primarily protein kinase A. After inflammation, cAMP also activates the exchange proteins activated by cAMP (Epacs) to produce exaggerated PGE2-mediated hyperalgesia. The role of cAMP-Epac signaling in the generation of hypersensitivity is the topic of this review.
Collapse
Affiliation(s)
| | - Yanping Gu
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch Galveston, TX 77555-1069, USA
| |
Collapse
|
38
|
AKAP1 Protects from Cerebral Ischemic Stroke by Inhibiting Drp1-Dependent Mitochondrial Fission. J Neurosci 2018; 38:8233-8242. [PMID: 30093535 PMCID: PMC6146498 DOI: 10.1523/jneurosci.0649-18.2018] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 07/29/2018] [Accepted: 08/01/2018] [Indexed: 12/20/2022] Open
Abstract
Mitochondrial fission and fusion impact numerous cellular functions and neurons are particularly sensitive to perturbations in mitochondrial dynamics. Here we describe that male mice lacking the mitochondrial A-kinase anchoring protein 1 (AKAP1) exhibit increased sensitivity in the transient middle cerebral artery occlusion model of focal ischemia. At the ultrastructural level, AKAP1-/- mice have smaller mitochondria and increased contacts between mitochondria and the endoplasmic reticulum in the brain. Mechanistically, deletion of AKAP1 dysregulates complex II of the electron transport chain, increases superoxide production, and impairs Ca2+ homeostasis in neurons subjected to excitotoxic glutamate. Ca2+ deregulation in neurons lacking AKAP1 can be attributed to loss of inhibitory phosphorylation of the mitochondrial fission enzyme dynamin-related protein 1 (Drp1) at the protein kinase A (PKA) site Ser637. Our results indicate that inhibition of Drp1-dependent mitochondrial fission by the outer mitochondrial AKAP1/PKA complex protects neurons from ischemic stroke by maintaining respiratory chain activity, inhibiting superoxide production, and delaying Ca2+ deregulation. They also provide the first genetic evidence that Drp1 inhibition may be of therapeutic relevance for the treatment of stroke and neurodegeneration.SIGNIFICANCE STATEMENT Previous work suggests that activation of dynamin-related protein 1 (Drp1) and mitochondrial fission contribute to ischemic injury in the brain. However, the specificity and efficacy of the pharmacological Drp1 inhibitor mdivi-1 that was used has now been discredited by several high-profile studies. Our report is timely and highly impactful because it provides the first evidence that genetic disinhibition of Drp1 via knock-out of the mitochondrial protein kinase A (PKA) scaffold AKAP1 exacerbates stroke injury in mice. Mechanistically, we show that electron transport deficiency, increased superoxide production, and Ca2+ overload result from genetic disinhibition of Drp1. In summary, our work settles current controversies regarding the role of mitochondrial fission in neuronal injury, provides mechanisms, and suggests that fission inhibitors hold promise as future therapeutic agents.
Collapse
|
39
|
Local administration of mangiferin prevents experimental inflammatory mechanical hyperalgesia through CINC-1/epinephrine/PKA pathway and TNF-α inhibition. Eur J Pharmacol 2018; 830:87-94. [DOI: 10.1016/j.ejphar.2018.04.030] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 04/24/2018] [Accepted: 04/26/2018] [Indexed: 12/27/2022]
|
40
|
Rivera-Pagán AF, Méndez-González MP, Rivera-Aponte DE, Malpica-Nieves CJ, Melnik-Martínez KV, Zayas-Santiago A, Maldonado-Martínez G, Shuba YM, Skatchkov SN, Eaton MJ. A-Kinase-Anchoring Protein (AKAP150) is expressed in Astrocytes and Upregulated in Response to Ischemia. Neuroscience 2018; 384:54-63. [PMID: 29800717 DOI: 10.1016/j.neuroscience.2018.05.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 05/11/2018] [Accepted: 05/14/2018] [Indexed: 10/16/2022]
Abstract
A-kinase-anchoring proteins, AKAPs, are scaffolding proteins that associate with kinases and phosphatases, and direct them to a specific submembrane site to coordinate signaling events. AKAP150, a rodent ortholog of human AKAP79, has been extensively studied in neurons, but very little is known about the localization and function of AKAP150 in astrocytes, the major cell type in brain. Thus, in this study, we assessed the localization of AKAP150 in astrocytes and elucidated its role during physiological and ischemic conditions. Herein, we demonstrate that AKAP150 is localized in astrocytes and is up-regulated during ischemia both in vitro and in vivo. Knock-down of AKAP150 by RNAi depolarizes the astrocytic membrane potential and substantially reduces by 80% the ability of astrocytes to take up extracellular potassium during ischemic conditions. Therefore, upregulation of AKAP150 during ischemia preserves potassium conductance and the associated hyperpolarized membrane potential of astrocytes; properties of astrocytes needed to maintain extracellular brain homeostasis. Taken together, these data suggest that AKAP150 may play a pivotal role in the neuroprotective mechanism of astrocytes during pathological conditions.
