1
|
Sankaranarayanan I, Kume M, Mohammed A, Mwirigi JM, Inturi NN, Munro G, Petersen KA, Tavares-Ferreira D, Price TJ. Persistent changes in the dorsal root ganglion nociceptor translatome governs hyperalgesic priming in mice: roles of GPR88 and Meteorin. Pain 2025:00006396-990000000-00811. [PMID: 39878635 DOI: 10.1097/j.pain.0000000000003523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 10/08/2024] [Indexed: 01/31/2025]
Abstract
ABSTRACT Hyperalgesic priming is a model system that has been widely used to understand plasticity in painful stimulus-detecting sensory neurons, called nociceptors. A key feature of this model system is that following priming, stimuli that do not normally cause hyperalgesia now readily provoke this state. We hypothesized that hyperalgesic priming occurs because of reorganization of translation of mRNA in nociceptors. To test this hypothesis, we used paclitaxel treatment as the priming stimulus and translating ribosome affinity purification to measure persistent changes in mRNA translation in Nav1.8+ nociceptors. Translating ribosome affinity purification sequencing revealed 161 genes with persistently altered mRNA translation in the primed state. Among these genes, we identified Gpr88 as upregulated and Metrn as downregulated. To provide functional evidence for these changes in hyperalgesic priming in a related priming model, we used the interleukin-6 priming model. A GPR88 agonist injection into the paw had no effect in naive mice but caused mechanical hypersensitivity and grimacing responses in female primed mice. Systemic Meteorin treatment in primed mice completely reversed established hyperalgesic priming mechanical hypersensitivity and grimacing responses to prostaglandin E2 in female mice. Our work demonstrates that altered nociceptor translatomes are causative in producing hyperalgesic priming in multiple models in female mice.
Collapse
Affiliation(s)
- Ishwarya Sankaranarayanan
- Department of Neuroscience, Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX
| | - Moeno Kume
- Department of Neuroscience, Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX
| | - Ayaan Mohammed
- Department of Neuroscience, Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX
| | - Juliet M Mwirigi
- Department of Neuroscience, Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX
| | - Nikhil Nageswar Inturi
- Department of Neuroscience, Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX
| | | | | | - Diana Tavares-Ferreira
- Department of Neuroscience, Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX
| | - Theodore J Price
- Department of Neuroscience, Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX
| |
Collapse
|
2
|
Picco F, Zeboudj L, Oggero S, Prato V, Burgoyne T, Gamper N, Malcangio M. Macrophage to neuron communication via extracellular vesicles in neuropathic pain conditions. Heliyon 2025; 11:e41268. [PMID: 39811367 PMCID: PMC11730208 DOI: 10.1016/j.heliyon.2024.e41268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 11/27/2024] [Accepted: 12/15/2024] [Indexed: 01/16/2025] Open
Abstract
Neuropathic pain following peripheral nerve injury results from maladaptive changes in neurons and immune cells contribution to mechanisms underlying chronic pain. Specifically, in dorsal root ganglia (DRG), sensory neuron cell bodies release extracellular vesicles (EVs) which promote pro-inflammatory macrophage accumulation that facilitates nociceptive signalling. Here, we show that macrophages shuttle EVs to neurons. Indeed, bone marrow-derived macrophages (BMDMs) release EVs containing microRNA-155 (miR-155) which are taken up by cultured sensory neurons. EV-mediated transfer of miR-155 suppresses phosphatase Ship1 expression and increases cytokine interleukin-6 (IL-6) contents. Intrathecal-injected BMDM-derived EVs accumulate in lumbar DRG and EVs containing miR-155 antagomir result in Ship1 upregulation, Il6 downregulation in neurons in concomitance to attenuation of neuropathic mechanical hypersensitivity. These data suggest that, under neuropathic conditions, pro-inflammatory macrophages shuttle EV-containing miR-155 to neurons and contribute to the expression of pronociceptive IL-6 in DRG.
Collapse
Affiliation(s)
- Francesca Picco
- Wolfson Sensory, Pain and Regeneration Centre, King’s College London, London, United Kingdom
| | - Lynda Zeboudj
- Wolfson Sensory, Pain and Regeneration Centre, King’s College London, London, United Kingdom
| | - Silvia Oggero
- Wolfson Sensory, Pain and Regeneration Centre, King’s College London, London, United Kingdom
| | - Vincenzo Prato
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| | - Thomas Burgoyne
- UCL Institute of Ophthalmology, University College London, London, EC1V 9EL, United Kingdom
- Royal Brompton Hospital, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Nikita Gamper
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| | - Marzia Malcangio
- Wolfson Sensory, Pain and Regeneration Centre, King’s College London, London, United Kingdom
| |
Collapse
|
3
|
Vazquez E, Richter F, Natura G, König C, Eitner A, Schaible HG. Direct Effects of the Janus Kinase Inhibitor Baricitinib on Sensory Neurons. Int J Mol Sci 2024; 25:11943. [PMID: 39596013 PMCID: PMC11593535 DOI: 10.3390/ijms252211943] [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: 10/15/2024] [Revised: 11/01/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024] Open
Abstract
Therapeutically, the Janus kinase (Jak) 1/Jak2 inhibitor baricitinib reduces the pathology of rheumatoid arthritis and may also reduce pain. Here, we investigated whether baricitinib directly affects joint nociceptors. We recorded action potentials from nociceptive C- and A∂-fibers of the normal and inflamed knee joint in anesthetized rats to monitor their responses to innocuous and noxious joint rotation. In isolated and cultured dorsal root ganglion (DRG) neurons, we examined Stat3 activation using Western blots and monitored excitability using patch-clamp recordings. Intra-articular injection of baricitinib did not alter C- and A∂-fiber responses to innocuous and noxious rotations of the normal knee but reduced C-fiber responses to these stimuli in inflamed joints. Baricitinib prevented the increase in C-fiber responses to joint rotation evoked by interleukin (IL)-6 plus soluble interleukin-6 receptor (sIL-6R) but not the increase evoked by TNF. In DRG neurons, baricitinib blocked Stat3 activation by hyper-IL-6, and baricitinib or the Stat3 inhibitor Sta21 prevented induction of hyperexcitability by IL-6 plus sIL-6R. Thus, neuronal Jaks are involved in the generation of C-fiber hyperexcitability induced by inflammation and IL-6. Pain reduction by baricitinib may result, at least in part, from direct effects on joint nociceptors.
Collapse
Affiliation(s)
- Enrique Vazquez
- Institute of Physiology 1/Neurophysiology, Jena University Hospital, Friedrich-Schiller-University, D-07740 Jena, Germany; (E.V.); (F.R.); (G.N.); (C.K.)
| | - Frank Richter
- Institute of Physiology 1/Neurophysiology, Jena University Hospital, Friedrich-Schiller-University, D-07740 Jena, Germany; (E.V.); (F.R.); (G.N.); (C.K.)
| | - Gabriel Natura
- Institute of Physiology 1/Neurophysiology, Jena University Hospital, Friedrich-Schiller-University, D-07740 Jena, Germany; (E.V.); (F.R.); (G.N.); (C.K.)
| | - Christian König
- Institute of Physiology 1/Neurophysiology, Jena University Hospital, Friedrich-Schiller-University, D-07740 Jena, Germany; (E.V.); (F.R.); (G.N.); (C.K.)
| | - Annett Eitner
- Department of Trauma, Hand and Reconstructive Surgery, Experimental Trauma Surgery, Jena University Hospital, Friedrich-Schiller-University, D-07740 Jena, Germany;
| | - Hans-Georg Schaible
- Institute of Physiology 1/Neurophysiology, Jena University Hospital, Friedrich-Schiller-University, D-07740 Jena, Germany; (E.V.); (F.R.); (G.N.); (C.K.)
| |
Collapse
|
4
|
Leisengang S. Pain research in a petri dish? Advantages and limitations of neuro-glial primary cell cultures from structures of the nociceptive system. Brain Behav Immun Health 2024; 41:100854. [PMID: 39308957 PMCID: PMC11415590 DOI: 10.1016/j.bbih.2024.100854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 06/26/2024] [Accepted: 09/03/2024] [Indexed: 09/25/2024] Open
Abstract
How can we learn more about pain without causing pain in humans or animals? This short review focuses on neuro-glial primary cell cultures as models to study neuro-immune interactions in the context of pain and discusses their advantages and limitations. The field of basic pain research places scientists in an ethical dilemma. We aim to understand underlying mechanisms of pain for an improved pain therapy for humans and animals. At the same time, this regularly includes the induction of pain in model animals. Within the field of psychoneuroimmunology, the examination of the complexity of neuro-immune interactions in health and disease as well as the bi-directional communication between the brain and the periphery make animal experiments an inevitable part of pain research. To address ethical and legal considerations as well as the growing societal awareness for animal welfare, scientists push for the identification and characterization of complementary methods to implement the 3R principle of Russel and Burch. As such, methods to replace animal studies, reduce the number of animals used, and refine experiments are tested. Neuro-glial primary cell cultures of structures of the nociceptive system, such as dorsal root ganglia (DRG) or the spinal dorsal horn (SDH) represent useful in vitro tools, when research comes to a cellular and molecular level. They allow for studying mechanisms of neuronal sensitization, glial cell activation, or the role of specific inflammatory mediators and intracellular signaling cascades involved in the development of inflammatory and neuropathic pain. Moreover, DRG/SDH-cultures provide the opportunity to test novel strategies for interventions, such as pharmaceuticals or cell-based therapies targeting neuroinflammatory processes. Thereby, in vitro models contribute to a better understanding of neuron-glia-immune communication in the context of pain and in the advancement of pain therapies. However, this can only be one piece in a large puzzle. Our knowledge about the complexity of pain will depend on studies in humans and animals applied in vitro and in vivo and will benefit from clear and open-minded interdisciplinary communication and transparency in public outreach.
Collapse
Affiliation(s)
- Stephan Leisengang
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University Giessen, Frankfurter Strasse 100, 35392 Giessen, Germany
- Translational Neuroscience Network Giessen (TNNG), Justus Liebig University Giessen, Germany
- Center for Mind, Brain and Behavior (CMBB), Philipps University Marburg & Justus Liebig University Giessen, Germany
| |
Collapse
|
5
|
Salib AMN, Crane MJ, Jamieson AM, Lipscombe D. Peripheral Ca V2.2 Channels in the Skin Regulate Prolonged Heat Hypersensitivity during Neuroinflammation. eNeuro 2024; 11:ENEURO.0311-24.2024. [PMID: 39433408 PMCID: PMC11599794 DOI: 10.1523/eneuro.0311-24.2024] [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: 07/13/2024] [Revised: 09/13/2024] [Accepted: 09/26/2024] [Indexed: 10/23/2024] Open
Abstract
Neuroinflammation can lead to chronic maladaptive pain affecting millions of people worldwide. Neurotransmitters, cytokines, and ion channels are implicated in neuroimmune cell signaling, but their roles in specific behavioral responses are not fully elucidated. Voltage-gated CaV2.2 channel activity in skin controls rapid and transient heat hypersensitivity induced by intradermal (i.d.) capsaicin via IL-1ɑ cytokine signaling. CaV2.2 channels are not, however, involved in mechanical hypersensitivity that developed in the i.d. capsaicin animal model. Here, we show that CaV2.2 channels are also critical for heat hypersensitivity induced by i.d. complete Freund adjuvant (CFA). i.d. CFA, a model of chronic neuroinflammation, involves ongoing cytokine signaling for days leading to pronounced edema and hypersensitivity to sensory stimuli. Peripheral CaV2.2 channel activity in the skin was required for the full development and week-long time course of heat hypersensitivity induced by i.d. CFA, but paw edema and mechanical hypersensitivity were independent of CaV2.2 channel activity. CFA induced increases in several cytokines in hindpaw fluid including IL-6 which was also dependent on CaV2.2 channel activity. Using IL-6-specific neutralizing antibodies in vivo, we show that IL-6 contributes to heat hypersensitivity and that neutralizing both IL-1ɑ and IL-6 was even more effective at reducing the magnitude and duration of CFA-induced heat hypersensitivity. Our findings demonstrate a functional link between CaV2.2 channel activity and the release of IL-6 in the skin and show that CaV2.2 channels have a privileged role in the induction and maintenance of heat hypersensitivity during chronic forms of neuroinflammation in the skin.
Collapse
Affiliation(s)
- Anne-Mary N Salib
- Departments of Neuroscience, Brown University, Providence, Rhode Island 02912
- Carney Institute for Brain Science, Brown University, Providence, Rhode Island 02912
| | - Meredith J Crane
- Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island 02912
| | - Amanda M Jamieson
- Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island 02912
| | - Diane Lipscombe
- Departments of Neuroscience, Brown University, Providence, Rhode Island 02912
- Carney Institute for Brain Science, Brown University, Providence, Rhode Island 02912
| |
Collapse
|
6
|
Sankaranarayanan I, Kume M, Mohammed A, Mwirigi JM, Inturi NN, Munro G, Petersen KA, Tavares-Ferreira D, Price TJ. Persistent changes in nociceptor translatomes govern hyperalgesic priming in mouse models. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.07.606891. [PMID: 39149295 PMCID: PMC11326310 DOI: 10.1101/2024.08.07.606891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Hyperalgesic priming is a model system that has been widely used to understand plasticity in painful stimulus-detecting sensory neurons, called nociceptors. A key feature of this model system is that following priming, stimuli that do not normally cause hyperalgesia now readily provoke this state. We hypothesized that hyperalgesic priming occurs due to reorganization of translation of mRNA in nociceptors. To test this hypothesis, we used paclitaxel treatment as the priming stimulus and translating ribosome affinity purification (TRAP) to measure persistent changes in mRNA translation in Nav1.8+ nociceptors. TRAP sequencing revealed 161 genes with persistently altered mRNA translation in the primed state. We identified Gpr88 as upregulated and Metrn as downregulated. We confirmed a functional role for these genes, wherein a GPR88 agonist causes pain only in primed mice and established hyperalgesic priming is reversed by Meteorin. Our work demonstrates that altered nociceptor translatomes are causative in producing hyperalgesic priming.
Collapse
Affiliation(s)
- Ishwarya Sankaranarayanan
- Pain Neurobiology Research Group, Department of Neuroscience, Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas
| | - Moeno Kume
- Pain Neurobiology Research Group, Department of Neuroscience, Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas
| | - Ayaan Mohammed
- Pain Neurobiology Research Group, Department of Neuroscience, Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas
| | - Juliet M Mwirigi
- Pain Neurobiology Research Group, Department of Neuroscience, Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas
| | - Nikhil Nageswar Inturi
- Pain Neurobiology Research Group, Department of Neuroscience, Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas
| | | | | | - Diana Tavares-Ferreira
- Pain Neurobiology Research Group, Department of Neuroscience, Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas
| | - Theodore J Price
- Pain Neurobiology Research Group, Department of Neuroscience, Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas
| |
Collapse
|
7
|
Salib AMN, Crane MJ, Jamieson AM, Lipscombe D. Peripheral Ca V2.2 channels in skin regulate prolonged heat hypersensitivity during neuroinflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.13.603149. [PMID: 39071304 PMCID: PMC11275762 DOI: 10.1101/2024.07.13.603149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Neuroinflammation can lead to chronic maladaptive pain affecting millions of people worldwide. Neurotransmitters, cytokines, and ion channels are implicated in neuro-immune cell signaling but their roles in specific behavioral responses are not fully elucidated. Voltage-gated CaV2.2 channel activity in skin controls rapid and transient heat hypersensitivity induced by intradermal capsaicin via IL-1α cytokine signaling. CaV2.2 channels are not, however, involved in mechanical hypersensitivity that developed in the same animal model. Here, we show that CaV2.2 channels are also critical for heat hypersensitivity induced by the intradermal (id) Complete Freund's Adjuvant (CFA) model of chronic neuroinflammation that involves ongoing cytokine signaling for days. Ongoing CFA-induced cytokine signaling cascades in skin lead to pronounced edema, and hypersensitivity to sensory stimuli. Peripheral CaV2.2 channel activity in skin is required for the full development and week-long time course of heat hypersensitivity induced by id CFA. CaV2.2 channels, by contrast, are not involved in paw edema and mechanical hypersensitivity. CFA induced increases in cytokines in hind paws including IL-6 which was dependent on CaV2.2 channel activity. Using IL-6 specific neutralizing antibodies, we show that IL-6 contributes to heat hypersensitivity and, neutralizing both IL-1α and IL-6 was even more effective at reducing the magnitude and duration of CFA-induced heat hypersensitivity. Our findings demonstrate a functional link between CaV2.2 channel activity and the release of IL-6 in skin and show that CaV2.2 channels have a privileged role in the induction and maintenance of heat hypersensitivity during chronic forms of neuroinflammation in skin.
