1
|
Schaible HG, König C, Ebersberger A. Spinal pain processing in arthritis: Neuron and glia (inter)actions. J Neurochem 2024; 168:3644-3662. [PMID: 36520021 DOI: 10.1111/jnc.15742] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 12/02/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022]
Abstract
Diseases of joints are among the most frequent causes of chronic pain. In the course of joint diseases, the peripheral and the central nociceptive system develop persistent hyperexcitability (peripheral and central sensitization). This review addresses the mechanisms of spinal sensitization evoked by arthritis. Electrophysiological recordings in anesthetized rats from spinal cord neurons with knee input in a model of acute arthritis showed that acute spinal sensitization is dependent on spinal glutamate receptors (AMPA, NMDA, and metabotropic glutamate receptors) and supported by spinal actions of neuropeptides such as neurokinins and CGRP, by prostaglandins, and by proinflammatory cytokines. In several chronic arthritis models (including immune-mediated arthritis and osteoarthritis) spinal glia activation was observed to be coincident with behavioral mechanical hyperalgesia which was attenuated or prevented by intrathecal application of minocycline, fluorocitrate, and pentoxyfylline. Some studies identified specific pathways of micro- and astroglia activation such as the purinoceptor- (P2X7-) cathepsin S/CX3CR1 pathway, the mobility group box-1 protein (HMGB1), and toll-like receptor 4 (TLR4) activation, spinal NFκB/p65 activation and others. The spinal cytokines TNF, interleukin-6, interleukin-1β, and others form a functional spinal network characterized by an interaction between neurons and glia cells which is required for spinal sensitization. Neutralization of spinal cytokines by intrathecal interventions attenuates mechanical hyperalgesia. This effect may in part result from local suppression of spinal sensitization and in part from efferent effects which attenuate the inflammatory process in the joint. In summary, arthritis evokes significant spinal hyperexcitability which is likely to contribute to the phenotype of arthritis pain in patients.
Collapse
Affiliation(s)
- Hans-Georg Schaible
- Institute of Physiology 1/Neurophysiology, Jena University Hospital, Friedrich-Schiller-University of Jena, Jena, Germany
| | - Christian König
- Institute of Physiology 1/Neurophysiology, Jena University Hospital, Friedrich-Schiller-University of Jena, Jena, Germany
| | - Andrea Ebersberger
- Institute of Physiology 1/Neurophysiology, Jena University Hospital, Friedrich-Schiller-University of Jena, Jena, Germany
| |
Collapse
|
2
|
Tyagi S, Higerd-Rusli GP, Ghovanloo MR, Dib-Hajj F, Zhao P, Liu S, Kim DH, Shim JS, Park KS, Waxman SG, Choi JS, Dib-Hajj SD. Compartment-specific regulation of Na V1.7 in sensory neurons after acute exposure to TNF-α. Cell Rep 2024; 43:113685. [PMID: 38261513 PMCID: PMC10947185 DOI: 10.1016/j.celrep.2024.113685] [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: 08/08/2023] [Revised: 11/09/2023] [Accepted: 01/02/2024] [Indexed: 01/25/2024] Open
Abstract
Tumor necrosis factor α (TNF-α) is a major pro-inflammatory cytokine, important in many diseases, that sensitizes nociceptors through its action on a variety of ion channels, including voltage-gated sodium (NaV) channels. We show here that TNF-α acutely upregulates sensory neuron excitability and current density of threshold channel NaV1.7. Using electrophysiological recordings and live imaging, we demonstrate that this effect on NaV1.7 is mediated by p38 MAPK and identify serine 110 in the channel's N terminus as the phospho-acceptor site, which triggers NaV1.7 channel insertion into the somatic membrane. We also show that the N terminus of NaV1.7 is sufficient to mediate this effect. Although acute TNF-α treatment increases NaV1.7-carrying vesicle accumulation at axonal endings, we did not observe increased channel insertion into the axonal membrane. These results identify molecular determinants of TNF-α-mediated regulation of NaV1.7 in sensory neurons and demonstrate compartment-specific effects of TNF-α on channel insertion in the neuronal plasma membrane.