Collapse
Affiliation(s)
- Aixa F Rivera-Pagán
- Department of Biochemistry, Universidad Central del Caribe, Bayamón, PR, United States
| | - Miguel P Méndez-González
- Department of Biochemistry, Universidad Central del Caribe, Bayamón, PR, United States; University of Puerto Rico, Natural Sciences Department, Aguadilla, PR, United States
| | - David E Rivera-Aponte
- Department of Biochemistry, Universidad Central del Caribe, Bayamón, PR, United States
| | | | | | - Astrid Zayas-Santiago
- Department of Pathology and Laboratory Medicine, Universidad Central del Caribe, Bayamón, PR, United States
| | | | - Yaroslav M Shuba
- Bogomoletz Institute of Physiology and International Center of Molecular Physiology of the National Academy of Sciences of Ukraine, Kiev, Ukraine
| | - Serguei N Skatchkov
- Department of Biochemistry, Universidad Central del Caribe, Bayamón, PR, United States; Department of Physiology, Universidad Central del Caribe, Bayamón, PR, United States.
| | - Misty J Eaton
- Department of Biochemistry, Universidad Central del Caribe, Bayamón, PR, United States.
| |
Collapse
|
41
|
Serum response factor mediates nociceptor inflammatory pain plasticity. Pain Rep 2018; 3:e658. [PMID: 29922747 PMCID: PMC5999410 DOI: 10.1097/pr9.0000000000000658] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 04/04/2018] [Accepted: 04/08/2018] [Indexed: 01/09/2023] Open
Abstract
Supplemental Digital Content is Available in the Text. Serum response factor upregulates A-Kinase Anchoring Protein 79/150 expression in afferent sensory neurons through metabotropic glutamate receptor signaling. Introduction: Chronic metabotropic glutamate receptor activation in nociceptive afferents may upregulate A-Kinase Anchoring Protein 150 (AKAP150) expression and/or function. Objectives: To quantify transcriptional changes in AKAP150 expression and/or function after long-term mGluR5 agonist exposure, and identify transcriptional elements responsible. Methods: Dorsal root ganglia (DRG) were dissected from Sprague-Dawley rats and cultured for biochemical analysis of AKAP150 expression after prolonged mGluR5 agonist exposure. Serum response factor (SRF) expression was knocked down through siRNA in cultures to demonstrate significance to AKAP150 upregulation. Serum response factor was also knocked down in vivo through intrathecal injections of specifically targeted oligonucleotides to demonstrate significance to hyperalgesic priming behavior in persistent mechanical hypersensitivity. Results: Serum response factor and AKAP150 are coexpressed in TRPV1(+) DRG neurons in intact DRG. Prolonged mGluR5 agonist exposure increases SRF-dependent transcription and AKAP150 expression in a manner sensitive to protein kinase C inhibition and SRF knock down. Serum response factor in vivo knock down reduces mechanical hyperalgesic priming. Conclusion: Serum response factor transcription plays an important role in transcriptional upregulation of AKAP and hyperalgesic priming behavior, and may contribute to the increased role of AKAP150 in the transition from acute to chronic pain.