Collapse
Affiliation(s)
- Anne-Mary N Salib
- Department of Neuroscience & the Carney Institute for Brain Science Brown University, Providence, RI 02912, USA
| | - Meredith J Crane
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA
| | - Amanda M Jamieson
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA
| | - Diane Lipscombe
- Department of Neuroscience & the Carney Institute for Brain Science Brown University, Providence, RI 02912, USA
| |
Collapse
|
8
|
Roy A, Segond von Banchet G, Gimeno-Ferrer F, König C, Eitner A, Ebersberger A, Ebbinghaus M, Leuchtweis J, Schaible HG. Impact of Interleukin-6 Activation and Arthritis on Epidermal Growth Factor Receptor (EGFR) Activation in Sensory Neurons and the Spinal Cord. Int J Mol Sci 2024; 25:7168. [PMID: 39000275 PMCID: PMC11241234 DOI: 10.3390/ijms25137168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/16/2024] Open
Abstract
In tumor cells, interleukin-6 (IL-6) signaling can lead to activation of the epidermal growth factor receptor (EGFR), which prolongs Stat3 activation. In the present experiments, we tested the hypothesis that IL-6 signaling activates EGFR signaling in peripheral and spinal nociception and examined whether EGFR localization and activation coincide with pain-related behaviors in arthritis. In vivo in anesthetized rats, spinal application of the EGFR receptor blocker gefitinib reduced the responses of spinal cord neurons to noxious joint stimulation, but only after spinal pretreatment with IL-6 and soluble IL-6 receptor. Using Western blots, we found that IL-6-induced Stat3 activation was reduced by gefitinib in microglial cells of the BV2 cell line, but not in cultured DRG neurons. Immunohistochemistry showed EGFR localization in most DRG neurons from normal rats, but significant downregulation in the acute and most painful arthritis phase. In the spinal cord of mice, EGFR was highly activated mainly in the chronic phase of inflammation, with localization in neurons. These data suggest that spinal IL-6 signaling may activate spinal EGFR signaling. Downregulation of EGFR in DRG neurons in acute arthritis may limit nociception, but pronounced delayed activation of EGFR in the spinal cord may be involved in chronic inflammatory pain.
Collapse
Affiliation(s)
- Anutosh Roy
- Institute of Physiology 1/Neurophysiology, Jena University Hospital, Friedrich-Schiller-University, 07743 Jena, Germany; (A.R.); (G.S.v.B.); (F.G.-F.); (C.K.); (A.E.); (M.E.); (J.L.)
| | - Gisela Segond von Banchet
- Institute of Physiology 1/Neurophysiology, Jena University Hospital, Friedrich-Schiller-University, 07743 Jena, Germany; (A.R.); (G.S.v.B.); (F.G.-F.); (C.K.); (A.E.); (M.E.); (J.L.)
| | - Fátima Gimeno-Ferrer
- Institute of Physiology 1/Neurophysiology, Jena University Hospital, Friedrich-Schiller-University, 07743 Jena, Germany; (A.R.); (G.S.v.B.); (F.G.-F.); (C.K.); (A.E.); (M.E.); (J.L.)
| | - Christian König
- Institute of Physiology 1/Neurophysiology, Jena University Hospital, Friedrich-Schiller-University, 07743 Jena, Germany; (A.R.); (G.S.v.B.); (F.G.-F.); (C.K.); (A.E.); (M.E.); (J.L.)
| | - Annett Eitner
- Department of Trauma, Hand and Reconstructive Surgery, Experimental Trauma Surgery, Jena University Hospital, Friedrich-Schiller-University, 07743 Jena, Germany;
| | - Andrea Ebersberger
- Institute of Physiology 1/Neurophysiology, Jena University Hospital, Friedrich-Schiller-University, 07743 Jena, Germany; (A.R.); (G.S.v.B.); (F.G.-F.); (C.K.); (A.E.); (M.E.); (J.L.)
| | - Matthias Ebbinghaus
- Institute of Physiology 1/Neurophysiology, Jena University Hospital, Friedrich-Schiller-University, 07743 Jena, Germany; (A.R.); (G.S.v.B.); (F.G.-F.); (C.K.); (A.E.); (M.E.); (J.L.)
| | - Johannes Leuchtweis
- Institute of Physiology 1/Neurophysiology, Jena University Hospital, Friedrich-Schiller-University, 07743 Jena, Germany; (A.R.); (G.S.v.B.); (F.G.-F.); (C.K.); (A.E.); (M.E.); (J.L.)
| | - Hans-Georg Schaible
- Institute of Physiology 1/Neurophysiology, Jena University Hospital, Friedrich-Schiller-University, 07743 Jena, Germany; (A.R.); (G.S.v.B.); (F.G.-F.); (C.K.); (A.E.); (M.E.); (J.L.)
| |
Collapse
|
9
|
Zhu X, Jia Z, Zhou Y, Wu J, Cao M, Hu C, Yu L, Chen Z. Current advances in the pain treatment and mechanisms of Traditional Chinese Medicine. Phytother Res 2024. [PMID: 39031847 DOI: 10.1002/ptr.8259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 05/20/2024] [Accepted: 05/20/2024] [Indexed: 07/22/2024]
Abstract
Traditional Chinese Medicine (TCM), as a unique medical model in China, has been shown to be effective in the treatment of many diseases. It has been proven that TCM can increase the pain threshold, increase the level of endorphins and enkephalins in the body, and reduce the body's response to adverse stimuli. In recent years, TCM scholars have made valuable explorations in the field of pain treatment, using methods such as internal and external application of TCM and acupuncture to carry out research on pain treatment and have achieved more satisfactory results. TCM treats pain in a variety of ways, and with the discovery of a variety of potential bioactive substances for pain treatment. With the new progress in the research of other TCM treatment methods for pain, TCM will have greater potential in the clinical application of pain.
Collapse
Affiliation(s)
- Xiaoli Zhu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhuolin Jia
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ye Zhou
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jie Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Mayijie Cao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Changjiang Hu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lingying Yu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhimin Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
10
|
Liu PW, Zhang H, Werley CA, Pichler M, Ryan SJ, Lewarch CL, Jacques J, Grooms J, Ferrante J, Li G, Zhang D, Bremmer N, Barnett A, Chantre R, Elder AE, Cohen AE, Williams LA, Dempsey GT, McManus OB. A phenotypic screening platform for chronic pain therapeutics using all-optical electrophysiology. Pain 2024; 165:922-940. [PMID: 37963235 PMCID: PMC10950549 DOI: 10.1097/j.pain.0000000000003090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 08/30/2023] [Indexed: 11/16/2023]
Abstract
ABSTRACT Chronic pain associated with osteoarthritis (OA) remains an intractable problem with few effective treatment options. New approaches are needed to model the disease biology and to drive discovery of therapeutics. We present an in vitro model of OA pain, where dorsal root ganglion (DRG) sensory neurons were sensitized by a defined mixture of disease-relevant inflammatory mediators, here called Sensitizing PAin Reagent Composition or SPARC. Osteoarthritis-SPARC components showed synergistic or additive effects when applied in combination and induced pain phenotypes in vivo. To measure the effect of OA-SPARC on neural firing in a scalable format, we used a custom system for high throughput all-optical electrophysiology. This system enabled light-based membrane voltage recordings from hundreds of neurons in parallel with single cell and single action potential resolution and a throughput of up to 500,000 neurons per day. A computational framework was developed to construct a multiparameter OA-SPARC neuronal phenotype and to quantitatively assess phenotype reversal by candidate pharmacology. We screened ∼3000 approved drugs and mechanistically focused compounds, yielding data from over 1.2 million individual neurons with detailed assessment of functional OA-SPARC phenotype rescue and orthogonal "off-target" effects. Analysis of confirmed hits revealed diverse potential analgesic mechanisms including ion channel modulators and other mechanisms including MEK inhibitors and tyrosine kinase modulators. Our results suggest that the Raf-MEK-ERK axis in DRG neurons may integrate the inputs from multiple upstream inflammatory mediators found in osteoarthritis patient joints, and MAPK pathway activation in DRG neurons may contribute to chronic pain in patients with osteoarthritis.
Collapse
Affiliation(s)
- Pin W. Liu
- Quiver Bioscience, Cambridge, MA, United States
| | | | | | | | | | | | | | | | | | - Guangde Li
- Quiver Bioscience, Cambridge, MA, United States
| | - Dawei Zhang
- Quiver Bioscience, Cambridge, MA, United States
| | | | | | | | | | - Adam E. Cohen
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, United States
| | | | | | | |
Collapse
|
11
|
Sawicki CM, Janal MN, Gonzalez SH, Wu AK, Schmidt BL, Albertson DG. Measurement of the Association of Pain with Clinical Characteristics in Oral Cancer Patients at Diagnosis and Prior to Cancer Treatment. J Pain Res 2024; 17:501-508. [PMID: 38328017 PMCID: PMC10848821 DOI: 10.2147/jpr.s423318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 09/25/2023] [Indexed: 02/09/2024] Open
Abstract
Aim Oral cancer patients suffer pain at the site of the cancer, which degrades quality of life (QoL). The University of California San Francisco Oral Cancer Pain Questionnaire (UCSFOCPQ), the only validated instrument specifically designed for measuring oral cancer pain, measures the intensity and nature of pain and the level of functional restriction due to pain. Purpose The aim of this study was to compare pain reported by untreated oral cancer patients on the UCSFOCPQ with pain they reported on the Brief Pain Inventory (BPI), an instrument widely used to evaluate cancer and non-cancer pain. Patients and Methods The correlation between pain measured by the two instruments and clinical characteristics were analyzed. Thirty newly diagnosed oral cancer patients completed the UCSFOCPQ and the BPI. Results Pain severity measurements made by the UCSFOCPQ and BPI were concordant; however, the widely used BPI average pain over 24 hours score appeared less sensitive to detect association of oral cancer pain with clinical characteristics of patients prior to treatment (nodal status, depth of invasion, DOI). A BPI average score that includes responses to questions that measure both pain severity and interference with function performs similarly to the UCSFOCPQ in detection of associations with nodal status, pathologic T stage (pT stage), stage and depth of invasion (DOI). Conclusion Pain assessment instruments that measure sensory and interference dimensions of oral cancer pain correlate with biologic features and clinical behavior.
Collapse
Affiliation(s)
- Caroline M Sawicki
- Department of Pediatric Dentistry, New York University College of Dentistry, New York, NY, USA
| | - Malvin N Janal
- Department of Epidemiology & Health Promotion New York University College of Dentistry, New York, NY, USA
| | - Sung Hye Gonzalez
- NYU Dentistry Translational Research Center, New York University College of Dentistry, New York, NY, USA
| | - Angie K Wu
- NYU Dentistry Translational Research Center, New York University College of Dentistry, New York, NY, USA
| | - Brian L Schmidt
- NYU Dentistry Translational Research Center, New York University College of Dentistry, New York, NY, USA
- Department of Oral and Maxillofacial Surgery, New York University College of Dentistry, New York, NY, USA
- NYU Pain Research Center, New York University College of Dentistry, New York, NY, USA
| | - Donna G Albertson
- NYU Dentistry Translational Research Center, New York University College of Dentistry, New York, NY, USA
- Department of Oral and Maxillofacial Surgery, New York University College of Dentistry, New York, NY, USA
- NYU Pain Research Center, New York University College of Dentistry, New York, NY, USA
| |
Collapse
|
12
|
Donovan LJ, Brewer CL, Bond SF, Lopez AP, Hansen LH, Jordan CE, González OC, de Lecea L, Kauer JA, Tawfik VL. Aging and injury drive neuronal senescence in the dorsal root ganglia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.20.576299. [PMID: 39829815 PMCID: PMC11741248 DOI: 10.1101/2024.01.20.576299] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Aging negatively impacts central nervous system function; however, the cellular impact of aging in the peripheral nervous system remains poorly understood. Aged individuals are more likely to experience increased pain and slower recovery after trauma. Such injury can damage vulnerable peripheral axons of dorsal root ganglion (DRG) neurons resulting in somatosensory dysfunction. One cellular mechanism common to both aging and injury is cellular senescence, a complex cell state that can contribute to the aged pro-inflammatory environment. We uncovered, for the first time, DRG neuron senescence in the context of aging and pain-inducing peripheral nerve injury in young and aged mice. Aged DRG neurons displayed multiple markers of senescence (SA-β-gal, p21, p16, IL6) when compared to young DRG neurons. Peripheral nerve injury triggered a further accumulation of senescent DRG neurons over time post-injury in young and aged DRG. These senescent neurons were dynamic and heterogeneous in their expression of senescence markers, p16, p21, and senescence-associated secretory phenotype (SASP) expression of IL6, which was influenced by age. An electrophysiological characterization of senescence marker-expressing neurons revealed high-firing and nociceptor-like phenotypes within these populations. In addition, we observed improvement in nociceptive behaviors in young and aged nerve-injured mice after treatment with a senolytic agent that eliminates senescent cells. Finally, we confirmed in human post-mortem DRG samples that neuronal senescence is present and increases with age. Overall, we describe a susceptibility of the peripheral nervous system to neuronal senescence with age or injury that may be a targetable mechanism to treat sensory dysfunction, such as chronic pain, particularly in aged populations.
Collapse
Affiliation(s)
- Lauren J. Donovan
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA 94305, USA
| | - Chelsie L. Brewer
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Sabrina F. Bond
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA 94305, USA
| | - Aleishai Pena Lopez
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA 94305, USA
| | - Linus H. Hansen
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA 94305, USA
| | - Claire E. Jordan
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA 94305, USA
| | - Oscar C. González
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Luis de Lecea
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Julie A. Kauer
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Vivianne L. Tawfik
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
13
|
Saloman JL, Li Y, Stello K, Li W, Li S, Phillips AE, Hall K, Fogel EL, Vege SS, Li L, Andersen DK, Fisher WE, Forsmark CE, Hart PA, Pandol SJ, Park WG, Topazian MD, Van Den Eeden SK, Serrano J, Conwell DL, Yadav D. Serum Biomarkers of Nociceptive and Neuropathic Pain in Chronic Pancreatitis. THE JOURNAL OF PAIN 2023; 24:2199-2210. [PMID: 37451493 PMCID: PMC10787046 DOI: 10.1016/j.jpain.2023.07.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 07/03/2023] [Accepted: 07/07/2023] [Indexed: 07/18/2023]
Abstract
Debilitating abdominal pain is a common symptom affecting most patients with chronic pancreatitis (CP). There are multiple underlying mechanisms that contribute to CP-related pain, which makes successful treatment difficult. The identification of biomarkers for subtypes of pain could provide viable targets for nonopioid interventions and the development of mechanistic approaches to pain management in CP. Nineteen inflammation- and nociception-associated proteins were measured in serum collected from 358 subjects with definite CP enrolled in PROspective Evaluation of Chronic Pancreatitis for EpidEmiologic and Translational StuDies, a prospective observational study of pancreatitis in US adult subjects. First, serum levels of putative biomarkers were compared between CP subjects with and without pain. Only platelet-derived growth factor B (PDGF-B) stood out, with levels significantly higher in the CP pain group as compared to subjects with no pain. Subjects with pain were then stratified into 4 pain subtypes (Neuropathic, Nociceptive, Mixed, and Unclassified). A comparison of putative biomarker concentration among 5 groups (no pain and 4 pain subtypes) identified unique proteins that were correlated with pain subtypes. Serum transforming growth factor beta 1 (TGFβ1) level was significantly higher in the Nociceptive pain group compared to the No pain group, suggesting that TGFβ1 may be a biomarker for nociceptive pain. The Neuropathic pain only group was too small to detect statistical differences. However, glycoprotein 130 (GP130), a coreceptor for interleukin 6, was significantly higher in the Mixed pain group compared to the groups lacking a neuropathic pain component. These data suggest that GP130 may be a biomarker for neuropathic pain in CP. PERSPECTIVE: Serum TGFβ1 and GP130 may be biomarkers for nociceptive and neuropathic CP pain, respectively. Preclinical data suggest inhibiting TGFβ1 or GP130 reduces CP pain in rodent models, indicating that additional translational and clinical studies may be warranted to develop a precision medicine approach to the management of pain in CP.