Collapse
Affiliation(s)
- Sidharth Tyagi
- Medical Scientist Training Program, Yale School of Medicine, New Haven, CT 06511, USA; Center for Neuroscience and Regeneration Research, West Haven, CT 06516, USA; Department of Neurology, Yale School of Medicine, New Haven, CT 06516, USA; Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT 06516, USA.
| | - Grant P Higerd-Rusli
- Medical Scientist Training Program, Yale School of Medicine, New Haven, CT 06511, USA; Center for Neuroscience and Regeneration Research, West Haven, CT 06516, USA; Department of Neurology, Yale School of Medicine, New Haven, CT 06516, USA; Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Mohammad-Reza Ghovanloo
- Center for Neuroscience and Regeneration Research, West Haven, CT 06516, USA; Department of Neurology, Yale School of Medicine, New Haven, CT 06516, USA; Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Fadia Dib-Hajj
- Center for Neuroscience and Regeneration Research, West Haven, CT 06516, USA; Department of Neurology, Yale School of Medicine, New Haven, CT 06516, USA; Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Peng Zhao
- Center for Neuroscience and Regeneration Research, West Haven, CT 06516, USA; Department of Neurology, Yale School of Medicine, New Haven, CT 06516, USA; Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Shujun Liu
- Center for Neuroscience and Regeneration Research, West Haven, CT 06516, USA; Department of Neurology, Yale School of Medicine, New Haven, CT 06516, USA; Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Dong-Hyun Kim
- Integrated Research Institute of Pharmaceutical Science, College of Pharmacy, The Catholic University of Korea, Bucheon 14662, South Korea; New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, South Korea
| | - Ji Seon Shim
- Department of Physiology, Kyung Hee University School of Medicine, Seoul 02447, South Korea
| | - Kang-Sik Park
- Department of Physiology, Kyung Hee University School of Medicine, Seoul 02447, South Korea
| | - Stephen G Waxman
- Center for Neuroscience and Regeneration Research, West Haven, CT 06516, USA; Department of Neurology, Yale School of Medicine, New Haven, CT 06516, USA; Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT 06516, USA.
| | - Jin-Sung Choi
- Integrated Research Institute of Pharmaceutical Science, College of Pharmacy, The Catholic University of Korea, Bucheon 14662, South Korea.
| | - Sulayman D Dib-Hajj
- Center for Neuroscience and Regeneration Research, West Haven, CT 06516, USA; Department of Neurology, Yale School of Medicine, New Haven, CT 06516, USA; Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT 06516, USA.
| |
Collapse
|
3
|
Chang JH, Chen KW, Tsai SY, Zeng YJ, Li CY, Chen KB, Wen YR. Role of ERK in gender difference of fibromyalgia pain. Mol Pain 2024; 20:17448069241261940. [PMID: 38818809 PMCID: PMC11168046 DOI: 10.1177/17448069241261940] [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: 09/22/2023] [Revised: 05/06/2024] [Accepted: 05/29/2024] [Indexed: 06/01/2024] Open
Abstract
This study investigated the ERK pathway of the peripheral nervous system and discovered a gender-specific pattern of ERK activation in the dorsal root ganglion of an acid-induced chronic widespread muscular pain model. We employed a twice acid-induced chronic musculoskeletal pain model in rats to evaluate mechanical pain behavior in both male and female groups. We further conducted protein analysis of dissected dorsal root ganglions from both genders. Both male and female rats exhibited a similar pain behavior trend, with females demonstrating a lower pain threshold. Protein analysis of the dorsal root ganglion (DRG) showed a significant increase in phosphorylated ERK after the second acid injection in all groups. However, phosphorylation of ERK was observed in the dorsal root ganglion, with higher levels in the male ipsilateral group compared to the female group. Moreover, there was a no difference between the left and right sides in males, whereas the significant difference was observed in females. In conclusions, the administration of acid injections induced painful behavior in rats, and concurrent with this, a significant upregulation of pERK was observed in the dorsal root ganglia, with a greater magnitude of increase in males than females, and in the contralateral side compared to the ipsilateral side. Our findings shed light on the peripheral mechanisms underlying chronic pain disorders and offer potential avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Ju-Hsin Chang