Collapse
|
42
|
Frey E, Karney-Grobe S, Krolak T, Milbrandt J, DiAntonio A. TRPV1 Agonist, Capsaicin, Induces Axon Outgrowth after Injury via Ca 2+/PKA Signaling. eNeuro 2018; 5:ENEURO.0095-18.2018. [PMID: 29854941 PMCID: PMC5975717 DOI: 10.1523/eneuro.0095-18.2018] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Revised: 05/15/2018] [Accepted: 05/15/2018] [Indexed: 12/22/2022] Open
Abstract
Preconditioning nerve injuries activate a pro-regenerative program that enhances axon regeneration for most classes of sensory neurons. However, nociceptive sensory neurons and central nervous system neurons regenerate poorly. In hopes of identifying novel mechanisms that promote regeneration, we screened for drugs that mimicked the preconditioning response and identified a nociceptive ligand that activates a preconditioning-like response to promote axon outgrowth. We show that activating the ion channel TRPV1 with capsaicin induces axon outgrowth of cultured dorsal root ganglion (DRG) sensory neurons, and that this effect is blocked in TRPV1 knockout neurons. Regeneration occurs only in NF200-negative nociceptive neurons, consistent with a cell-autonomous mechanism. Moreover, we identify a signaling pathway in which TRPV1 activation leads to calcium influx and protein kinase A (PKA) activation to induce a preconditioning-like response. Finally, capsaicin administration to the mouse sciatic nerve activates a similar preconditioning-like response and induces enhanced axonal outgrowth, indicating that this pathway can be induced in vivo. These findings highlight the use of local ligands to induce regeneration and suggest that it may be possible to target selective neuronal populations for repair, including cell types that often fail to regenerate.
Collapse
Affiliation(s)
- Erin Frey
- Department of Developmental Biology, Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Scott Karney-Grobe
- Department of Developmental Biology, Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Trevor Krolak
- Department of Developmental Biology, Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jeff Milbrandt
- Department of Genetics, Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Aaron DiAntonio
- Department of Developmental Biology, Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
43
|
Kullmann FA, Chang HH, Gauthier C, McDonnell BM, Yeh JC, Clayton DR, Kanai AJ, de Groat WC, Apodaca GL, Birder LA. Serotonergic paraneurones in the female mouse urethral epithelium and their potential role in peripheral sensory information processing. Acta Physiol (Oxf) 2018; 222:10.1111/apha.12919. [PMID: 28719042 PMCID: PMC5963688 DOI: 10.1111/apha.12919] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 06/19/2017] [Accepted: 07/13/2017] [Indexed: 01/01/2023]
Abstract
AIM The mechanisms underlying detection and transmission of sensory signals arising from visceral organs, such as the urethra, are poorly understood. Recently, specialized ACh-expressing cells embedded in the urethral epithelium have been proposed as chemosensory sentinels for detection of bacterial infection. Here, we examined the morphology and potential role in sensory signalling of a different class of specialized cells that express serotonin (5-HT), termed paraneurones. METHODS Urethrae, dorsal root ganglia neurones and spinal cords were isolated from adult female mice and used for immunohistochemistry and calcium imaging. Visceromotor reflexes (VMRs) were recorded in vivo. RESULTS We identified two morphologically distinct groups of 5-HT+ cells with distinct regional locations: bipolar-like cells predominant in the mid-urethra and multipolar-like cells predominant in the proximal and distal urethra. Sensory nerve fibres positive for calcitonin gene-related peptide, substance P, and TRPV1 were found in close proximity to 5-HT+ paraneurones. In vitro 5-HT (1 μm) stimulation of urethral primary afferent neurones, mimicking 5-HT release from paraneurones, elicited changes in the intracellular calcium concentration ([Ca2+ ]i ) mediated by 5-HT2 and 5-HT3 receptors. Approximately 50% of 5-HT responding cells also responded to capsaicin with changes in the [Ca2+ ]i . In vivo intra-urethral 5-HT application increased VMRs induced by urethral distention and activated pERK in lumbosacral spinal cord neurones. CONCLUSION These morphological and functional findings provide insights into a putative paraneurone-neural network within the urethra that utilizes 5-HT signalling, presumably from paraneurones, to modulate primary sensory pathways carrying nociceptive and non-nociceptive (mechano-sensitive) information to the central nervous system.