Collapse
Affiliation(s)
- Jami L. Saloman
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, School of Medicine, University of Pittsburgh, PA, USA
- Pittsburgh Center for Pain Research, School of Medicine, University of Pittsburgh, PA, USA
- Department of Neurobiology, School of Medicine, University of Pittsburgh, PA, USA
| | - Yan Li
- Department of Biostatistics, M.D. Anderson Cancer Center, Houston, TX, USA
| | - Kimberly Stello
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, School of Medicine, University of Pittsburgh, PA, USA
| | - Wenhao Li
- Department of Biostatistics, M.D. Anderson Cancer Center, Houston, TX, USA
| | - Shuang Li
- Department of Biostatistics, M.D. Anderson Cancer Center, Houston, TX, USA
| | - Anna Evans Phillips
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, School of Medicine, University of Pittsburgh, PA, USA
| | - Kristen Hall
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, School of Medicine, University of Pittsburgh, PA, USA
| | - Evan L. Fogel
- Digestive and Liver Disorders, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
| | | | - Liang Li
- Department of Biostatistics, M.D. Anderson Cancer Center, Houston, TX, USA
| | - Dana K. Andersen
- Division of Digestive Diseases and Nutrition, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | - William E. Fisher
- Division of General Surgery, Baylor College of Medicine, Houston, TX
| | - Christopher E. Forsmark
- Division of Gastroenterology, Hepatology, and Nutrition. University of Florida, Gainesville, FL
| | - Phil A. Hart
- Division of Gastroenterology, Hepatology, and Nutrition, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Stephen J. Pandol
- Division of Digestive and Liver Diseases, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Walter G. Park
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | - Mark D. Topazian
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | | | - Jose Serrano
- Division of Digestive Diseases and Nutrition, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | - Darwin L. Conwell
- Department of Internal Medicine, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Dhiraj Yadav
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, School of Medicine, University of Pittsburgh, PA, USA
| |
Collapse
|
14
|
Maldonado-García JL, Pérez-Sánchez G, Becerril-Villanueva E, Alvarez-Herrera S, Pavón L, Sánchez-Torres L, Gutiérrez-Ospina G, Girón-Pérez MI, Damian-Morales G, Maldonado-Tapia JO, López-Santiago R, Moreno-Lafont MC. Imipramine Administration in Brucella abortus 2308-Infected Mice Restores Hippocampal Serotonin Levels, Muscle Strength, and Mood, and Decreases Spleen CFU Count. Pharmaceuticals (Basel) 2023; 16:1525. [PMID: 38004391 PMCID: PMC10674296 DOI: 10.3390/ph16111525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/25/2023] [Accepted: 09/28/2023] [Indexed: 11/26/2023] Open
Abstract
Brucellosis infection causes non-specific symptoms such as fever, chills, sweating, headaches, myalgia, arthralgia, anorexia, fatigue, and mood disorders. In mouse models, it has been associated with increased levels of IL-6, TNF-α, and IFN-γ, a decrease in serotonin and dopamine levels within the hippocampus, induced loss of muscle strength and equilibrium, and increased anxiety and hopelessness. Imipramine (ImiP), a tricyclic antidepressant, is used to alleviate neuropathic pain. This study evaluated the effects of ImiP on Balb/c mice infected with Brucella abortus 2308 (Ba) at 14- and 28-days post-infection. Serum levels of six cytokines (IFN-γ, IL-6, TNF-α, IL-12, MCP-1. and IL-10) were assessed by FACS, while the number of bacteria in the spleen was measured via CFU. Serotonin levels in the hippocampus were analyzed via HPLC, and behavioral tests were conducted to assess strength, equilibrium, and mood. Our results showed that mice infected with Brucella abortus 2308 and treated with ImiP for six days (Im6Ba14) had significantly different outcomes compared to infected mice (Ba14) at day 14 post-infection. The mood was enhanced in the forced swimming test (FST) (p < 0.01), tail suspension test (TST) (p < 0.0001), and open-field test (p < 0.0001). Additionally, there was an increase in serotonin levels in the hippocampus (p < 0.001). Furthermore, there was an improvement in equilibrium (p < 0.0001) and muscle strength (p < 0.01). Lastly, there was a decrease in IL-6 levels (p < 0.05) and CFU count in the spleen (p < 0.0001). At 28 days, infected mice that received ImiP for 20 days (Im20Ba28) showed preservation of positive effects compared to infected mice (Ba28). These effects include the following: (1) improved FST (p < 0.0001) and TST (p < 0.0001); (2) better equilibrium (p < 0.0001) and muscle strength (p < 0.0001); (3) decreased IL-6 levels (p < 0.05); and (4) reduced CFU count in the spleen (p < 0.0001). These findings suggest the potential for ImiP to be used as an adjuvant treatment for the symptoms of brucellosis, which requires future studies.
Collapse
Affiliation(s)
- José Luis Maldonado-García
- Laboratorio de Psicoinmunología, Dirección de Investigaciones en Neurociencias del Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico
- Laboratorio de Inmunología Celular, Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 07738, Mexico
| | - Gilberto Pérez-Sánchez
- Laboratorio de Psicoinmunología, Dirección de Investigaciones en Neurociencias del Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico
| | - Enrique Becerril-Villanueva
- Laboratorio de Psicoinmunología, Dirección de Investigaciones en Neurociencias del Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico
| | - Samantha Alvarez-Herrera
- Laboratorio de Psicoinmunología, Dirección de Investigaciones en Neurociencias del Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico
| | - Lenin Pavón
- Laboratorio de Psicoinmunología, Dirección de Investigaciones en Neurociencias del Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico
| | - Luvia Sánchez-Torres
- Laboratorio de Inmunología de los Microorganismos, Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 07738, Mexico
| | - Gabriel Gutiérrez-Ospina
- Laboratorio de Biología de Sistemas, Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas y Coordinación de Psicobiología y Neurociencias, Facultad de Psicología, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | | | - Gabriela Damian-Morales
- Laboratorio de Inmunología Celular, Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 07738, Mexico
| | - Jesús Octavio Maldonado-Tapia
- Laboratorio de Psicoinmunología, Dirección de Investigaciones en Neurociencias del Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 14370, Mexico
- Laboratorio de Inmunología Celular, Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 07738, Mexico
| | - Rubén López-Santiago
- Laboratorio de Inmunología Celular, Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 07738, Mexico
| | - Martha C Moreno-Lafont
- Laboratorio de Inmunología Celular, Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 07738, Mexico
| |
Collapse
|
15
|
Smith PA. Neuropathic pain; what we know and what we should do about it. FRONTIERS IN PAIN RESEARCH 2023; 4:1220034. [PMID: 37810432 PMCID: PMC10559888 DOI: 10.3389/fpain.2023.1220034] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 09/05/2023] [Indexed: 10/10/2023] Open
Abstract
Neuropathic pain can result from injury to, or disease of the nervous system. It is notoriously difficult to treat. Peripheral nerve injury promotes Schwann cell activation and invasion of immunocompetent cells into the site of injury, spinal cord and higher sensory structures such as thalamus and cingulate and sensory cortices. Various cytokines, chemokines, growth factors, monoamines and neuropeptides effect two-way signalling between neurons, glia and immune cells. This promotes sustained hyperexcitability and spontaneous activity in primary afferents that is crucial for onset and persistence of pain as well as misprocessing of sensory information in the spinal cord and supraspinal structures. Much of the current understanding of pain aetiology and identification of drug targets derives from studies of the consequences of peripheral nerve injury in rodent models. Although a vast amount of information has been forthcoming, the translation of this information into the clinical arena has been minimal. Few, if any, major therapeutic approaches have appeared since the mid 1990's. This may reflect failure to recognise differences in pain processing in males vs. females, differences in cellular responses to different types of injury and differences in pain processing in humans vs. animals. Basic science and clinical approaches which seek to bridge this knowledge gap include better assessment of pain in animal models, use of pain models which better emulate human disease, and stratification of human pain phenotypes according to quantitative assessment of signs and symptoms of disease. This can lead to more personalized and effective treatments for individual patients. Significance statement: There is an urgent need to find new treatments for neuropathic pain. Although classical animal models have revealed essential features of pain aetiology such as peripheral and central sensitization and some of the molecular and cellular mechanisms involved, they do not adequately model the multiplicity of disease states or injuries that may bring forth neuropathic pain in the clinic. This review seeks to integrate information from the multiplicity of disciplines that seek to understand neuropathic pain; including immunology, cell biology, electrophysiology and biophysics, anatomy, cell biology, neurology, molecular biology, pharmacology and behavioral science. Beyond this, it underlines ongoing refinements in basic science and clinical practice that will engender improved approaches to pain management.
Collapse
Affiliation(s)
- Peter A. Smith
- Neuroscience and Mental Health Institute and Department of Pharmacology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
16
|
Liu H, Wang D, Wu F, Dong Z, Yu S. Association between inflammatory potential of diet and self-reported severe headache or migraine: A cross-sectional study of the National Health and Nutrition Examination Survey. Nutrition 2023; 113:112098. [PMID: 37356381 DOI: 10.1016/j.nut.2023.112098] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 03/01/2023] [Accepted: 05/22/2023] [Indexed: 06/27/2023]
Abstract
OBJECTIVE Inflammation plays an important role in migraine development. Dietary interventions that reduce the inflammatory state are effective for migraine prophylaxis. However, little is yet known about the association between the inflammatory potential of diet and migraine in the general US adult population. The aim of this study was to evaluate whether the inflammatory potential of diet is associated with severe headache or migraine in US adults. METHODS This cross-sectional study analyzed data from adult participants in the 1999-2004 National Health and Nutrition Examination Survey. Severe headache or migraine was determined based on self-reported information. Severe headache or migraine was defined as an affirmative response to the question: "Have you had a severe headache or migraine in the past 3 mo?" The dietary inflammatory index (DII) was used to assess the inflammatory potential of the diet. Multivariable logistic regression models were used to determine the odds ratio (OR) and 95 % confidence interval (95% CI) for the association between DII and severe headache or migraine. A multivariable-adjusted restricted cubic spline model was constructed to establish the OR curves at 3 knots to examine the possible non-linear dose-response association between DII and severe headache or migraine. RESULTS Of 10359 participants included in this study (mean age, 46.3 [0.3] y; 5116 [49%] men), 2083 (22%) reported previous episodes of severe headache or migraine, whereas 8276 (78%) did not. After adjusting for potential confounders, the DII score was associated with severe headache or migraine (odds ratio [OR], 1.09; 95% confidence interval [CI], 1.03-1.15; P = 0.003). Compared with participants with the lowest tertile (T1) of DII score (i.e., T1; ≤-4.42 to ≤0.77), those with the highest tertile (i.e., T3; <2.44 to ≤5.18) had an adjusted OR for severe headache or migraine of 1.46 (95% CI, 1.20-1.77; P < 0.001). The multivariable restricted cubic spline showed a non-linear association between DII and severe headache or migraine (P = 0.012). In two piecewise regression models, the adjusted OR of developing a severe headache or migraine was 1.19 (95% CI, 1.04-1.37; P = 0.012) in participants with a DII score ≥1.48, whereas there was no association between DII and severe headache or migraine in participants with a DII score <1.48. Subgroup analyses showed that DII was associated with severe headache or migraine in women (OR, 1.73; 95% CI, 1.31-2.28), individuals 20 to 50 y of age (OR, 1.66; 95% CI, 1.29-2.14), those who were married or living with a partner (OR, 1.49; 95% CI, 1.19-1.85), individuals living alone (OR, 1.36; 95% CI, 1.02-1.81), those with a high level of education (OR, 1.72; 95% CI, 1.26-2.36), individuals with a low family income (OR, 1.58; 95% CI, 1.10-2.28), those with a medium or high family income (OR, 1.48; 95% CI, 1.17-1.86), and body mass index <25 kg/m2 (OR, 1.71; 95% CI, 1.21-2.41). CONCLUSION The inflammatory potential of the diet is associated with severe headache or migraine in US adults.
Collapse
Affiliation(s)
- Huanxian Liu
- Department of Neurology, First Medical Center of Chinese PLA General Hospital, Beijing, China.
| | - Dan Wang
- Department of Neurology, General Hospital of Northern Theater Command, Shenyang, China.
| | - Fei Wu
- Department of Neurology, General Hospital of Chinese PLA General Central Theater Command, Wuhan, China.
| | - Zhao Dong
- Department of Neurology, First Medical Center of Chinese PLA General Hospital, Beijing, China; International Headache Center, Chinese PLA General Hospital, Beijing, China.
| | - Shengyuan Yu
- Department of Neurology, First Medical Center of Chinese PLA General Hospital, Beijing, China; International Headache Center, Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
17
|
Hernandez LM, Montersino A, Niu J, Guo S, Faezov B, Sanders SS, Dunbrack RL, Thomas GM. Palmitoylation-dependent control of JAK1 kinase signaling governs responses to neuropoietic cytokines and survival in DRG neurons. J Biol Chem 2023; 299:104965. [PMID: 37356718 PMCID: PMC10413081 DOI: 10.1016/j.jbc.2023.104965] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 06/11/2023] [Accepted: 06/13/2023] [Indexed: 06/27/2023] Open
Abstract
Janus Kinase-1 (JAK1) plays key roles during neurodevelopment and following neuronal injury, while activatory JAK1 mutations are linked to leukemia. In mice, Jak1 genetic deletion results in perinatal lethality, suggesting non-redundant roles and/or regulation of JAK1 for which other JAKs cannot compensate. Proteomic studies reveal that JAK1 is more likely palmitoylated compared to other JAKs, implicating palmitoylation as a possible JAK1-specific regulatory mechanism. However, the importance of palmitoylation for JAK1 signaling has not been addressed. Here, we report that JAK1 is palmitoylated in transfected HEK293T cells and endogenously in cultured Dorsal Root Ganglion (DRG) neurons. We further use comprehensive screening in transfected non-neuronal cells and shRNA-mediated knockdown in DRG neurons to identify the related enzymes ZDHHC3 and ZDHHC7 as dominant protein acyltransferases (PATs) for JAK1. Surprisingly, we found palmitoylation minimally affects JAK1 localization in neurons, but is critical for JAK1's kinase activity in cells and even in vitro. We propose this requirement is likely because palmitoylation facilitates transphosphorylation of key sites in JAK1's activation loop, a possibility consistent with structural models of JAK1. Importantly, we demonstrate a leukemia-associated JAK1 mutation overrides the palmitoylation-dependence of JAK1 activity, potentially explaining why this mutation is oncogenic. Finally, we show that JAK1 palmitoylation is important for neuropoietic cytokine-dependent signaling and neuronal survival and that combined Zdhhc3/7 loss phenocopies loss of palmitoyl-JAK1. These findings provide new insights into the control of JAK signaling in both physiological and pathological contexts.