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
- Department of Anesthesiology, China Medical University Hospital, Taichung, Taiwan
| | - Ke-Wei Chen
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
- Division of Cardiology, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Shih-Ying Tsai
- Department of Anesthesiology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Yen-Jing Zeng
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
| | - Chi-Yuan Li
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
- Department of Anesthesiology, China Medical University Hospital, Taichung, Taiwan
| | - Kuen-Bao Chen
- Department of Anesthesiology, China Medical University Hospital, Taichung, Taiwan
| | - Yeong-Ray Wen
- Department of Anesthesiology, China Medical University Hospital, Taichung, Taiwan
- Department of Anesthesiology, School of Medicine, China Medical University, Taichung, Taiwan
- Graduate Institute of Acupuncture Science, College of Chinese Medicine, China Medical University, Taichung, Taiwan
- Acupuncture Research Center, China Medical University, Taichung, Taiwan
| |
Collapse
|
4
|
Spicarova D, Nerandzic V, Muzik D, Pontearso M, Bhattacharyya A, Nagy I, Palecek J. Inhibition of synaptic transmission by anandamide precursor 20:4-NAPE is mediated by TRPV1 receptors under inflammatory conditions. Front Mol Neurosci 2023; 16:1188503. [PMID: 37426071 PMCID: PMC10325575 DOI: 10.3389/fnmol.2023.1188503] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 05/24/2023] [Indexed: 07/11/2023] Open
Abstract
Transient receptor potential ion channel, vanilloid subfamily, type 1 (TRPV1) cation channel, and cannabinoid receptor 1 (CB1) are essential in the modulation of nociceptive signaling in the spinal cord dorsal horn that underlies different pathological pain states. TRPV1 and CB1 receptors share the endogenous agonist anandamide (AEA), produced from N-arachidonoylphosphatidylethanolamine (20:4-NAPE). We investigated the effect of the anandamide precursor 20:4-NAPE on synaptic activity in naive and inflammatory conditions. Patch-clamp recordings of miniature excitatory postsynaptic currents (mEPSCs) from superficial dorsal horn neurons in rat acute spinal cord slices were used. Peripheral inflammation was induced by subcutaneous injection of carrageenan. Under naive conditions, mEPSCs frequency (0.96 ± 0.11 Hz) was significantly decreased after 20 μM 20:4-NAPE application (55.3 ± 7.4%). This 20:4-NAPE-induced inhibition was blocked by anandamide-synthesizing enzyme N-acyl phosphatidylethanolamine phospholipase D (NAPE-PLD) inhibitor LEI-401. In addition, the inhibition was prevented by the CB1 receptor antagonist PF 514273 (0.2 μM) but not by the TRPV1 receptor antagonist SB 366791 (10 μM). Under inflammatory conditions, 20:4-NAPE (20 μM) also exhibited a significant inhibitory effect (74.5 ± 8.9%) on the mEPSCs frequency that was prevented by the TRPV1 receptor antagonist SB 366791 but not by PF 514273 application. Our results show that 20:4-NAPE application has a significant modulatory effect on spinal cord nociceptive signaling that is mediated by both TRPV1 and CB1 presynaptic receptors, whereas peripheral inflammation changes the underlying mechanism. The switch between TRPV1 and CB1 receptor activation by the AEA precursor 20:4-NAPE during inflammation may play an important role in nociceptive processing, hence the development of pathological pain.