Collapse
Affiliation(s)
- F. A. Kullmann
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - H. H. Chang
- Department of Urology, University of Southern California, Los Angeles, CA, USA
| | - C. Gauthier
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - B. M. McDonnell
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - J.-C. Yeh
- Department of Urology, University of Southern California, Los Angeles, CA, USA
| | - D. R. Clayton
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - A. J. Kanai
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - W. C. de Groat
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - G. L. Apodaca
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - L. A. Birder
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
44
|
Wild AR, Dell'Acqua ML. Potential for therapeutic targeting of AKAP signaling complexes in nervous system disorders. Pharmacol Ther 2017; 185:99-121. [PMID: 29262295 DOI: 10.1016/j.pharmthera.2017.12.004] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
A common feature of neurological and neuropsychiatric disorders is a breakdown in the integrity of intracellular signal transduction pathways. Dysregulation of ion channels and receptors in the cell membrane and the enzymatic mediators that link them to intracellular effectors can lead to synaptic dysfunction and neuronal death. However, therapeutic targeting of these ubiquitous signaling elements can lead to off-target side effects due to their widespread expression in multiple systems of the body. A-kinase anchoring proteins (AKAPs) are multivalent scaffolding proteins that compartmentalize a diverse range of receptor and effector proteins to streamline signaling within nanodomain signalosomes. A number of essential neurological processes are known to critically depend on AKAP-directed signaling and an understanding of the role AKAPs play in nervous system disorders has emerged in recent years. Selective targeting of AKAP protein-protein interactions may be a means to uncouple pathologically active signaling pathways in neurological disorders with a greater degree of specificity. In this review we will discuss the role of AKAPs in both regulating normal nervous system function and dysfunction associated with disease, and the potential for therapeutic targeting of AKAP signaling complexes.
Collapse
Affiliation(s)
- Angela R Wild
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Mark L Dell'Acqua
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA.
| |
Collapse
|
45
|
Georgescu SR, Sârbu MI, Matei C, Ilie MA, Caruntu C, Constantin C, Neagu M, Tampa M. Capsaicin: Friend or Foe in Skin Cancer and Other Related Malignancies? Nutrients 2017; 9:E1365. [PMID: 29258175 PMCID: PMC5748815 DOI: 10.3390/nu9121365] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 12/11/2017] [Accepted: 12/12/2017] [Indexed: 02/07/2023] Open
Abstract
Capsaicin is the main pungent in chili peppers, one of the most commonly used spices in the world; its analgesic and anti-inflammatory properties have been proven in various cultures for centuries. It is a lipophilic substance belonging to the class of vanilloids and an agonist of the transient receptor potential vanilloid 1 receptor. Taking into consideration the complex neuro-immune impact of capsaicin and the potential link between inflammation and carcinogenesis, the effect of capsaicin on muco-cutaneous cancer has aroused a growing interest. The aim of this review is to look over the most recent data regarding the connection between capsaicin and muco-cutaneous cancers, with emphasis on melanoma and muco-cutaneous squamous cell carcinoma.
Collapse
Affiliation(s)
- Simona-Roxana Georgescu
- Department of Dermatology, Carol DavilaUniversity of Medicine and Pharmacy, 020021 Bucharest, Romania.
| | - Maria-Isabela Sârbu
- Department of Dermatology, Carol DavilaUniversity of Medicine and Pharmacy, 020021 Bucharest, Romania.
| | - Clara Matei
- Department of Dermatology, Carol DavilaUniversity of Medicine and Pharmacy, 020021 Bucharest, Romania.
| | - Mihaela Adriana Ilie
- Department of Biochemistry, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania.
| | - Constantin Caruntu
- Department of Physiology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania.
- Department of Dermatology, Prof. N.C. Paulescu National Institute of Diabetes, Nutrition and Metabolic Diseases, 011233 Bucharest, Romania.
| | - Carolina Constantin
- Immunology Department, Victor Babes National Institute of Pathology, 050096 Bucharest, Romania.
| | - Monica Neagu
- Immunology Department, Victor Babes National Institute of Pathology, 050096 Bucharest, Romania.
- Faculty of Biology, University of Bucharest, 76201 Bucharest, Romania.
| | - Mircea Tampa
- Department of Dermatology, Carol DavilaUniversity of Medicine and Pharmacy, 020021 Bucharest, Romania.