Collapse
Affiliation(s)
- Luiselys M Hernandez
- Shriners Hospitals Pediatric Research Center (Center for Neurorehabilitation and Neural Repair), Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | - Audrey Montersino
- Shriners Hospitals Pediatric Research Center (Center for Neurorehabilitation and Neural Repair), Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | - Jingwen Niu
- Shriners Hospitals Pediatric Research Center (Center for Neurorehabilitation and Neural Repair), Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | - Shuchi Guo
- Shriners Hospitals Pediatric Research Center (Center for Neurorehabilitation and Neural Repair), Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | - Bulat Faezov
- Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA; Kazan Federal University, Kazan, Russian Federation
| | - Shaun S Sanders
- Shriners Hospitals Pediatric Research Center (Center for Neurorehabilitation and Neural Repair), Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | - Roland L Dunbrack
- Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
| | - Gareth M Thomas
- Shriners Hospitals Pediatric Research Center (Center for Neurorehabilitation and Neural Repair), Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA; Department of Neural Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
18
|
Stover JD, Trone MA, Lawrence B, Bowles RD. Multiplex epigenome editing of ion channel expression in nociceptive neurons abolished degenerative IVD-conditioned media-induced mechanical sensitivity. JOR Spine 2023; 6:e1253. [PMID: 37361323 PMCID: PMC10285767 DOI: 10.1002/jsp2.1253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 01/15/2023] [Accepted: 02/27/2023] [Indexed: 06/28/2023] Open
Abstract
Background Low back pain is a major contributor to disability worldwide and generates a tremendous socioeconomic impact. The degenerative intervertebral disc (IVD) has been hypothesized to contribute to discogenic pain by sensitizing nociceptive neurons innervating the disc to stimuli that is nonpainful in healthy patients. Previously, we demonstrated the ability of degenerative IVDs to sensitize neurons to mechanical stimuli; however, elucidation of degenerative IVDs discogenic pain mechanisms is required to develop therapeutic strategies that directly target these mechanisms. Aims In this study, we utilized CRISPR epigenome editing of nociceptive neurons to identify mechanisms of degenerative IVD-induced changes to mechanical nociception and demonstrated the ability of multiplex CRISPR epigenome editing of nociceptive neurons to modulate inflammation-induced mechanical nociception. Methods and Results Utilizing an in vitro model, we demonstrated degenerative IVD-produced IL-6-induced increases in nociceptive neuron activity in response to mechanical stimuli, mediated by TRPA1, ASIC3, and Piezo2 ion channel activity. Once these ion channels were identified as mediators of degenerative IVD-induced mechanical nociception, we developed singleplex and multiplex CRISPR epigenome editing vectors that modulate endogenous expression of TRPA1, ASIC3, and Piezo2 via targeted gene promoter histone methylation. When delivered to nociceptive neurons, the multiplex CRISPR epigenome editing vectors abolished degenerative IVD-induced mechanical nociception while preserving nonpathologic neuron activity. Conclusion This work demonstrates the potential of multiplex CRISPR epigenome editing as a highly targeted gene-based neuromodulation strategy for the treatment of discogenic pain, specifically; and, for the treatment of inflammatory chronic pain conditions, more broadly.
Collapse
Affiliation(s)
- Joshua D. Stover
- Department of BioengineeringUniversity of UtahSalt Lake CityUtahUSA
| | - Matthew A. Trone
- Department of BioengineeringUniversity of UtahSalt Lake CityUtahUSA
| | - Brandon Lawrence
- Department of OrthopaedicsUniversity of UtahSalt Lake CityUtahUSA
| | - Robby D. Bowles
- Department of BioengineeringUniversity of UtahSalt Lake CityUtahUSA
- Department of OrthopaedicsUniversity of UtahSalt Lake CityUtahUSA
| |
Collapse
|
19
|
Yamakita S, Fujita D, Sudo K, Ishikawa D, Kushimoto K, Horii Y, Amaya F. Activation of neurons and satellite glial cells in the DRG produces morphine-induced hyperalgesia. Mol Pain 2023; 19:17448069231181973. [PMID: 37254240 PMCID: PMC10291868 DOI: 10.1177/17448069231181973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 05/18/2023] [Accepted: 05/29/2023] [Indexed: 06/01/2023] Open
Abstract
Activation of neurons and glial cells in the dorsal root ganglion is one of the key mechanisms for the development of hyperalgesia. The aim of the present study was to examine the role of neuroglial activity in the development of opioid-induced hyperalgesia. Male rats were treated with morphine daily for 3 days. The resultant phosphorylation of extracellular signal-regulated kinase (ERK) 1/2 in the dorsal root ganglion was analyzed by immunohistochemistry and Western blotting. Pain hypersensitivity was analyzed using behavioral studies. The amount of cytokine expression in the dorsal root ganglion was also analyzed. Repeated morphine treatment induced hyperalgesia and marked induction of phosphorylated ERK1/2 in the neurons and satellite glial cells on day 3. An opioid receptor antagonist, toll like receptor-4 inhibitor, MAP/ERK kinase (MEK) inhibitor and gap junction inhibitor inhibited morphine-induced hyperalgesia and ERK1/2 phosphorylation. Morphine treatment induced alteration of cytokine expression, which was inhibited by the opioid receptor antagonist, toll like receptor-4 inhibitor, MEK inhibitor and gap junction inhibitor. Dexamethasone inhibited morphine-induced hyperalgesia and ERK1/2 phosphorylation after morphine treatment. The peripherally restricted opioid receptor antagonist, methylnaltrexone, inhibited hyperalgesia and ERK1/2 phosphorylation. Morphine activates ERK1/2 in neurons and satellite glial cells in the dorsal root ganglion via the opioid receptor and toll like receptor-4. ERK1/2 phosphorylation is gap junction-dependent and is associated with the alteration of cytokine expression. Inhibition of neuroinflammation by activation of neurons and glia might be a promising target to prevent opioid-induced hyperalgesia.
Collapse
Affiliation(s)
- Shunsuke Yamakita
- Department of Anesthesiology, institution-id-type="Ringgold" />Kyoto Prefectural University of Medicine, Kyoto, Japan
- Research Unit for the Neurobiology of Pain, institution-id-type="Ringgold" />Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Daisuke Fujita
- Department of Anesthesiology, institution-id-type="Ringgold" />Kyoto Prefectural University of Medicine, Kyoto, Japan
- Research Unit for the Neurobiology of Pain, institution-id-type="Ringgold" />Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kazuki Sudo
- Department of Anesthesiology, institution-id-type="Ringgold" />Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Daiki Ishikawa
- Department of Anesthesiology, institution-id-type="Ringgold" />Kyoto Prefectural University of Medicine, Kyoto, Japan
- Research Unit for the Neurobiology of Pain, institution-id-type="Ringgold" />Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kohsuke Kushimoto
- Department of Anesthesiology, institution-id-type="Ringgold" />Kyoto Prefectural University of Medicine, Kyoto, Japan
- Research Unit for the Neurobiology of Pain, institution-id-type="Ringgold" />Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yasuhiko Horii
- Department of Anesthesiology, institution-id-type="Ringgold" />Kyoto Prefectural University of Medicine, Kyoto, Japan
- Research Unit for the Neurobiology of Pain, institution-id-type="Ringgold" />Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Fumimasa Amaya
- Research Unit for the Neurobiology of Pain, institution-id-type="Ringgold" />Kyoto Prefectural University of Medicine, Kyoto, Japan
- Department of Pain Management and Palliative Care Medicine, institution-id-type="Ringgold" />Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
20
|
Zheng B, Li S, Xiang Y, Zong W, Ma Q, Wang S, Wu H, Song H, Ren H, Chen J, Liu J, Zhao F. Netrin-1 mediates nerve innervation and angiogenesis leading to discogenic pain. J Orthop Translat 2022; 39:21-33. [PMID: 36605621 PMCID: PMC9804017 DOI: 10.1016/j.jot.2022.11.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 09/16/2022] [Accepted: 11/28/2022] [Indexed: 12/24/2022] Open
Abstract
Objective Discogenic low back pain (LBP) is associated with nociceptive nerve fibers that grow into degenerated intervertebral discs (IVD) but the etiopathogenesis of disease is not fully understood. The purpose of this study was to clarify the role of Netrin-1 in causing discogenic LBP. Methods The level of nociceptive nerve innervation was examined in disc degenerative patients and rat needle-punctured models by immunohistochemistry. Nucleus pulposus (NP) cells were isolated from IVD tissues of rats and induced degeneration by interleukin-1β (IL-1β) or tumor necrosis factor α (TNFα). The candidate genes related to neuron outgrowth and migration were selected by Next-generation sequencing (NGS). CRISPR/Cas9 was used to knockdown Netrin-1 in NP cells. The impact of Netrin-1 on nerve innervation were evaluated with P2X2、NF200 staining and microfluidics assay. Meanwhile the CD31 staining and transwell assay were used to evaluate the impact of Netrin-1 in angiogenesis. The proteins and RNA extracted from NP cells related to catabolism and anabolism were examined by western blot assay and RT-qPCR experiment. ChIP and luciferase experiments were used to assess the intracellular transcriptional regulation of Netrin-1. Further, a needle-punctured rat model followed by histomorphometry and immunofluorescence histochemistry was used to explore the potential effect of Netrin-1 on LBP in vivo. Results The level of nerve innervation was increased in severe disc degenerative patients while the expression of Netrin-1 was upregulated. The supernatants of NP cells stimulated with IL-1β or TNFα containing more Netrin-1 could promote axon growth and vascular endothelial cells migration. Knocking down Netrin-1 or overexpressing transcription factor TCF3 as a negative regulator of Netrin-1 attenuated this effect. The needle-punctured rat model brought significant spinal hypersensitivity, nerve innervation and angiogenesis, nevertheless knocking down Netrin-1 effectively prevented disc degeneration-induced adverse impacts. Conclusion Discogenic LBP was induced by Netrin-1, which mediated nerve innervation and angiogenesis in disc degeneration. Knocking down Netrin-1 by CRISPR/Cas9 or negatively regulating Netrin1 by transcription factor TCF3 could alleviate spinal hypersensitivity. The translational potential of this article This study on Netrin-1 could provide a new target and theoretical basis for the prevention and treatment for discogenic back pain.
Collapse
Affiliation(s)
- Bingjie Zheng
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310027, China,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, Zhejiang, 310027, China
| | - Shengwen Li
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, 1055 Sanxiang Road, 215003, Suzhou, Jiangsu, China,Second Department of Orthopaedics Haining People's Hospital, Jiaxing, Zhejiang, 314400, China
| | - Yufeng Xiang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310027, China,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, Zhejiang, 310027, China
| | - Wentian Zong
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310027, China,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, Zhejiang, 310027, China
| | - Qingliang Ma
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310027, China,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, Zhejiang, 310027, China
| | - Shiyu Wang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310027, China,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, Zhejiang, 310027, China
| | - Haihao Wu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310027, China,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, Zhejiang, 310027, China,Department of Orthopedics, Hwa Mei Hospital, University of Chinese Academy of Sciences, No. 41 Northwest Street, Ningbo, Zhejiang, 315010, China
| | - Haixin Song
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310027, China,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, Zhejiang, 310027, China
| | - Hong Ren
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310027, China
| | - Jian Chen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310027, China,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, Zhejiang, 310027, China
| | - Junhui Liu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310027, China,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, Zhejiang, 310027, China,Corresponding author. Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310027, China.
| | - Fengdong Zhao
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310027, China,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, Zhejiang, 310027, China,Corresponding author. Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310027, China.
| |
Collapse
|
21
|
Sawicki CM, Janal MN, Nicholson SJ, Wu AK, Schmidt BL, Albertson DG. Oral cancer patients experience mechanical and chemical sensitivity at the site of the cancer. BMC Cancer 2022; 22:1165. [PMID: 36368973 PMCID: PMC9650819 DOI: 10.1186/s12885-022-10282-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 11/04/2022] [Indexed: 11/13/2022] Open
Abstract
INTRODUCTION Oral cancer patients suffer severe chronic and mechanically-induced pain at the site of the cancer. Our clinical experience is that oral cancer patients report new sensitivity to spicy foods. We hypothesized that in cancer patients, mechanical and chemical sensitivity would be greater when measured at the cancer site compared to a contralateral matched normal site. METHODS We determined mechanical pain thresholds (MPT) on the right and left sides of the tongue of 11 healthy subjects, and at the cancer and contralateral matched normal site in 11 oral cancer patients in response to von Frey filaments in the range of 0.008 to 300 g (normally not reported as painful). We evaluated chemical sensitivity in 13 healthy subjects and seven cancer patients, who rated spiciness/pain on a visual analog scale in response to exposure to six paper strips impregnated with capsaicin (0-10 mM). RESULTS Mechanical detection thresholds (MDT) were recorded for healthy subjects, but not MPTs. By contrast, MPTs were measured at the site of the cancer in oral cancer patients (7/11 patients). No MPTs were measured at the cancer patients' contralateral matched normal sites. Measured MPTs were correlated with patients' responses to the University of California Oral Cancer Pain Questionnaire. Capsaicin sensitivity at the site of the cancer was evident in cancer patients by a leftward shift of the cancer site capsaicin dose-response curve compared to that of the patient's contralateral matched normal site. We detected no difference in capsaicin sensitivity on the right and left sides of tongues of healthy subjects. CONCLUSIONS Mechanical and chemical sensitivity testing was well tolerated by the majority of oral cancer patients. Sensitivity is greater at the site of the cancer than at a contralateral matched normal site.
Collapse
Affiliation(s)
- Caroline M. Sawicki
- grid.137628.90000 0004 1936 8753Department of Pediatric Dentistry, New York University College of Dentistry, 421 First Avenue, Room 233W, New York, NY 10010 USA
| | - Malvin N. Janal
- grid.137628.90000 0004 1936 8753Department of Epidemiology & Health Promotion, New York University College of Dentistry, Room 301, 433 First Avenue, New York, NY 10010 USA
| | - Samuel J. Nicholson
- grid.137628.90000 0004 1936 8753Department of Oral and Maxillofacial Surgery, New York University College of Dentistry, 421 First Avenue, Room 233W, New York, NY 10010 USA
| | - Angie K. Wu
- grid.137628.90000 0004 1936 8753Bluestone Center for Clinical Research, New York University College of Dentistry, 421 First Avenue, Room 233W, New York, NY 10010 USA
| | - Brian L. Schmidt
- grid.137628.90000 0004 1936 8753Department of Oral and Maxillofacial Surgery, New York University College of Dentistry, 421 First Avenue, Room 233W, New York, NY 10010 USA ,grid.137628.90000 0004 1936 8753Bluestone Center for Clinical Research, New York University College of Dentistry, 421 First Avenue, Room 233W, New York, NY 10010 USA ,grid.137628.90000 0004 1936 8753NYU Oral Cancer Center, New York University College of Dentistry, 421 First Avenue, Room 233W, New York, NY 10010 USA
| | - Donna G. Albertson
- grid.137628.90000 0004 1936 8753Department of Oral and Maxillofacial Surgery, New York University College of Dentistry, 421 First Avenue, Room 233W, New York, NY 10010 USA ,grid.137628.90000 0004 1936 8753Bluestone Center for Clinical Research, New York University College of Dentistry, 421 First Avenue, Room 233W, New York, NY 10010 USA ,grid.137628.90000 0004 1936 8753NYU Oral Cancer Center, New York University College of Dentistry, 421 First Avenue, Room 233W, New York, NY 10010 USA
| |
Collapse
|
22
|
Proinflammatory cytokines and their receptors as druggable targets to alleviate pathological pain. Pain 2022; 163:S79-S98. [DOI: 10.1097/j.pain.0000000000002737] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 07/14/2022] [Indexed: 02/07/2023]
|
23
|
Dubeykovskaya ZA, Tu NH, Garcia PDR, Schmidt BL, Albertson DG. Oral Cancer Cells Release Vesicles that Cause Pain. Adv Biol (Weinh) 2022; 6:e2200073. [PMID: 35802912 PMCID: PMC9474716 DOI: 10.1002/adbi.202200073] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/02/2022] [Indexed: 01/28/2023]
Abstract
Oral cancer pain is attributed to the release from cancers of mediators that sensitize and activate sensory neurons. Intraplantar injection of conditioned media (CM) from human tongue cancer cell line HSC-3 or OSC-20 evokes nociceptive behavior. By contrast, CM from noncancer cell lines, DOK, and HaCaT are non-nociceptive. Pain mediators are carried by extracellular vesicles (EVs) released from cancer cells. Depletion of EVs from cancer cell line CM reverses mechanical allodynia and thermal hyperalgesia. CM from non-nociceptive cell lines become nociceptive when reconstituted with HSC-3 EVs. Two miRNAs (hsa-miR-21-5p and hsa-miR-221-3p) are identified that are present in increased abundance in EVs from HSC-3 and OSC-20 CM compared to HaCaT CM. The miRNA target genes suggest potential involvement in oral cancer pain of the toll like receptor 7 (TLR7) and 8 (TLR8) pathways, as well as signaling through interleukin 6 cytokine family signal transducer receptor (gp130, encoded by IL6ST) and colony stimulating factor receptor (G-CSFR, encoded by CSF3R), Janus kinase and signal transducer and activator of transcription 3 (JAK/STAT3). These studies confirm the recent discovery of the role of cancer EVs in pain and add to the repertoire of algesic and analgesic cancer pain mediators and pathways that contribute to oral cancer pain.