Collapse
Affiliation(s)
- Diana Spicarova
- Laboratory of Pain Research, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| | - Vladimir Nerandzic
- Laboratory of Pain Research, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| | - David Muzik
- Laboratory of Pain Research, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| | - Monica Pontearso
- Laboratory of Pain Research, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| | - Anirban Bhattacharyya
- Laboratory of Pain Research, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| | - Istvan Nagy
- Section of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Imperial College London, Faculty of Medicine, Chelsea and Westminster Hospital, London, United Kingdom
- Department of Physiology, University of Debrecen, Debrecen, Hungary
| | - Jiri Palecek
- Laboratory of Pain Research, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
5
|
Ban D, Yu P, Xiang Z, Liu Y. TNF-like weak inducer of apoptosis / nuclear factor κB axis feedback loop promotes spinal cord injury by inducing astrocyte activation. Bioengineered 2022; 13:11503-11516. [PMID: 35506163 PMCID: PMC9275888 DOI: 10.1080/21655979.2022.2068737] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Non-canonical signaling pathways have been proved to act as potent sites of astrocytes osmotic expanding or proliferation, which promotes the regeneration of axons in areas with non-neural spinal cord injury (SCI). However, the relevant signal pathway that induces autophagic cell death in astrocytes and its function relative to the TNF-like weak inducer of apoptosis/nuclear factor κB (TWEAK/NF-κB) axis remains elusive. The SCI model was established by vertically striking the spinal cord according to Allen’s model. Astrocytes and neuronal cells were prepared from spinal cells extracted from spinal cord tissues of SCI or normal C57BL/6 newborn mice. After co-culturing astrocytes and neurons, cell viability and autophagy were determined by CCK-8, transmission electron microscopy (TEM), and western blot. The expression of TWEAK, NF-κB and inflammatory cytokines was confirmed by qRT-PCR, western blot, Immunofluorescence and ELISA assay. Chromatin immunoprecipitation (CHIP) was used to evaluate the interaction between TWEAK and NF-κB. Our results demonstrated that knockdown of TWEAK and NF-κB inhibited secretion of high levels of TNF-α/IL-1β, partially counteracted by adding Rap. TWEAK/NF-κB was the positive correlation feedback loop regulating the proliferation and autophagy of astrocytes involved in SCI. Moreover, restraining the excess growth of astrocytes was beneficial to the growth of neurons. Collectively, our findings illustrated that the TWEAK/NF-κB pathway might act as a positive modulator of SCI by inducing astrocyte activation, shedding new insights for SCI treatment.
Collapse
Affiliation(s)
- Dexiang Ban
- Department of Orthopaedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Peng Yu
- Department of Orthopaedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhenyang Xiang
- Department of Orthopaedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Yang Liu
- Department of Orthopaedics, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
6
|
Go EJ, Ji J, Kim YH, Berta T, Park CK. Transient Receptor Potential Channels and Botulinum Neurotoxins in Chronic Pain. Front Mol Neurosci 2021; 14:772719. [PMID: 34776867 PMCID: PMC8586451 DOI: 10.3389/fnmol.2021.772719] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 10/11/2021] [Indexed: 12/30/2022] Open
Abstract
Pain afflicts more than 1.5 billion people worldwide, with hundreds of millions suffering from unrelieved chronic pain. Despite widespread recognition of the importance of developing better interventions for the relief of chronic pain, little is known about the mechanisms underlying this condition. However, transient receptor potential (TRP) ion channels in nociceptors have been shown to be essential players in the generation and progression of pain and have attracted the attention of several pharmaceutical companies as therapeutic targets. Unfortunately, TRP channel inhibitors have failed in clinical trials, at least in part due to their thermoregulatory function. Botulinum neurotoxins (BoNTs) have emerged as novel and safe pain therapeutics because of their regulation of exocytosis and pro-nociceptive neurotransmitters. However, it is becoming evident that BoNTs also regulate the expression and function of TRP channels, which may explain their analgesic effects. Here, we summarize the roles of TRP channels in pain, with a particular focus on TRPV1 and TRPA1, their regulation by BoNTs, and briefly discuss the use of BoNTs for the treatment of chronic pain.