| |
Collapse
|
46
|
Chen Y, Huang LYM. A simple and fast method to image calcium activity of neurons from intact dorsal root ganglia using fluorescent chemical Ca 2+ indicators. Mol Pain 2017; 13:1744806917748051. [PMID: 29212403 PMCID: PMC5731619 DOI: 10.1177/1744806917748051] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Chemical calcium indicators have been commonly used to monitor calcium (Ca2+) activity in cell bodies, i.e., somata, of isolated dorsal root ganglion neurons. Recent studies have shown that dorsal root ganglion somata play an essential role in soma–glia interactions and actively participate in the transmission of nociceptive signals. It is therefore desirable to develop methods to study Ca2+ activity in neurons and glia in intact dorsal root ganglia. In our previous studies, we found that incubation of intact dorsal root ganglia with acetoxymethyl dye resulted in efficient Ca2+ dye loading into glial cells but limited dye loading into neurons. Here, we introduce a useful method to load Ca2+ dyes in intact dorsal root ganglion neurons through electroporation. We found that electroporation greatly facilitated loading of Fluo-4 acetoxymethyl, Oregon green bapta-1-488 acetoxymethyl, and Fluo-4 pentapotassium salt into dorsal root ganglion neurons. In contrast, electroporation did not further facilitate dye loading into glia. Using electroporation followed by incubation of acetoxymethyl form Ca2+ dye, we can load acetoxymethyl Ca2+ dye well in both neurons and glia. With this approach, we found that inflammation induced by complete Freund’s adjuvant significantly increased the incidence of neuron–glia interactions in dorsal root ganglia. We also confirmed the actions of capsaicin and morphine on Ca2+ responses in dorsal root ganglion neurons. Thus, by promoting the loading of Ca2+ dye in neurons and glia through electroporation and incubation, Ca2+ activities in neurons and neuron–glia interactions can be well studied in intact dorsal root ganglia.
Collapse
Affiliation(s)
- Yong Chen
- 1 Department of Neuroscience, Cell Biology and Anatomy, 12338 University of Texas Medical Branch, Galveston , TX, USA
| | - Li-Yen M Huang
- 1 Department of Neuroscience, Cell Biology and Anatomy, 12338 University of Texas Medical Branch, Galveston , TX, USA
| |
Collapse
|
47
|
Deftu AF, Filippi A, Gheorghe RO, Ristoiu V. CXCL1 activates TRPV1 via Gi/o protein and actin filaments. Life Sci 2017; 193:282-291. [PMID: 28966134 DOI: 10.1016/j.lfs.2017.09.041] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 09/18/2017] [Accepted: 09/27/2017] [Indexed: 02/07/2023]
Abstract
AIMS CXCL1 is a chemokine with pleiotropic effects, including pain and itch. Itch, an unpleasant sensation that elicits the desire or reflex to scratch, it is evoked mainly from the skin and implicates activation of a specific subset of IB4+, C-type primary afferents. In previous studies we showed that acute application of CXCL1 induced a Ca2+ influx of low amplitude and slow kinetics in a subpopulation of transient receptor potential vanilloid type 1 (TRPV1)+/isolectin B4 (IB4)+dorsal root ganglia neurons which also responded to other itch-inducing agents. In this study we explored the mechanism behind the Ca2+ influx to better understand how CXCL1 acts on primary sensitive neurons to induce itch. MATERIALS AND METHODS Intracellular Ca2+ imaging and patch-clamp recordings on dorsal root ganglia neurons primary cultures and HEK293T cell transiently transfected with TRPV1 and CXCR2 plasmids were used to investigate the acute effect (12min application) of 4nM CXCL1. In primary cultures, the focus was on TRPV1+/IB4+ cells to which the itch-sensitive neurons belong. KEY FINDINGS The results showed that the Ca2+ influx induced by the acute application of CXCL1 is mediated mainly by TRPV1 receptors and depends on extracellular Ca2+ not on intracellular stores. TRPV1 was activated, not sensitized by CXCL1, in a CXCR2 receptors- and actin filaments-dependent manner, since specific blockers and actin depolymerizing agents disrupted the CXCL1 effect. SIGNIFICANCE This study brings additional data about the itch inducing mechanism of CXCL1 chemokine and about a new mechanism of TRPV1 activation via actin filaments.
Collapse
Affiliation(s)
- Alexandru Florian Deftu
- Department of Anatomy, Animal Physiology and Biophysics, Faculty of Biology, University of Bucharest, Splaiul Independenţei 91-95, 050095 Bucharest, Romania
| | - Alexandru Filippi
- Department of Anatomy, Animal Physiology and Biophysics, Faculty of Biology, University of Bucharest, Splaiul Independenţei 91-95, 050095 Bucharest, Romania; Department of Medical Biophysics, University of Medicine and Pharmacy "Carol Davila", Bulevardul Eroilor Sanitari 8, 050474 Bucharest, Romania
| | - Roxana Olimpia Gheorghe
- Department of Anatomy, Animal Physiology and Biophysics, Faculty of Biology, University of Bucharest, Splaiul Independenţei 91-95, 050095 Bucharest, Romania
| | - Violeta Ristoiu
- Department of Anatomy, Animal Physiology and Biophysics, Faculty of Biology, University of Bucharest, Splaiul Independenţei 91-95, 050095 Bucharest, Romania.