Collapse
Affiliation(s)
- Zinaida A Dubeykovskaya
- Bluestone Center for Clinical Research and Department of Oral and Maxillofacial Surgery, New York University College of Dentistry, New York, NY, 10010, USA
| | - Nguyen Huu Tu
- Bluestone Center for Clinical Research and Department of Oral and Maxillofacial Surgery, New York University College of Dentistry, New York, NY, 10010, USA
| | - Paulina D Ramírez Garcia
- Bluestone Center for Clinical Research and Department of Oral and Maxillofacial Surgery, New York University College of Dentistry, New York, NY, 10010, USA
| | - Brian L Schmidt
- Bluestone Center for Clinical Research and Department of Oral and Maxillofacial Surgery, New York University College of Dentistry, New York, NY, 10010, USA
| | - Donna G Albertson
- Bluestone Center for Clinical Research and Department of Oral and Maxillofacial Surgery, New York University College of Dentistry, New York, NY, 10010, USA
| |
Collapse
|
24
|
Liao Y, Ren Y, Luo X, Mirando AJ, Long JT, Leinroth A, Ji RR, Hilton MJ. Interleukin-6 signaling mediates cartilage degradation and pain in posttraumatic osteoarthritis in a sex-specific manner. Sci Signal 2022; 15:eabn7082. [PMID: 35881692 PMCID: PMC9382892 DOI: 10.1126/scisignal.abn7082] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Osteoarthritis (OA) and posttraumatic OA (PTOA) are caused by an imbalance in catabolic and anabolic processes in articular cartilage and proinflammatory changes throughout the joint, leading to joint degeneration and pain. We examined whether interleukin-6 (IL-6) signaling contributed to cartilage degradation and pain in PTOA. Genetic ablation of Il6 in male mice decreased PTOA-associated cartilage catabolism, innervation of the knee joint, and nociceptive signaling without improving PTOA-associated subchondral bone sclerosis or chondrocyte apoptosis. These effects were not observed in female Il6-/- mice. Compared with wild-type mice, the activation of the IL-6 downstream mediators STAT3 and ERK was reduced in the knees and dorsal root ganglia (DRG) of male Il6-/- mice after knee injury. Janus kinases (JAKs) were critical for STAT and ERK signaling in cartilage catabolism and DRG pain signaling in tissue explants. Whereas STAT3 signaling was important for cartilage catabolism, ERK signaling mediated neurite outgrowth and the activation of nociceptive neurons. These data demonstrate that IL-6 mediates both cartilage degradation and pain associated with PTOA in a sex-specific manner and identify tissue-specific contributions of downstream effectors of IL-6 signaling, which are potential therapeutic targets for disease-modifying OA drugs.
Collapse
Affiliation(s)
- Yihan Liao
- Departments of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA,Departments of Orthopaedic Surgery, Duke Orthopaedic Cellular, Developmental, and Genome Laboratories, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Yinshi Ren
- Departments of Orthopaedic Surgery, Duke Orthopaedic Cellular, Developmental, and Genome Laboratories, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Xin Luo
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Anthony J. Mirando
- Departments of Orthopaedic Surgery, Duke Orthopaedic Cellular, Developmental, and Genome Laboratories, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Jason T. Long
- Departments of Orthopaedic Surgery, Duke Orthopaedic Cellular, Developmental, and Genome Laboratories, Duke University School of Medicine, Durham, NC, 27710, USA,Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Abigail Leinroth
- Departments of Orthopaedic Surgery, Duke Orthopaedic Cellular, Developmental, and Genome Laboratories, Duke University School of Medicine, Durham, NC, 27710, USA,Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Matthew J. Hilton
- Departments of Orthopaedic Surgery, Duke Orthopaedic Cellular, Developmental, and Genome Laboratories, Duke University School of Medicine, Durham, NC, 27710, USA,Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA,Corresponding author.
| |
Collapse
|
25
|
Leisengang S, Gluding D, Hörster J, Peek V, Ott D, Rummel C, Schmidt MJ. Expression of adipokines and adipocytokines by epidural adipose tissue in cauda equina syndrome in dogs. J Vet Intern Med 2022; 36:1373-1381. [PMID: 35838307 PMCID: PMC9308421 DOI: 10.1111/jvim.16483] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 06/15/2022] [Indexed: 11/28/2022] Open
Abstract
Background Compression of epidural adipose tissue (EAT) within the scope of cauda equina syndrome (CES) could lead to an enhanced expression of inflammatory mediators, possibly contributing to pain amplification in dogs. Objectives To analyze expression of inflammatory adipo(‐cyto)kines within the EAT of dogs with CES. Animals Client‐owned dogs: 15 dogs with CES and 9 dogs euthanized for unrelated medical reasons (controls). Methods Prospective, experimental study. Epidural adipose tissue and subcutaneous adipose tissue were collected during dorsal laminectomy and used for real‐time quantitative polymerase chain reaction. Tissue explants were cultured for measurements of inflammation‐induced release of cytokines. Results Results show a CES‐associated upregulation of the cytokines tumor necrosis factor alpha (TNFα: mean ± SD: 18.88 ± 11.87, 95% CI: 10.90‐26.86 vs 9.66 ± 5.22, 95% CI: 5.29‐14.02, *: P = .04) and interleukin‐ (IL‐) 10 (20.1 ± 9.15, 95% CI: 14.82‐25.39 vs 11.52 ± 6.82, 95% CI: 5.82‐17.22, *: P = .03), whereas the expression of the adipokine leptin was attenuated in EAT of dogs with CES (3.07 ± 2.29, 95% CI: 1.80‐3.34 vs 9.83 ± 8.42, 95% CI: 3.36‐16.30, **: P = .007). Inflammatory stimulation of EAT explant cultures resulted in an enhanced release of IL‐6 (LPS: 5491.55 ± 4438, 95% CI: 833.7‐10 149; HMGB1: 1001.78 ± 522.2, 95% CI: 518.8‐1485; PBS: 310.9 ± 98.57, 95% CI: 228.5‐393.3, ***: P < .001). Conclusion and Clinical Importance Expression profile of inflammatory adipo(‐cyto)kines by EAT is influenced from compressive forces acting in dogs with CES and might contribute to amplification of pain.
Collapse
Affiliation(s)
- Stephan Leisengang
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University Giessen, Giessen, Germany.,Center for Mind, Brain and Behavior - CMBB, Philipps University Marburg & Justus Liebig University Giessen, Giessen, Germany.,Institute of Medical Psychology and Behavioral Immunobiology, Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Dennis Gluding
- Department of Veterinary Clinical Sciences, Clinic for Small Animals (Surgery), Justus Liebig University Giessen, Giessen, Germany
| | - Julia Hörster
- Department of Veterinary Clinical Sciences, Small Animal Clinic - Neurosurgery, Neuroradiology and Clinical Neurology, Justus Liebig University Giessen, Giessen, Germany
| | - Verena Peek
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University Giessen, Giessen, Germany
| | - Daniela Ott
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University Giessen, Giessen, Germany
| | - Christoph Rummel
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University Giessen, Giessen, Germany.,Center for Mind, Brain and Behavior - CMBB, Philipps University Marburg & Justus Liebig University Giessen, Giessen, Germany
| | - Martin J Schmidt
- Department of Veterinary Clinical Sciences, Small Animal Clinic - Neurosurgery, Neuroradiology and Clinical Neurology, Justus Liebig University Giessen, Giessen, Germany
| |
Collapse
|
26
|
Liu NQ, Chen S, Geng D, Lei J, Zhang J, Li L, Lin Y, Ouyang Y, Shkhyan R, Van Handel B, Bian F, Mkaratigwa T, Chai Y, Evseenko D. Local Drug-Induced Modulation of gp130 Receptor Signaling Delays Disease Progression in a Pig Model of Temporo-Mandibular Joint Osteoarthritis. FRONTIERS IN DENTAL MEDICINE 2022. [DOI: 10.3389/fdmed.2022.937819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Temporomandibular joint disorders (TMJs) are a multifaceted group of chronic disorders characterized by stiffness in the jaw, limited jaw mobility and pain when opening or closing the mouth. TMJs are relatively common, with incidence rates in the range of 5–12%, with nearly twice as many women as men being affected. One of the primary causes of TMJs is a degenerative disease of joints, such as osteoarthritis (OA), characterized by progressive loss of cartilage which causes stiffness, swelling, and pain. Currently, there are no disease-modifying agents on the market for OA. We have recently discovered a small molecule, R805 acting as a modulator of glycoprotein 130 (gp130) receptor for IL-6 family of cytokines. R805 enables regenerative outputs of endogenous joint stem and progenitor cells through immunomodulation in the joint microenvironment by reducing the levels of destructive cytokines and supporting chondrocyte survival and anabolism. Extensive testing has shown R805 to be safe at doses far above the therapeutic level. Here, we have conducted a pivotal efficacy study in our newly-established pig model of TMJ post-traumatic OA. IA injection of R805 has shown a highly significant reduction of articular cartilage degeneration, reduced synovitis and degenerative changes in subchondral bone in the mandibular condyle compared to the vehicle-treated group. These data will support additional pre-clinical development of R805 as a first-in-class injectable therapeutic for TMJ osteoarthritis.
Collapse
|
27
|
Zheng Q, Dong X, Green DP, Dong X. Peripheral mechanisms of chronic pain. MEDICAL REVIEW 2022; 2:251-270. [PMID: 36067122 PMCID: PMC9381002 DOI: 10.1515/mr-2022-0013] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 06/13/2022] [Indexed: 11/15/2022]
Abstract
Abstract
Acutely, pain serves to protect us from potentially harmful stimuli, however damage to the somatosensory system can cause maladaptive changes in neurons leading to chronic pain. Although acute pain is fairly well controlled, chronic pain remains difficult to treat. Chronic pain is primarily a neuropathic condition, but studies examining the mechanisms underlying chronic pain are now looking beyond afferent nerve lesions and exploring new receptor targets, immune cells, and the role of the autonomic nervous system in contributing chronic pain conditions. The studies outlined in this review reveal how chronic pain is not only confined to alterations in the nervous system and presents findings on new treatment targets and for this debilitating disease.
Collapse
Affiliation(s)
- Qin Zheng
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Xintong Dong
- The Solomon H. Snyder Department of Neuroscience, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Dustin P. Green
- Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch, Galveston, TX, USA
| | - Xinzhong Dong
- The Solomon H. Snyder Department of Neuroscience, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
- Howard Hughes Medical Institute, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
28
|
Kalpachidou T, Malsch P, Qi Y, Mair N, Geley S, Quarta S, Kummer KK, Kress M. Genetic and functional evidence for gp130/IL6ST-induced transient receptor potential ankyrin 1 upregulation in uninjured but not injured neurons in a mouse model of neuropathic pain. Pain 2022; 163:579-589. [PMID: 34252913 PMCID: PMC8832546 DOI: 10.1097/j.pain.0000000000002402] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/27/2021] [Accepted: 06/30/2021] [Indexed: 11/26/2022]
Abstract
ABSTRACT Peripheral nerve injuries result in pronounced alterations in dorsal root ganglia, which can lead to the development of neuropathic pain. Although the polymodal mechanosensitive transient receptor potential ankyrin 1 (TRPA1) ion channel is emerging as a relevant target for potential analgesic therapies, preclinical studies do not provide unequivocal mechanistic insight into its relevance for neuropathic pain pathogenesis. By using a transgenic mouse model with a conditional depletion of the interleukin-6 (IL-6) signal transducer gp130 in Nav1.8 expressing neurons (SNS-gp130-/-), we provide a mechanistic regulatory link between IL-6/gp130 and TRPA1 in the spared nerve injury (SNI) model. Spared nerve injury mice developed profound mechanical hypersensitivity as indicated by decreased withdrawal thresholds in the von Frey behavioral test in vivo, as well as a significant increase in mechanosensitivity of unmyelinated nociceptive primary afferents in ex vivo skin-nerve recordings. In contrast to wild type and control gp130fl/fl animals, SNS-gp130-/- mice did not develop mechanical hypersensitivity after SNI and exhibited low levels of Trpa1 mRNA in sensory neurons, which were partially restored by adenoviral gp130 re-expression in vitro. Importantly, uninjured but not injured neurons developed increased responsiveness to the TRPA1 agonist cinnamaldehyde, and neurons derived from SNS-gp130-/- mice after SNI were significantly less responsive to cinnamaldehyde. Our study shows for the first time that TRPA1 upregulation is attributed specifically to uninjured neurons in the SNI model, and this depended on the IL-6 signal transducer gp130. We provide a solution to the enigma of TRPA1 regulation after nerve injury and stress its significance as an important target for neuropathic pain disorders.
Collapse
Affiliation(s)
- Theodora Kalpachidou
- Institute of Physiology, DPMP, Medical University of Innsbruck, Innsbruck, Austria
| | - Philipp Malsch
- Institute of Physiology, DPMP, Medical University of Innsbruck, Innsbruck, Austria
| | - Yanmei Qi
- Institute of Physiology, DPMP, Medical University of Innsbruck, Innsbruck, Austria
| | - Norbert Mair
- Institute of Physiology, DPMP, Medical University of Innsbruck, Innsbruck, Austria
| | - Stephan Geley
- Institute of Pathophysiology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Serena Quarta
- Institute of Physiology, DPMP, Medical University of Innsbruck, Innsbruck, Austria
| | - Kai K. Kummer
- Institute of Physiology, DPMP, Medical University of Innsbruck, Innsbruck, Austria
| | - Michaela Kress
- Institute of Physiology, DPMP, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
29
|
Zhang L, Bing S, Dong M, Lu X, Xiong Y. Targeting ion channels for the treatment of lung cancer. Biochim Biophys Acta Rev Cancer 2021; 1876:188629. [PMID: 34610420 DOI: 10.1016/j.bbcan.2021.188629] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 09/02/2021] [Accepted: 09/19/2021] [Indexed: 02/06/2023]
Abstract
Lung cancer is caused by several environmental and genetic variables and is globally associated with elevated morbidity and mortality. Among these variables, membrane-bound ion channels have a key role in regulating multiple signaling pathways in tumor cells and dysregulation of ion channel expression and function is closely related to proliferation, migration, and metastasis of lung cancer. This work reviews and summarizes current knowledge about the role of ion channels in lung cancer, focusing on the changes in the expression and function of various ion channels in lung cancer and how these changes affect lung cancer cell biology both in vitro and in vivo as evidenced by both genetic and pharmacological studies. It can help understand the molecular mechanisms of various ion channels influencing the initiation and progression of lung cancer and shed new insights into their roles in the development and treatment of this deadly disease.
Collapse
Affiliation(s)
- Liqin Zhang
- The First Affiliated Hospital of Wannan Medical College,Yijishan Hospital,2 Zheshan West Road, Wuhu 241000,China.
| | - Shuya Bing
- The First Affiliated Hospital of Wannan Medical College,Yijishan Hospital,2 Zheshan West Road, Wuhu 241000,China
| | - Mo Dong
- The First Affiliated Hospital of Wannan Medical College,Yijishan Hospital,2 Zheshan West Road, Wuhu 241000,China
| | - Xiaoqiu Lu
- The First Affiliated Hospital of Wannan Medical College,Yijishan Hospital,2 Zheshan West Road, Wuhu 241000,China
| | - Yuancheng Xiong
- The First Affiliated Hospital of Wannan Medical College,Yijishan Hospital,2 Zheshan West Road, Wuhu 241000,China
| |
Collapse
|
30
|
Jenkins RH, Hughes STO, Figueras AC, Jones SA. Unravelling the broader complexity of IL-6 involvement in health and disease. Cytokine 2021; 148:155684. [PMID: 34411990 DOI: 10.1016/j.cyto.2021.155684] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/20/2021] [Accepted: 08/04/2021] [Indexed: 02/07/2023]
Abstract
The classification of interleukin-6 (IL-6) as a pro-inflammatory cytokine undervalues the biological impact of this cytokine in health and disease. With broad activities affecting the immune system, tissue homeostasis and metabolic processes, IL-6 displays complex biology. The significance of these involvements has become increasingly important in clinical settings where IL-6 is identified as a prominent target for therapy. Here, clinical experience with IL-6 antagonists emphasises the need to understand the context-dependent properties of IL-6 within an inflammatory environment and the anticipated or unexpected consequences of IL-6 blockade. In this review, we will describe the immunobiology of IL-6 and explore the gamut of IL-6 bioactivity affecting the clinical response to biological drugs targeting this cytokine pathway.