Collapse
Affiliation(s)
- Eun Jin Go
- Department of Physiology, Gachon Pain Center, Gachon University College of Medicine, Incheon, South Korea
| | - Jeongkyu Ji
- Gachon University College of Medicine, Incheon, South Korea
| | - Yong Ho Kim
- Department of Physiology, Gachon Pain Center, Gachon University College of Medicine, Incheon, South Korea
| | - Temugin Berta
- Department of Anesthesiology, Pain Research Center, University of Cincinnati Medical Center, Cincinnati, OH, United States
| | - Chul-Kyu Park
- Department of Physiology, Gachon Pain Center, Gachon University College of Medicine, Incheon, South Korea
| |
Collapse
|
7
|
O'Reilly ML, Mironets E, Shapiro TM, Crowther K, Collyer E, Bethea JR, Tom VJ. Pharmacological Inhibition of Soluble Tumor Necrosis Factor-Alpha Two Weeks after High Thoracic Spinal Cord Injury Does Not Affect Sympathetic Hyperreflexia. J Neurotrauma 2021; 38:2186-2191. [PMID: 33397170 PMCID: PMC8309421 DOI: 10.1089/neu.2020.7504] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
After a severe, high-level spinal cord injury (SCI), plasticity to intraspinal circuits below injury results in heightened spinal sympathetic reflex activity and detrimentally impacts peripheral organ systems. Such sympathetic hyperreflexia is immediately apparent as an episode of autonomic dysreflexia (AD), a life-threatening condition characterized by sudden hypertension and reflexive bradycardia following below-level sensory inputs; for example, pressure sores or impacted fecal matter. Over time, plasticity within the spinal sympathetic reflex (SSR) circuit contributes to the progressive intensification of AD events, as the frequency and severity of AD events increase greatly beginning ∼2 weeks post-injury (wpi). The neuroimmune system has been implicated in driving sympathetic hyperreflexia, as inhibition of the cytokine soluble tumor necrosis factor-alpha (sTNFα) using the biological mimetic XPro1595 beginning within days post-SCI has been shown to attenuate the development of AD. Here, we sought to further understand the effective therapeutic time window of XPro1595 to diminish sympathetic hyperreflexia, as indicated by AD. We delayed the commencement of continuous intrathecal administration of XPro1595 until 2 weeks after a complete, thoracic level 3 injury in adult rats. We examined the severity of colorectal distension-induced AD biweekly. We found that initiation of sTNFα inhibition at 2 wpi does not attenuate the severity or intensification of sympathetic hyperreflexia compared with saline-treated controls. Coupled with previous data from our group, these findings suggest that central sTNFα signaling must be targeted prior to 2 weeks post-SCI in order to decrease sympathetic hyperreflexia.
Collapse
Affiliation(s)
- Micaela L. O'Reilly
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Eugene Mironets
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Tatiana M. Shapiro
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Kallon Crowther
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Eileen Collyer
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - John R. Bethea
- Department of Biology, Drexel University, Philadelphia, Pennsylvania, USA
| | - Veronica J. Tom
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
8
|
Meng J, Li Y, Fischer MJM, Steinhoff M, Chen W, Wang J. Th2 Modulation of Transient Receptor Potential Channels: An Unmet Therapeutic Intervention for Atopic Dermatitis. Front Immunol 2021; 12:696784. [PMID: 34276687 PMCID: PMC8278285 DOI: 10.3389/fimmu.2021.696784] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 06/15/2021] [Indexed: 12/12/2022] Open
Abstract
Atopic dermatitis (AD) is a multifaceted, chronic relapsing inflammatory skin disease that affects people of all ages. It is characterized by chronic eczema, constant pruritus, and severe discomfort. AD often progresses from mild annoyance to intractable pruritic inflammatory lesions associated with exacerbated skin sensitivity. The T helper-2 (Th2) response is mainly linked to the acute and subacute phase, whereas Th1 response has been associated in addition with the chronic phase. IL-17, IL-22, TSLP, and IL-31 also play a role in AD. Transient receptor potential (TRP) cation channels play a significant role in neuroinflammation, itch and pain, indicating neuroimmune circuits in AD. However, the Th2-driven cutaneous sensitization of TRP channels is underappreciated. Emerging findings suggest that critical Th2-related cytokines cause potentiation of TRP channels, thereby exaggerating inflammation and itch sensation. Evidence involves the following: (i) IL-13 enhances TRPV1 and TRPA1 transcription levels; (ii) IL-31 sensitizes TRPV1 via transcriptional and channel modulation, and indirectly modulates TRPV3 in keratinocytes; (iii) The Th2-cytokine TSLP increases TRPA1 synthesis in sensory neurons. These changes could be further enhanced by other Th2 cytokines, including IL-4, IL-25, and IL-33, which are inducers for IL-13, IL-31, or TSLP in skin. Taken together, this review highlights that Th2 cytokines potentiate TRP channels through diverse mechanisms under different inflammatory and pruritic conditions, and link this effect to distinct signaling cascades in AD. This review strengthens the notion that interrupting Th2-driven modulation of TRP channels will inhibit transition from acute to chronic AD, thereby aiding the development of effective therapeutics and treatment optimization.