| |
Collapse
|
48
|
Stover JD, Farhang N, Berrett KC, Gertz J, Lawrence B, Bowles RD. CRISPR Epigenome Editing of AKAP150 in DRG Neurons Abolishes Degenerative IVD-Induced Neuronal Activation. Mol Ther 2017; 25:2014-2027. [PMID: 28676344 PMCID: PMC5589089 DOI: 10.1016/j.ymthe.2017.06.010] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 06/08/2017] [Accepted: 06/09/2017] [Indexed: 01/07/2023] Open
Abstract
Back pain is a major contributor to disability and has significant socioeconomic impacts worldwide. The degenerative intervertebral disc (IVD) has been hypothesized to contribute to back pain, but a better understanding of the interactions between the degenerative IVD and nociceptive neurons innervating the disc and treatment strategies that directly target these interactions is needed to improve our understanding and treatment of back pain. We investigated degenerative IVD-induced changes to dorsal root ganglion (DRG) neuron activity and utilized CRISPR epigenome editing as a neuromodulation strategy. By exposing DRG neurons to degenerative IVD-conditioned media under both normal and pathological IVD pH levels, we demonstrate that degenerative IVDs trigger interleukin (IL)-6-induced increases in neuron activity to thermal stimuli, which is directly mediated by AKAP and enhanced by acidic pH. Utilizing this novel information on AKAP-mediated increases in nociceptive neuron activity, we developed lentiviral CRISPR epigenome editing vectors that modulate endogenous expression of AKAP150 by targeted promoter histone methylation. When delivered to DRG neurons, these epigenome-modifying vectors abolished degenerative IVD-induced DRG-elevated neuron activity while preserving non-pathologic neuron activity. This work elucidates the potential for CRISPR epigenome editing as a targeted gene-based pain neuromodulation strategy.
Collapse
Affiliation(s)
- Joshua D Stover
- Department of Bioengineering, University of Utah, Salt Lake City, UT 84112, USA
| | - Niloofar Farhang
- Department of Bioengineering, University of Utah, Salt Lake City, UT 84112, USA
| | - Kristofer C Berrett
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112, USA; Hunstman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Jason Gertz
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112, USA; Hunstman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Brandon Lawrence
- Department of Orthopaedics, University of Utah, Salt Lake City, UT 84112, USA
| | - Robby D Bowles
- Department of Bioengineering, University of Utah, Salt Lake City, UT 84112, USA; Department of Orthopaedics, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
49
|
The Complement System Component C5a Produces Thermal Hyperalgesia via Macrophage-to-Nociceptor Signaling That Requires NGF and TRPV1. J Neurosci 2017; 36:5055-70. [PMID: 27147658 DOI: 10.1523/jneurosci.3249-15.2016] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 03/21/2016] [Indexed: 12/31/2022] Open
Abstract
UNLABELLED The complement cascade is a principal component of innate immunity. Recent studies have underscored the importance of C5a and other components of the complement system in inflammatory and neuropathic pain, although the underlying mechanisms are largely unknown. In particular, it is unclear how the complement system communicates with nociceptors and which ion channels and receptors are involved. Here we demonstrate that inflammatory thermal and mechanical hyperalgesia induced by complete Freund's adjuvant was accompanied by C5a upregulation and was markedly reduced by C5a receptor (C5aR1) knock-out or treatment with the C5aR1 antagonist PMX53. Direct administration of C5a into the mouse hindpaw produced strong thermal hyperalgesia, an effect that was absent in TRPV1 knock-out mice, and was blocked by the TRPV1 antagonist AMG9810. Immunohistochemistry of mouse plantar skin showed prominent expression of C5aR1 in macrophages. Additionally, C5a evoked strong Ca(2+) mobilization in macrophages. Macrophage depletion in transgenic macrophage Fas-induced apoptosis mice abolished C5a-dependent thermal hyperalgesia. Examination of inflammatory mediators following C5a injection revealed a rapid upregulation of NGF, a mediator known to sensitize TRPV1. Preinjection of an NGF-neutralizing antibody or Trk inhibitor GNF-5837 prevented C5a-induced thermal hyperalgesia. Notably, NGF-induced thermal hyperalgesia was unaffected by macrophage depletion. Collectively, these results suggest that complement fragment C5a induces thermal hyperalgesia by triggering macrophage-dependent signaling that involves mobilization of NGF and NGF-dependent sensitization of TRPV1. Our findings highlight the importance of macrophage-to-neuron signaling in pain processing and identify C5a, NGF, and TRPV1 as key players in this cross-cellular communication. SIGNIFICANCE STATEMENT This study provides mechanistic insight into how the complement system, a key component of innate immunity, regulates the development of pain hypersensitivity. We demonstrate a crucial role of the C5a receptor, C5aR1, in the development of inflammatory thermal and mechanical sensitization. By focusing on the mechanisms of C5a-induced thermal hyperalgesia, we show that this process requires recruitment of macrophages and initiation of macrophage-to-nociceptor signaling. At the molecular level, we demonstrate that this signaling depends on NGF and is mediated by the heat-sensitive nociceptive channel TRPV1. This deeper understanding of how immune cells and neurons interact to regulate pain processing is expected to facilitate mechanism-based approaches in the development of new analgesics.