Collapse
Affiliation(s)
- Robert H Jenkins
- Division of Infection & Immunity, The School of Medicine, Cardiff University, Cardiff, Wales, UK; Systems Immunity Research Institute, The School of Medicine, Cardiff University, Cardiff, Wales, UK
| | - Stuart T O Hughes
- Division of Infection & Immunity, The School of Medicine, Cardiff University, Cardiff, Wales, UK; Systems Immunity Research Institute, The School of Medicine, Cardiff University, Cardiff, Wales, UK
| | - Ana Cardus Figueras
- Division of Infection & Immunity, The School of Medicine, Cardiff University, Cardiff, Wales, UK; Systems Immunity Research Institute, The School of Medicine, Cardiff University, Cardiff, Wales, UK
| | - Simon A Jones
- Division of Infection & Immunity, The School of Medicine, Cardiff University, Cardiff, Wales, UK; Systems Immunity Research Institute, The School of Medicine, Cardiff University, Cardiff, Wales, UK.
| |
Collapse
|
31
|
Kummer KK, Zeidler M, Kalpachidou T, Kress M. Role of IL-6 in the regulation of neuronal development, survival and function. Cytokine 2021; 144:155582. [PMID: 34058569 DOI: 10.1016/j.cyto.2021.155582] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/07/2021] [Accepted: 05/11/2021] [Indexed: 12/17/2022]
Abstract
The pleiotropic cytokine interleukin-6 (IL-6) is emerging as a molecule with both beneficial and destructive potentials. It can exert opposing actions triggering either neuron survival after injury or causing neurodegeneration and cell death in neurodegenerative or neuropathic disorders. Importantly, neurons respond differently to IL-6 and this critically depends on their environment and whether they are located in the peripheral or the central nervous system. In addition to its hub regulator role in inflammation, IL-6 is recently emerging as an important regulator of neuron function in health and disease, offering exciting possibilities for more mechanistic insight into the pathogenesis of mental, neurodegenerative and pain disorders and for developing novel therapies for diseases with neuroimmune and neurogenic pathogenic components.
Collapse
Affiliation(s)
- Kai K Kummer
- Institute of Physiology, Medical University of Innsbruck, Austria
| | | | | | - Michaela Kress
- Institute of Physiology, Medical University of Innsbruck, Austria.
| |
Collapse
|
32
|
Nürnberger F, Leisengang S, Ott D, Murgott J, Gerstberger R, Rummel C, Roth J. Manifestation of lipopolysaccharide-induced tolerance in neuro-glial primary cultures of the rat afferent somatosensory system. Inflamm Res 2021; 70:429-444. [PMID: 33582876 PMCID: PMC8012319 DOI: 10.1007/s00011-021-01440-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 01/12/2021] [Accepted: 01/28/2021] [Indexed: 01/24/2023] Open
Abstract
OBJECTIVE Bacterial lipopolysaccharide (LPS) may contribute to the manifestation of inflammatory pain within structures of the afferent somatosensory system. LPS can induce a state of refractoriness to its own effects termed LPS tolerance. We employed primary neuro-glial cultures from rat dorsal root ganglia (DRG) and the superficial dorsal horn (SDH) of the spinal cord, mainly including the substantia gelatinosa to establish and characterize a model of LPS tolerance within these structures. METHODS Tolerance was induced by pre-treatment of both cultures with 1 µg/ml LPS for 18 h, followed by a short-term stimulation with a higher LPS dose (10 µg/ml for 2 h). Cultures treated with solvent were used as controls. Cells from DRG or SDH were investigated by means of RT-PCR (expression of inflammatory genes) and immunocytochemistry (translocation of inflammatory transcription factors into nuclei of cells from both cultures). Supernatants from both cultures were assayed for tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) by highly sensitive bioassays. RESULTS At the mRNA-level, pre-treatment with 1 µg/ml LPS caused reduced expression of TNF-α and enhanced IL-10/TNF-α expression ratios in both cultures upon subsequent stimulation with 10 µg/ml LPS, i.e. LPS tolerance. SDH cultures further showed reduced release of TNF-α into the supernatants and attenuated TNF-α immunoreactivity in microglial cells. In the state of LPS tolerance macrophages from DRG and microglial cells from SDH showed reduced LPS-induced nuclear translocation of the inflammatory transcription factors NFκB and NF-IL6. Nuclear immunoreactivity of the IL-6-activated transcription factor STAT3 was further reduced in neurons from DRG and astrocytes from SDH in LPS tolerant cultures. CONCLUSION A state of LPS tolerance can be induced in primary cultures from the afferent somatosensory system, which is characterized by a down-regulation of pro-inflammatory mediators. Thus, this model can be applied to study the effects of LPS tolerance at the cellular level, for example possible modifications of neuronal reactivity patterns upon inflammatory stimulation.
Collapse
Affiliation(s)
- Franz Nürnberger
- Department of Veterinary Physiology and Biochemistry, Justus-Liebig-University Giessen, Frankfurter Strasse 100, 35392, Giessen, Germany
| | - Stephan Leisengang
- Department of Veterinary Physiology and Biochemistry, Justus-Liebig-University Giessen, Frankfurter Strasse 100, 35392, Giessen, Germany
| | - Daniela Ott
- Department of Veterinary Physiology and Biochemistry, Justus-Liebig-University Giessen, Frankfurter Strasse 100, 35392, Giessen, Germany
| | - Jolanta Murgott
- Department of Veterinary Physiology and Biochemistry, Justus-Liebig-University Giessen, Frankfurter Strasse 100, 35392, Giessen, Germany
| | - Rüdiger Gerstberger
- Department of Veterinary Physiology and Biochemistry, Justus-Liebig-University Giessen, Frankfurter Strasse 100, 35392, Giessen, Germany
| | - Christoph Rummel
- Department of Veterinary Physiology and Biochemistry, Justus-Liebig-University Giessen, Frankfurter Strasse 100, 35392, Giessen, Germany
| | - Joachim Roth
- Department of Veterinary Physiology and Biochemistry, Justus-Liebig-University Giessen, Frankfurter Strasse 100, 35392, Giessen, Germany.
| |
Collapse
|
33
|
Sensory neuron-associated macrophages as novel modulators of neuropathic pain. Pain Rep 2021; 6:e873. [PMID: 33981924 PMCID: PMC8108583 DOI: 10.1097/pr9.0000000000000873] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/13/2020] [Accepted: 10/19/2020] [Indexed: 12/28/2022] Open
Abstract
The peripheral nervous system comprises an infinity of neural networks that act in the communication between the central nervous system and the most diverse tissues of the body. Along with the extension of the primary sensory neurons (axons and cell bodies), a population of resident macrophages has been described. These newly called sensory neuron-associated macrophages (sNAMs) seem to play an essential role in physiological and pathophysiological processes, including infection, autoimmunity, nerve degeneration/regeneration, and chronic neuropathic pain. After different types of peripheral nerve injury, there is an increase in the number and activation of sNAMs in the sciatic nerve and sensory ganglia. The activation of sNAMs and their participation in neuropathic pain development depends on the stimulation of pattern recognition receptors such as Toll-like receptors and Nod-like receptors, chemokines/cytokines, and microRNAs. On activation, sNAMs trigger the production of critical inflammatory mediators such as proinflammatory cytokines (eg, TNF and IL-1β) and reactive oxygen species that can act in the amplification of primary sensory neurons sensitization. On the other hand, there is evidence that sNAMs can produce antinociceptive mediators (eg, IL-10) that counteract neuropathic pain development. This review will present the cellular and molecular mechanisms behind the participation of sNAMs in peripheral nerve injury-induced neuropathic pain development. Understanding how sNAMs are activated and responding to nerve injury can help set novel targets for the control of neuropathic pain.
Collapse
|
34
|
Sebba A. Pain: A Review of Interleukin-6 and Its Roles in the Pain of Rheumatoid Arthritis. Open Access Rheumatol 2021; 13:31-43. [PMID: 33707975 PMCID: PMC7943546 DOI: 10.2147/oarrr.s291388] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 01/22/2021] [Indexed: 12/29/2022] Open
Abstract
Pain is a major and common symptom reported as a top priority in patients with rheumatoid arthritis (RA). Intuitively, RA-related pain is often considered to be a natural consequence of peripheral inflammation, so treatment of RA is expected to manage pain concurrently as part of inflammation control. However, pain in patients with RA can be poorly correlated with objective measures of inflammation, for example, in patients who are otherwise in remission. Joint damage appears to account for only a fraction of this residual pain. Emerging evidence suggests that alteration of peripheral and central pain processing contributes to RA-related pain; this is parallel to, but somewhat independent of, joint inflammation. Interleukin (IL)-6 is a proinflammatory cytokine that contributes to the pathogenesis of RA. It exerts systemic effects via signaling through soluble forms of the IL-6 receptor (“trans-signaling”). Evidence from preclinical studies demonstrates that intra-articular IL-6 can produce long-lasting peripheral sensitization to mechanical stimulation and suggests an important role for IL-6 in central pain sensitization. This may be partly explained by its ability to activate neurons through trans-signaling, affecting nociceptive plasticity and nerve fiber regrowth. Local activity at neuron endings may culminate in altered pain processing in the central nervous system because of persistent signaling from sensitized peripheral neurons. Peripheral and central sensitization can promote the development of chronic pain, which can have a significant impact on patients’ health and quality of life. A proportion of pain in RA may be more appropriately managed as an entity separate from inflammation. Both the peripheral and central nervous systems should be recognized as important potential systems targeted by RA. The substantial burden of RA-related chronic pain suggests that pain should be a key focus in RA management and should be assessed and addressed early and separately from the inflammatory component.
Collapse
Affiliation(s)
- Anthony Sebba
- Division of Rheumatology, University of South Florida, Tampa, FL, USA
| |
Collapse
|
35
|
Bakare AO, Owoyele BV. Bromelain reduced pro-inflammatory mediators as a common pathway that mediate antinociceptive and anti-anxiety effects in sciatic nerve ligated Wistar rats. Sci Rep 2021; 11:289. [PMID: 33432004 PMCID: PMC7801445 DOI: 10.1038/s41598-020-79421-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 12/08/2020] [Indexed: 02/02/2023] Open
Abstract
The involvement of pro-inflammatory mediators complicates the complex mechanism in neuropathic pain (NP). This study investigated the roles of bromelain against pro-inflammatory mediators as a mechanism that underpins its antinociceptive and anti-anxiety effects in the peripheral model of NP. Sixty-four male Wistar rats randomly divided into eight groups, were used for the study. A chronic constriction injury model of peripheral neuropathy was used to induce NP. Tail-immersion and von Frey filaments tests were used to assess hyperalgesia while open field and elevated plus mazes were used to assess anxiety-like behaviour. NF-кB, iNOS, nitrate, and pro-inflammatory cytokines were investigated in the plasma, sciatic nerve, and brain tissues using ELISA, spectrophotometer, and immunohistochemistry techniques after twenty-one days of treatment. Bromelain significantly (p < 0.05) improved the cardinal signs of NP and inhibited anxiety-like behaviours in ligated Wistar rats. It mitigated the increases in cerebral cortex interleukin (IL) -1β, IL-6, and PGE2 levels. Bromelain reduced NF-кB, IL-1β, IL-6, TNF-α, PGE2, and nitrate concentrations as well as the expression of iNOS in the sciatic nerve. Hence, the antinociceptive and anxiolytic effects of bromelain in the sciatic nerve ligation model of NP is in part due to its ability to reduce nitrosative and inflammatory activities.
Collapse
Affiliation(s)
- Ahmed O Bakare
- Department of Physiology, Neuroscience and Inflammation Unit, Faculty of Basic Medical Sciences, University of Ilorin, Ilorin, Kwara State, Nigeria
| | - Bamidele V Owoyele
- Department of Physiology, Neuroscience and Inflammation Unit, Faculty of Basic Medical Sciences, University of Ilorin, Ilorin, Kwara State, Nigeria.
| |
Collapse
|
36
|
Simon LS, Taylor PC, Choy EH, Sebba A, Quebe A, Knopp KL, Porreca F. The Jak/STAT pathway: A focus on pain in rheumatoid arthritis. Semin Arthritis Rheum 2020; 51:278-284. [PMID: 33412435 DOI: 10.1016/j.semarthrit.2020.10.008] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 09/02/2020] [Accepted: 10/23/2020] [Indexed: 12/16/2022]
Abstract
Pain is a manifestation of rheumatoid arthritis (RA) that is mediated by inflammatory and non-inflammatory mechanisms and negatively affects quality of life. Recent findings from a Phase 3 clinical trial showed that patients with RA who were treated with a Janus kinase 1 (Jak1) and Janus kinase 2 (Jak2) inhibitor achieved significantly greater improvements in pain than those treated with a tumor necrosis factor blocker; both treatments resulted in similar changes in standard clinical measures and markers of inflammation. These findings suggest that Jak1 and Jak2 inhibition may relieve pain in RA caused by inflammatory and non-inflammatory mechanisms and are consistent with the overarching involvement of the Jak-signal transducer and activator of transcription (Jak/STAT) pathway in mediating the action, expression, and regulation of a multitude of pro- and anti-inflammatory cytokines. In this review, we provide an overview of pain in RA, the underlying importance of cytokines regulated directly or indirectly by the Jak/STAT pathway, and therapeutic targeting of the Jak/STAT pathway in RA. As highlighted herein, multiple cytokines directly or indirectly regulated by the Jak/STAT pathway play important roles in mediating various mechanisms underlying pain in RA. Having a better understanding of these mechanisms may help clinicians make treatment decisions that optimize the control of inflammation and pain.
Collapse
Affiliation(s)
| | - Peter C Taylor
- Botnar Research Centre, University of Oxford, Oxford, UK
| | - Ernest H Choy
- CREATE Centre, Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| | | | | | | | - Frank Porreca
- Department of Pharmacology, College of Medicine, University of Arizona, 1501 N. Campbell Avenue, Tucson, AZ 85718, USA.
| |
Collapse
|
37
|
Leisengang S, Nürnberger F, Ott D, Murgott J, Gerstberger R, Rummel C, Roth J. Primary culture of the rat spinal dorsal horn: a tool to investigate the effects of inflammatory stimulation on the afferent somatosensory system. Pflugers Arch 2020; 472:1769-1782. [PMID: 33098464 PMCID: PMC7691309 DOI: 10.1007/s00424-020-02478-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 08/28/2020] [Accepted: 10/14/2020] [Indexed: 02/06/2023]
Abstract
One maladaptive consequence of inflammatory stimulation of the afferent somatosensory system is the manifestation of inflammatory pain. We established and characterized a neuroglial primary culture of the rat superficial dorsal horn (SDH) of the spinal cord to test responses of this structure to neurochemical, somatosensory, or inflammatory stimulation. Primary cultures of the rat SDH consist of neurons (43%), oligodendrocytes (35%), astrocytes (13%), and microglial cells (9%). Neurons of the SDH responded to cooling (7%), heating (18%), glutamate (80%), substance P (43%), prostaglandin E2 (8%), and KCl (100%) with transient increases in the intracellular calcium [Ca2+]i. Short-term stimulation of SDH primary cultures with LPS (10 μg/ml, 2 h) caused increased expression of pro-inflammatory cytokines, inflammatory transcription factors, and inducible enzymes responsible for inflammatory prostaglandin E2 synthesis. At the protein level, increased concentrations of tumor necrosis factor-α (TNFα) and interleukin-6 (IL-6) were measured in the supernatants of LPS-stimulated SDH cultures and enhanced TNFα and IL-6 immunoreactivity was observed specifically in microglial cells. LPS-exposed microglial cells further showed increased nuclear immunoreactivity for the inflammatory transcription factors NFκB, NF-IL6, and pCREB, indicative of their activation. The short-term exposure to LPS further caused a reduction in the strength of substance P as opposed to glutamate-evoked Ca2+-signals in SDH neurons. However, long-term stimulation with a low dose of LPS (0.01 μg/ml, 24 h) resulted in a significant enhancement of glutamate-induced Ca2+ transients in SDH neurons, while substance P-evoked Ca2+ signals were not influenced. Our data suggest a critical role for microglial cells in the initiation of inflammatory processes within the SDH of the spinal cord, which are accompanied by a modulation of neuronal responses.
Collapse
Affiliation(s)
- Stephan Leisengang
- Department of Veterinary Physiology and Biochemistry, Justus-Liebig-University Giessen, Frankfurter Strasse 100, 35392, Giessen, Germany.,Center for Mind, Brain and Behavior - CMBB, Philipps-University Marburg, Marburg, Germany.,Center for Mind, Brain and Behavior - CMBB, Justus-Liebig-University of Giessen, Giessen, Germany
| | - Franz Nürnberger
- Department of Veterinary Physiology and Biochemistry, Justus-Liebig-University Giessen, Frankfurter Strasse 100, 35392, Giessen, Germany
| | - Daniela Ott
- Department of Veterinary Physiology and Biochemistry, Justus-Liebig-University Giessen, Frankfurter Strasse 100, 35392, Giessen, Germany
| | - Jolanta Murgott
- Department of Veterinary Physiology and Biochemistry, Justus-Liebig-University Giessen, Frankfurter Strasse 100, 35392, Giessen, Germany
| | - Rüdiger Gerstberger
- Department of Veterinary Physiology and Biochemistry, Justus-Liebig-University Giessen, Frankfurter Strasse 100, 35392, Giessen, Germany
| | - Christoph Rummel
- Department of Veterinary Physiology and Biochemistry, Justus-Liebig-University Giessen, Frankfurter Strasse 100, 35392, Giessen, Germany.,Center for Mind, Brain and Behavior - CMBB, Philipps-University Marburg, Marburg, Germany.,Center for Mind, Brain and Behavior - CMBB, Justus-Liebig-University of Giessen, Giessen, Germany
| | - Joachim Roth
- Department of Veterinary Physiology and Biochemistry, Justus-Liebig-University Giessen, Frankfurter Strasse 100, 35392, Giessen, Germany. .,Center for Mind, Brain and Behavior - CMBB, Philipps-University Marburg, Marburg, Germany. .,Center for Mind, Brain and Behavior - CMBB, Justus-Liebig-University of Giessen, Giessen, Germany.
| |
Collapse
|
38
|
Ray PR, Wangzhou A, Ghneim N, Yousuf MS, Paige C, Tavares-Ferreira D, Mwirigi JM, Shiers S, Sankaranarayanan I, McFarland AJ, Neerukonda SV, Davidson S, Dussor G, Burton MD, Price TJ. A pharmacological interactome between COVID-19 patient samples and human sensory neurons reveals potential drivers of neurogenic pulmonary dysfunction. Brain Behav Immun 2020; 89:559-568. [PMID: 32497778 PMCID: PMC7263237 DOI: 10.1016/j.bbi.2020.05.078] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/28/2020] [Accepted: 05/29/2020] [Indexed: 12/21/2022] Open
Abstract
The SARS-CoV-2 virus infects cells of the airway and lungs in humans causing the disease COVID-19. This disease is characterized by cough, shortness of breath, and in severe cases causes pneumonia and acute respiratory distress syndrome (ARDS) which can be fatal. Bronchial alveolar lavage fluid (BALF) and plasma from mild and severe cases of COVID-19 have been profiled using protein measurements and bulk and single cell RNA sequencing. Onset of pneumonia and ARDS can be rapid in COVID-19, suggesting a potential neuronal involvement in pathology and mortality. We hypothesized that SARS-CoV-2 infection drives changes in immune cell-derived factors that then interact with receptors expressed by the sensory neuronal innervation of the lung to further promote important aspects of disease severity, including ARDS. We sought to quantify how immune cells might interact with sensory innervation of the lung in COVID-19 using published data from patients, existing RNA sequencing datasets from human dorsal root ganglion neurons and other sources, and a genome-wide ligand-receptor pair database curated for pharmacological interactions relevant for neuro-immune interactions. Our findings reveal a landscape of ligand-receptor interactions in the lung caused by SARS-CoV-2 viral infection and point to potential interventions to reduce the burden of neurogenic inflammation in COVID-19 pulmonary disease. In particular, our work highlights opportunities for clinical trials with existing or under development rheumatoid arthritis and other (e.g. CCL2, CCR5 or EGFR inhibitors) drugs to treat high risk or severe COVID-19 cases.
Collapse
Affiliation(s)
- Pradipta R Ray
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA.
| | - Andi Wangzhou
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA
| | - Nizar Ghneim
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA
| | - Muhammad S Yousuf
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA
| | - Candler Paige
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA
| | - Diana Tavares-Ferreira
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA
| | - Juliet M Mwirigi
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA
| | - Stephanie Shiers
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA
| | - Ishwarya Sankaranarayanan
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA
| | - Amelia J McFarland
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA
| | - Sanjay V Neerukonda
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA
| | - Steve Davidson
- University of Cincinnati, College of Medicine, Department of Anesthesiology, USA
| | - Gregory Dussor
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA
| | - Michael D Burton
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Neuroimmunology and Behavior Research Group, USA
| | - Theodore J Price
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA.
| |
Collapse
|
39
|
Garza Carbajal A, Ebersberger A, Thiel A, Ferrari L, Acuna J, Brosig S, Isensee J, Moeller K, Siobal M, Rose-John S, Levine J, Schaible HG, Hucho T. Oncostatin M induces hyperalgesic priming and amplifies signaling of cAMP to ERK by RapGEF2 and PKA. J Neurochem 2020; 157:1821-1837. [PMID: 32885411 DOI: 10.1111/jnc.15172] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 08/19/2020] [Accepted: 08/20/2020] [Indexed: 12/19/2022]
Abstract
Hyperalgesic priming is characterized by enhanced nociceptor sensitization by pronociceptive mediators, prototypically PGE2 . Priming has gained interest as a mechanism underlying the transition to chronic pain. Which stimuli induce priming and what cellular mechanisms are employed remains incompletely understood. In adult male rats, we present the cytokine Oncostatin M (OSM), a member of the IL-6 family, as an inducer of priming by a novel mechanism. We used a high content microscopy based approach to quantify the activation of endogenous PKA-II and ERK of thousands sensory neurons in culture. Incubation with OSM increased and prolonged ERK activation by agents that increase cAMP production such as PGE2 , forskolin, and cAMP analogs. These changes were specific to IB4/CaMKIIα positive neurons, required protein translation, and increased cAMP-to-ERK signaling. In both, control and OSM-treated neurons, cAMP/ERK signaling involved RapGEF2 and PKA but not Epac. Similar enhancement of cAMP-to-ERK signaling could be induced by GDNF, which acts mostly on IB4/CaMKIIα-positive neurons, but not by NGF, which acts mostly on IB4/CaMKIIα-negative neurons. In vitro, OSM pretreatment rendered baseline TTX-R currents ERK-dependent and switched forskolin-increased currents from partial to full ERK-dependence in small/medium sized neurons. In summary, priming induced by OSM uses a novel mechanism to enhance and prolong coupling of cAMP/PKA to ERK1/2 signaling without changing the overall pathway structure.
Collapse
Affiliation(s)
- Anibal Garza Carbajal
- Department of Anaesthesiology and Intensive Care Medicine, Translational Pain Research, University Hospital Cologne, University Cologne, Cologne, Germany
| | | | - Alina Thiel
- Department of Anaesthesiology and Intensive Care Medicine, Translational Pain Research, University Hospital Cologne, University Cologne, Cologne, Germany
| | - Luiz Ferrari
- Department of Oral and Maxillofacial Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Jeremy Acuna
- Department of Anaesthesiology and Intensive Care Medicine, Translational Pain Research, University Hospital Cologne, University Cologne, Cologne, Germany
| | - Stephanie Brosig
- Department of Anaesthesiology and Intensive Care Medicine, Translational Pain Research, University Hospital Cologne, University Cologne, Cologne, Germany
| | - Joerg Isensee
- Department of Anaesthesiology and Intensive Care Medicine, Translational Pain Research, University Hospital Cologne, University Cologne, Cologne, Germany
| | - Katharina Moeller
- Department of Anaesthesiology and Intensive Care Medicine, Translational Pain Research, University Hospital Cologne, University Cologne, Cologne, Germany
| | - Maike Siobal
- Department of Anaesthesiology and Intensive Care Medicine, Translational Pain Research, University Hospital Cologne, University Cologne, Cologne, Germany
| | | | - Jon Levine
- Department of Oral and Maxillofacial Surgery, University of California San Francisco, San Francisco, CA, USA
| | | | - Tim Hucho
- Department of Anaesthesiology and Intensive Care Medicine, Translational Pain Research, University Hospital Cologne, University Cologne, Cologne, Germany
| |
Collapse
|
40
|
Abstract
A limited number of peripheral targets generate pain. Inflammatory mediators can sensitize these. The review addresses targets acting exclusively or predominantly on sensory neurons, mediators involved in inflammation targeting sensory neurons, and mediators involved in a more general inflammatory process, of which an analgesic effect secondary to an anti-inflammatory effect can be expected. Different approaches to address these systems are discussed, including scavenging proinflammatory mediators, applying anti-inflammatory mediators, and inhibiting proinflammatory or facilitating anti-inflammatory receptors. New approaches are contrasted to established ones; the current stage of progress is mentioned, in particular considering whether there is data from a molecular and cellular level, from animals, or from human trials, including an early stage after a market release. An overview of publication activity is presented, considering a IuPhar/BPS-curated list of targets with restriction to pain-related publications, which was also used to identify topics.
Collapse
Affiliation(s)
- Cosmin I Ciotu
- Center of Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstrasse 17, 1090, Vienna, Austria
| | - Michael J M Fischer
- Center of Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstrasse 17, 1090, Vienna, Austria.
| |
Collapse
|
41
|
Yaekura A, Yoshida K, Morii K, Oketani Y, Okumura I, Kaneshiro K, Shibanuma N, Sakai Y, Hashiramoto A. Chronotherapy targeting cytokine secretion attenuates collagen-induced arthritis in mice. Int Immunopharmacol 2020; 84:106549. [PMID: 32416449 DOI: 10.1016/j.intimp.2020.106549] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 04/25/2020] [Accepted: 04/27/2020] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Diurnal variation of symptoms are observed in rheumatoid arthritis, especially in productions of cytokines that show peak concentrations during mid night. In contrast, cytokines of collagen-induced arthritis (CIA) mice increase in daytimes under Mid-light condition. By using chronotherapy, differences in drug efficacies according to administration time of Baricitinib, a wide ranged cytokine blocker, were examined in CIA mice. METHODS CIA mice were administered a dose of 3 mg/kg of Baricitinib once a day at zeitgeber time (ZT) 0 or ZT12 for 21 days. Arthritis scores, histopathology and factors related to joint destruction in sera were examined. Phosphorylation of STAT3 in liver, expressions of cytokines in spleen, and Interleukin (IL)-6 and tumor necrosis factor (TNF)-α in sera were measured. RESULTS In CIA mice, diurnal variations were observed both in expressions of cytokines and phosphorylation of STAT3. Arthritis scores of ZT0/12 group decreased from day3 as compared to untreated mice, and those of ZT0 group significantly decreased as compared to ZT12 group from day12. Pathological findings, immunohistochemistry of cytokines and Receptor activator of nuclear factor kappa-Β ligand (RANKL)/osteoprotegerin ratio in sera well reflected results of arthritis scores. Diurnal variation of STAT3 phosphorylation was suppressed in ZT0 group. At ZT2, expressions of IL-6/Interferon-γ/TNF/granulocyte-macrophage colony-stimulating factor in ZT0 group were significantly decreased as compared to untreated mice, though not in ZT12 group. In ZT0 group, IL-6 and TNF-α in sera were decreased for longer time than that in ZT12 group. CONCLUSION Chronotherapy using Baricitinib targeting cytokine secretions is effective in CIA mice. Clinical applications of chronotherapy can be expected to enhance the drug efficacy.
Collapse
Affiliation(s)
- Arisa Yaekura
- Department of Biophysics, Division of Clinical of Immunology, Kobe University Graduate School of Health Sciences, Tomogaoka 7-10-2, Suma, Kobe 654-0142, Japan
| | - Kohsuke Yoshida
- Department of Biophysics, Division of Clinical of Immunology, Kobe University Graduate School of Health Sciences, Tomogaoka 7-10-2, Suma, Kobe 654-0142, Japan
| | - Kanta Morii
- Department of Biophysics, Division of Clinical of Immunology, Kobe University Graduate School of Health Sciences, Tomogaoka 7-10-2, Suma, Kobe 654-0142, Japan
| | - Yuto Oketani
- Department of Biophysics, Division of Clinical of Immunology, Kobe University Graduate School of Health Sciences, Tomogaoka 7-10-2, Suma, Kobe 654-0142, Japan
| | - Ikumi Okumura
- Department of Biophysics, Division of Clinical of Immunology, Kobe University Graduate School of Health Sciences, Tomogaoka 7-10-2, Suma, Kobe 654-0142, Japan
| | - Kenta Kaneshiro
- Department of Biophysics, Division of Clinical of Immunology, Kobe University Graduate School of Health Sciences, Tomogaoka 7-10-2, Suma, Kobe 654-0142, Japan
| | - Nao Shibanuma
- Department of Orthopaedic Surgery, Kobe Kaisei Hospital, Shinohara-Kita 3-11-15, Nada, Kobe 657-0068, Japan
| | - Yoshitada Sakai
- Department of Rehabilitation Science, Kobe University Graduate School of Medicine, Kusunoki 7-5-1, Cyuoh, Kobe 650-0017, Japan
| | - Akira Hashiramoto
- Department of Biophysics, Division of Clinical of Immunology, Kobe University Graduate School of Health Sciences, Tomogaoka 7-10-2, Suma, Kobe 654-0142, Japan.
| |
Collapse
|
42
|
Ray PR, Wangzhou A, Ghneim N, Yousuf MS, Paige C, Tavares-Ferreira D, Mwirigi JM, Shiers S, Sankaranarayanan I, McFarland AJ, Neerukonda SV, Davidson S, Dussor G, Burton MD, Price TJ. A pharmacological interactome between COVID-19 patient samples and human sensory neurons reveals potential drivers of neurogenic pulmonary dysfunction. SSRN 2020:3581446. [PMID: 32714114 PMCID: PMC7366818 DOI: 10.2139/ssrn.3581446] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/04/2020] [Indexed: 11/15/2022]
Abstract
The SARS-CoV-2 virus infects cells of the airway and lungs in humans causing the disease COVID-19. This disease is characterized by cough, shortness of breath, and in severe cases causes pneumonia and acute respiratory distress syndrome (ARDS) which can be fatal. Bronchial alveolar lavage fluid (BALF) and plasma from mild and severe cases of COVID-19 have been profiled using protein measurements and bulk and single cell RNA sequencing. Onset of pneumonia and ARDS can be rapid in COVID-19, suggesting a potential neuronal involvement in pathology and mortality. We sought to quantify how immune cells might interact with sensory innervation of the lung in COVID-19 using published data from patients, existing RNA sequencing datasets from human dorsal root ganglion neurons and other sources, and a genome-wide ligand-receptor pair database curated for pharmacological interactions relevant for neuro-immune interactions. Our findings reveal a landscape of ligand-receptor interactions in the lung caused by SARS-CoV-2 viral infection and point to potential interventions to reduce the burden of neurogenic inflammation in COVID-19 disease. In particular, our work highlights opportunities for clinical trials with existing or under development rheumatoid arthritis and other (e.g. CCL2, CCR5 or EGFR inhibitors) drugs to treat high risk or severe COVID-19 cases.
Collapse
Affiliation(s)
- Pradipta R. Ray
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Andi Wangzhou
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Nizar Ghneim
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Muhammad S. Yousuf
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Candler Paige
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Diana Tavares-Ferreira
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Juliet M. Mwirigi
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Stephanie Shiers
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Ishwarya Sankaranarayanan
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Amelia J. McFarland
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Sanjay V. Neerukonda
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Steve Davidson
- University of Cincinnati, College of Medicine, Department of Anesthesiology
| | - Gregory Dussor
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Michael D. Burton
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Theodore J. Price
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| |
Collapse
|
43
|
Leisengang S, Ott D, Murgott J, Nürnberger F, Gerstberger R, Rummel C, Schmidt M, Roth J. Effects of gabapentinoids on responses of primary cultures from rat dorsal root ganglia to inflammatory or somatosensory stimuli. J Basic Clin Physiol Pharmacol 2020; 31:/j/jbcpp.ahead-of-print/jbcpp-2019-0261/jbcpp-2019-0261.xml. [PMID: 32078575 DOI: 10.1515/jbcpp-2019-0261] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 11/26/2019] [Indexed: 11/15/2022]
Abstract
Background Gabapentinoids are known to reduce neuropathic pain. The aim of this experimental study was to investigate whether gabapentinoids exert anti-inflammatory and/or anti-nociceptive effects at the cellular level using primary cultures of rat dorsal root ganglia (DRG). Methods Cells from rat DRG were cultured in the presence of gabapentin or pregabalin, and we tested the effects of subsequent stimulation with lipopolysaccharide (LPS) on the expression of genes (real-time polymerase chain reaction) and production of tumor necrosis factor-α (TNFα) and interleukin-6 (IL-6) by specific bioassays. Using Ca2+ imaging, we further investigated in neurons the effects of gabapentinoids upon stimulation with the TRPV-1 agonist capsaicin. Results There is a small influence of gabapentinoids on the inflammatory response to LPS stimulation, namely, a significantly reduced expression of IL-6. Pregabalin and gabapentin further seem to exert a moderate inhibitory influence on capsaicin-induced Ca2+ signals in DRG neurons. Conclusions Although the single inhibitory effects of gabapentinoids on inflammatory and nociceptive responses are moderate, a combination of both effects might provide an explanation for the proposed function of these substances as an adjuvant for the reduction of neuropathic pain.