Collapse
|
50
|
Por ED, Sandoval ML, Thomas-Benson C, Burke TA, Doyle Brackley A, Jeske NA, Cleland JM, Lund BJ. Repeat low-level blast exposure increases transient receptor potential vanilloid 1 (TRPV1) and endothelin-1 (ET-1) expression in the trigeminal ganglion. PLoS One 2017; 12:e0182102. [PMID: 28797041 PMCID: PMC5552217 DOI: 10.1371/journal.pone.0182102] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 07/12/2017] [Indexed: 12/14/2022] Open
Abstract
Blast-associated sensory and cognitive trauma sustained by military service members is an area of extensively studied research. Recent studies in our laboratory have revealed that low-level blast exposure increased expression of transient receptor potential vanilloid 1 (TRPV1) and endothelin-1 (ET-1), proteins well characterized for their role in mediating pain transmission, in the cornea. Determining the functional consequences of these alterations in protein expression is critical to understanding blast-related sensory trauma. Thus, the purpose of this study was to examine TRPV1 and ET-1 expression in ocular associated sensory tissues following primary and tertiary blast. A rodent model of blast injury was used in which anesthetized animals, unrestrained or restrained, received a single or repeat blast (73.8 ± 5.5 kPa) from a compressed air shock tube once or daily for five consecutive days, respectively. Behavioral and functional analyses were conducted to assess blast effects on nocifensive behavior and TRPV1 activity. Immunohistochemistry and Western Blot were also performed with trigeminal ganglia (TG) to determine TRPV1, ET-1 and glial fibrillary associated protein (GFAP) expression following blast. Increased TRPV1, ET-1 and GFAP were detected in the TG of animals exposed to repeat blast. Increased nocifensive responses were also observed in animals exposed to repeat, tertiary blast as compared to single blast and control. Moreover, decreased TRPV1 desensitization was observed in TG neurons exposed to repeat blast. Repeat, tertiary blast resulted in increased TRPV1, ET-1 and GFAP expression in the TG, enhanced nociception and decreased TRPV1 desensitization.
Collapse
Affiliation(s)
- Elaine D. Por
- Ocular Trauma, United States Army Institute of Surgical Research, Fort Sam, Houston, Texas, United States of America
- * E-mail:
| | - Melody L. Sandoval
- Ocular Trauma, United States Army Institute of Surgical Research, Fort Sam, Houston, Texas, United States of America
| | - Chiquita Thomas-Benson
- Ocular Trauma, United States Army Institute of Surgical Research, Fort Sam, Houston, Texas, United States of America
| | - Teresa A. Burke
- Ocular Trauma, United States Army Institute of Surgical Research, Fort Sam, Houston, Texas, United States of America
| | - Allison Doyle Brackley
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Nathaniel A. Jeske
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Department of Oral and Maxillofacial Surgery, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Department of Physiology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Jeffery M. Cleland
- Ocular Trauma, United States Army Institute of Surgical Research, Fort Sam, Houston, Texas, United States of America
| | - Brian J. Lund
- Ocular Trauma, United States Army Institute of Surgical Research, Fort Sam, Houston, Texas, United States of America
| |
Collapse
|