Collapse
Affiliation(s)
- Stephan Leisengang
- Department of Veterinary-Physiology and -Biochemistry, Justus-Liebig University, Frankfurter Strasse 100, D-35392 Giessen, Germany
| | - Daniela Ott
- Department of Veterinary-Physiology and -Biochemistry, Justus-Liebig University, Frankfurter Strasse 100, D-35392 Giessen, Germany
| | - Jolanta Murgott
- Department of Veterinary-Physiology and -Biochemistry, Justus-Liebig University, Frankfurter Strasse 100, D-35392 Giessen, Germany
| | - Franz Nürnberger
- Department of Veterinary-Physiology and -Biochemistry, Justus-Liebig University, Frankfurter Strasse 100, D-35392 Giessen, Germany
| | - Rüdiger Gerstberger
- Department of Veterinary-Physiology and -Biochemistry, Justus-Liebig University, Frankfurter Strasse 100, D-35392 Giessen, Germany
| | - Christoph Rummel
- Department of Veterinary-Physiology and -Biochemistry, Justus-Liebig University, Frankfurter Strasse 100, D-35392 Giessen, Germany
| | - Martin Schmidt
- Department of Veterinary Clinical Sciences, Small Animal Clinic, Justus-Liebig University, Frankfurter Strasse 108, D-35392 Giessen, Germany
| | - Joachim Roth
- Department of Veterinary-Physiology and -Biochemistry, Justus-Liebig University, Frankfurter Strasse 100, D-35392 Giessen, Germany, Phone: +49-641-99-38150, Fax: +49-641-99-38159
| |
Collapse
|
44
|
Blockade of IL-6 signaling prevents paclitaxel-induced neuropathy in C57Bl/6 mice. Cell Death Dis 2020; 11:45. [PMID: 31969555 PMCID: PMC6976596 DOI: 10.1038/s41419-020-2239-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 01/08/2020] [Accepted: 01/08/2020] [Indexed: 12/13/2022]
Abstract
The microtubule-stabilizing agent paclitaxel frequently leads to chemotherapy-induced peripheral neuropathy (CIN), which further increases the burden of disease and often necessitates treatment limitations. The pathophysiology of CIN appears to involve both “upstream” effects including altered intracellular calcium signaling and activation of calcium dependent proteases such as calpain as well as subsequent “downstream” neuro-inflammatory reactions with cytokine release and macrophage infiltration of dorsal root ganglia. In this study, we aimed to investigate whether these processes are linked by the pro-inflammatory cytokine interleukin-6 (IL-6). We observed that paclitaxel exposure induced IL-6 synthesis in cultured sensory neurons from postnatal Wistar rats, which could be prevented by co-treatment with a calpain inhibitor. This suggests a calcium dependent process. We demonstrate that adult C57BL/6 mice deficient in IL-6 are protected from developing functional and histological changes of paclitaxel-induced neuropathy. Furthermore, pretreatment with an IL-6-neutralizing antibody resulted in the prevention of paclitaxel-induced neuropathy in C57BL/6 mice. Electrophysiological data from our preclinical model was adequately reflected by measurements of patients undergoing paclitaxel therapy for ovarian cancer. In this cohort, measured Il-6 levels correlated with the severity of neuropathy. Our findings demonstrate that IL-6 plays a pivotal role in the pathophysiology of paclitaxel-induced neuropathy per se and that pharmacological or genetic interference with this signaling pathway prevents the development of this potentially debilitating adverse effect. These findings provide a rationale for a clinical trial with IL-6 neutralizing antibodies to prevent dose-limiting neurotoxic adverse effects of paclitaxel chemotherapy.
Collapse
|
45
|
Larsson OJ, Georén SK, Cardell LO. Rapid activation of brainstem nuclei following TLR stimulation of the nasal mucosa. Acta Neurobiol Exp (Wars) 2020. [DOI: 10.21307/ane-2020-031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
46
|
Stover JD, Farhang N, Lawrence B, Bowles RD. Multiplex Epigenome Editing of Dorsal Root Ganglion Neuron Receptors Abolishes Redundant Interleukin 6, Tumor Necrosis Factor Alpha, and Interleukin 1β Signaling by the Degenerative Intervertebral Disc. Hum Gene Ther 2019; 30:1147-1160. [PMID: 31056946 PMCID: PMC6761584 DOI: 10.1089/hum.2019.032] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 04/27/2019] [Indexed: 01/07/2023] Open
Abstract
Back pain is the leading cause of disability worldwide and contributes to significant socioeconomic impacts. It has been hypothesized that the degenerative intervertebral disc (IVD) contributes to back pain by sensitizing nociceptive neurons innervating the IVD to stimuli that would not be painful to healthy patients. However, the inflammatory signaling networks mediating this sensitization remain poorly understood. A better understanding of the underlying mechanisms of degenerative IVD-induced changes in nociception is required to improve the understanding and treatment of back pain. Toward these ends, a novel in vitro model was developed to investigate degenerative IVD-induced changes in dorsal root ganglion (DRG) neuron activation by measuring DRG neuron activity following neuron seeding on human degenerative IVD tissue collected from patients undergoing surgical treatment for back pain. Lentiviral clustered regularly interspaced palindromic repeat (CRISPR) epigenome editing vectors were built to downregulate the inflammatory receptors TNFR1, IL1R1, and IL6st in DRG neurons in single- and multiplex. Multiplex CRISPR epigenome editing of inflammatory receptors demonstrated that degenerative IVD tissue drives thermal sensitization through the simultaneous and redundant signaling of interleukin (IL)-6, tumor necrosis factor alpha (TNF-α), and IL-1β. This work elucidates redundant signaling pathways in neuron interactions with the degenerative IVD and suggests the need for multiplex targeting of IL-6, TNF-α, and IL-1β for pain modulation in the degenerative IVD.
Collapse
Affiliation(s)
- Joshua D. Stover
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah
| | - Niloofar Farhang
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah
| | - Brandon Lawrence
- Department of Orthopaedics, University of Utah, Salt Lake City, Utah
| | - Robby D. Bowles
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah
- Department of Orthopaedics, University of Utah, Salt Lake City, Utah
| |
Collapse
|
47
|
Alvarez P, Bogen O, Levine JD. Interleukin 6 decreases nociceptor expression of the potassium channel KV1.4 in a rat model of hand-arm vibration syndrome. Pain 2019; 160:1876-1882. [PMID: 31335655 PMCID: PMC6668361 DOI: 10.1097/j.pain.0000000000001570] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Chronic muscle pain is a prominent symptom of the hand-arm vibration syndrome (HAVS), an occupational disease induced by exposure to vibrating power tools, but the underlying mechanism remains unknown. We evaluated the hypothesis that vibration induces an interleukin 6 (IL-6)-mediated downregulation of the potassium voltage-gated channel subfamily A member 4 (KV1.4) in nociceptors leading to muscle pain. Adult male rats were submitted to a protocol of mechanical vibration of the right hind limb. Twenty-four hours after vibration, muscle hyperalgesia was observed, concomitant to increased levels of IL-6 in the gastrocnemius muscle and decreased expression of KV1.4 in the dorsal root ganglia. Local injection of neutralizing antibodies against IL-6 attenuated the muscle hyperalgesia induced by vibration, whereas antisense knockdown of this channel in the dorsal root ganglia mimicked the muscle hyperalgesia observed in the model of HAVS. Finally, knockdown of the IL-6 receptor signaling subunit glycoprotein 130 (gp130) attenuated both vibration-induced muscle hyperalgesia and downregulation of KV1.4. These results support the hypothesis that IL-6 plays a central role in the induction of muscle pain in HAVS. This likely occurs through intracellular signaling downstream to the IL-6 receptor subunit gp130, which decreases the expression of KV1.4 in nociceptors.
Collapse
Affiliation(s)
- Pedro Alvarez
- Department of Oral and Maxillofacial Surgery, University of California, San Francisco, USA
| | - Oliver Bogen
- Department of Oral and Maxillofacial Surgery, University of California, San Francisco, USA
| | - Jon D. Levine
- Department of Oral and Maxillofacial Surgery, University of California, San Francisco, USA
- Department of Medicine, University of California, San Francisco, USA
| |
Collapse
|
48
|
Choy EHS, Calabrese LH. Neuroendocrine and neurophysiological effects of interleukin 6 in rheumatoid arthritis. Rheumatology (Oxford) 2019; 57:1885-1895. [PMID: 29186541 PMCID: PMC6199533 DOI: 10.1093/rheumatology/kex391] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Indexed: 11/14/2022] Open
Abstract
RA is a chronic, systemic, autoimmune disease characterized by inflammation and degradation of the joints, causing significant negative impact on quality of life. In addition to joint disease, symptoms and co-morbidities associated with RA-namely pain, fatigue and mood disorders-are often as debilitating as the disease itself. The pro-inflammatory cytokine IL-6 plays a critical role in RA-associated pathology. However, a greater understanding of the translational effects of IL-6 outside of the immune system is needed. This review discusses our current understanding of emerging aspects of IL-6 in RA-associated pain, fatigue and mood disorders such as depression and anxiety. This review also describes the clinical effects of IL-6 inhibition on these symptoms and co-morbidities in patients with RA.
Collapse
Affiliation(s)
- Ernest H S Choy
- Section of Rheumatology, Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| | | |
Collapse
|
49
|
North RY, Li Y, Ray P, Rhines LD, Tatsui CE, Rao G, Johansson CA, Zhang H, Kim YH, Zhang B, Dussor G, Kim TH, Price TJ, Dougherty PM. Electrophysiological and transcriptomic correlates of neuropathic pain in human dorsal root ganglion neurons. Brain 2019; 142:1215-1226. [PMID: 30887021 PMCID: PMC6487328 DOI: 10.1093/brain/awz063] [Citation(s) in RCA: 197] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 12/27/2018] [Accepted: 01/27/2019] [Indexed: 02/06/2023] Open
Abstract
Neuropathic pain encompasses a diverse array of clinical entities affecting 7-10% of the population, which is challenging to adequately treat. Several promising therapeutics derived from molecular discoveries in animal models of neuropathic pain have failed to translate following unsuccessful clinical trials suggesting the possibility of important cellular-level and molecular differences between animals and humans. Establishing the extent of potential differences between laboratory animals and humans, through direct study of human tissues and/or cells, is likely important in facilitating translation of preclinical discoveries to meaningful treatments. Patch-clamp electrophysiology and RNA-sequencing was performed on dorsal root ganglia taken from patients with variable presence of radicular/neuropathic pain. Findings establish that spontaneous action potential generation in dorsal root ganglion neurons is associated with radicular/neuropathic pain and radiographic nerve root compression. Transcriptome analysis suggests presence of sex-specific differences and reveals gene modules and signalling pathways in immune response and neuronal plasticity related to radicular/neuropathic pain that may suggest therapeutic avenues and that has the potential to predict neuropathic pain in future cohorts.
Collapse
Affiliation(s)
- Robert Y North
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas, USA
| | - Yan Li
- The Departments of Anesthesia and Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA, USA
| | - Pradipta Ray
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, Texas, USA
| | - Laurence D Rhines
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA, USA
| | - Claudio Esteves Tatsui
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA, USA
| | - Ganesh Rao
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA, USA
| | - Caj A Johansson
- The University of Texas Health Science Center, Houston, Texas, USA
| | - Hongmei Zhang
- The Departments of Anesthesia and Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA, USA
| | - Yeun Hee Kim
- Department of Biological Sciences and Center for Systems Biology, The University of Texas at Dallas, Richardson, Texas, USA
| | - Bo Zhang
- Department of Biological Sciences and Center for Systems Biology, The University of Texas at Dallas, Richardson, Texas, USA
| | - Gregory Dussor
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, Texas, USA
| | - Tae Hoon Kim
- Department of Biological Sciences and Center for Systems Biology, The University of Texas at Dallas, Richardson, Texas, USA
| | - Theodore J Price
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, Richardson, Texas, USA
| | - Patrick M Dougherty
- The Departments of Anesthesia and Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA, USA
| |
Collapse
|
50
|
Quarta S, Mitrić M, Kalpachidou T, Mair N, Schiefermeier-Mach N, Andratsch M, Qi Y, Langeslag M, Malsch P, Rose-John S, Kress M. Impaired mechanical, heat, and cold nociception in a murine model of genetic TACE/ADAM17 knockdown. FASEB J 2018; 33:4418-4431. [PMID: 30586315 PMCID: PMC6404580 DOI: 10.1096/fj.201801901r] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
TNF-α-converting enzyme, a member of the ADAM (A disintegrin and metalloproteinase) protease family and also known as ADAM17, regulates inflammation and regeneration in health and disease. ADAM17 targets are involved in pain development and hypersensitivity in animal models of inflammatory and neuropathic pain. However, the role of ADAM17 in the pain pathway is largely unknown. Therefore, we used the hypomorphic ADAM17 (ADAM17ex/ex) mouse model to investigate the importance of ADAM17 in nociceptive behavior, morphology, and function of primary afferent nociceptors. ADAM17ex/ex mice were hyposensitive to noxious stimulation, showing elevated mechanical thresholds as well as impaired heat and cold sensitivity. Despite these differences, skin thickness and innervation were comparable to controls. Although dorsal root ganglia of ADAM17ex/ex mice exhibited normal morphology of peptidergic and nonpeptidergic neurons, a small but significant reduction in the number of isolectin β-4–positive neurons was observed. Functional electrical properties of unmyelinated nociceptors showed differences in resting membrane potential, afterhyperpolarization, and firing patterns in specific subpopulations of sensory neurons in ADAM17ex/ex mice. However, spinal cord morphology and microglia activity in ADAM17ex/ex mice were not altered. Our data suggest that ADAM17 contributes to the processing of painful stimuli, with a complex mode of action orchestrating the function of neurons along the pain pathway.—Quarta, S., Mitrić, M., Kalpachidou, T., Mair, N., Schiefermeier-Mach, N., Andratsch, M., Qi, Y., Langeslag, M., Malsch, P., Rose-John, S., Kress, M. Impaired mechanical, heat, and cold nociception in a murine model of genetic TACE/ADAM17 knockdown.
Collapse
Affiliation(s)
- Serena Quarta
- Division of Physiology, Department of Physiology and Medical Physics, Innsbruck Medical University, Innsbruck, Austria; and
| | - Miodrag Mitrić
- Division of Physiology, Department of Physiology and Medical Physics, Innsbruck Medical University, Innsbruck, Austria; and
| | - Theodora Kalpachidou
- Division of Physiology, Department of Physiology and Medical Physics, Innsbruck Medical University, Innsbruck, Austria; and
| | - Norbert Mair
- Division of Physiology, Department of Physiology and Medical Physics, Innsbruck Medical University, Innsbruck, Austria; and
| | - Natalia Schiefermeier-Mach
- Division of Physiology, Department of Physiology and Medical Physics, Innsbruck Medical University, Innsbruck, Austria; and
| | - Manfred Andratsch
- Division of Physiology, Department of Physiology and Medical Physics, Innsbruck Medical University, Innsbruck, Austria; and
| | - Yanmei Qi
- Division of Physiology, Department of Physiology and Medical Physics, Innsbruck Medical University, Innsbruck, Austria; and
| | - Michiel Langeslag
- Division of Physiology, Department of Physiology and Medical Physics, Innsbruck Medical University, Innsbruck, Austria; and
| | - Philipp Malsch
- Division of Physiology, Department of Physiology and Medical Physics, Innsbruck Medical University, Innsbruck, Austria; and
| | - Stefan Rose-John
- Department of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Michaela Kress
- Division of Physiology, Department of Physiology and Medical Physics, Innsbruck Medical University, Innsbruck, Austria; and
| |
Collapse
|