Collapse
Affiliation(s)
- Jianghui Meng
- School of Life Sciences, Henan University, Kaifeng, China.,National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
| | - Yanqing Li
- School of Life Sciences, Henan University, Kaifeng, China
| | - Michael J M Fischer
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Martin Steinhoff
- Department of Dermatology and Venereology, Hamad Medical Corporation, Doha, Qatar.,Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar.,Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar.,Department of Dermatology, Weill Cornell Medicine-Qatar, Doha, Qatar.,Qatar University, College of Medicine, Doha, Qatar.,Department of Dermatology, Weill Cornell Medicine, New York, NY, United States
| | - Weiwei Chen
- School of Life Sciences, Henan University, Kaifeng, China
| | - Jiafu Wang
- School of Life Sciences, Henan University, Kaifeng, China.,School of Biotechnology, Faculty of Science and Health, Dublin City University, Dublin, Ireland
| |
Collapse
|
9
|
Schinke C, Fernandez Vallone V, Ivanov A, Peng Y, Körtvelyessy P, Nolte L, Huehnchen P, Beule D, Stachelscheid H, Boehmerle W, Endres M. Modeling chemotherapy induced neurotoxicity with human induced pluripotent stem cell (iPSC) -derived sensory neurons. Neurobiol Dis 2021; 155:105391. [PMID: 33984509 DOI: 10.1016/j.nbd.2021.105391] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 05/06/2021] [Accepted: 05/07/2021] [Indexed: 12/20/2022] Open
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a frequent, potentially irreversible adverse effect of cytotoxic chemotherapy often leading to a reduction or discontinuation of treatment which negatively impacts patients' prognosis. To date, however, neither predictive biomarkers nor preventive treatments for CIPN are available, which is partially due to a lack of suitable experimental models. We therefore aimed to evaluate whether sensory neurons derived from induced pluripotent stem cells (iPSC-DSN) can serve as human disease model system for CIPN. Treatment of iPSC-DSN for 24 h with the neurotoxic drugs paclitaxel, bortezomib, vincristine and cisplatin led to axonal blebbing and a dose dependent decline of cell viability in clinically relevant IC50 ranges, which was not observed for the non-neurotoxic compounds doxorubicin and 5-fluorouracil. Paclitaxel treatment effects were less pronounced after 24 h but prominent when treatment was applied for 72 h. Global transcriptome analyses performed at 24 h, i.e. before paclitaxel-induced cell death occurred, revealed the differential expression of genes of neuronal injury, cellular stress response, and sterol pathways. We further evaluated if known neuroprotective strategies can be reproduced in iPSC-DSN and observed protective effects of lithium replicating findings from rodent dorsal root ganglia cells. Comparing sensory neurons derived from two different healthy donors, we found preliminary evidence that these cell lines react differentially to neurotoxic drugs as expected from the variable presentation of CIPN in patients. In conclusion, iPSC-DSN are a promising platform to study the pathogenesis of CIPN and to evaluate neuroprotective treatment strategies. In the future, the application of patient-specific iPSC-DSN could open new avenues for personalized medicine with individual risk prediction, choice of chemotherapeutic compounds and preventive treatments.
Collapse
Affiliation(s)
- Christian Schinke
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Klinik und Hochschulambulanz für Neurologie, Charitéplatz 1, 10117 Berlin, Germany; Berlin Institute of Health at Charité, Universitätsmedizin Berlin, Anna-Louisa-Karsch Straße 2, 10178 Berlin, Germany
| | - Valeria Fernandez Vallone
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Stem Cell Core Facility, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Andranik Ivanov
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Core Unit Bioinformatics, Charitéplatz 1, 10117 Berlin, Germany
| | - Yangfan Peng
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Klinik und Hochschulambulanz für Neurologie, Charitéplatz 1, 10117 Berlin, Germany; Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institut für Neurophysiologie, Charitéplatz 1, 10117 Berlin, Germany
| | - Péter Körtvelyessy
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Klinik und Hochschulambulanz für Neurologie, Charitéplatz 1, 10117 Berlin, Germany; Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Neuropathology, Charitéplatz 1, 10117 Berlin, Germany; German Center for Neurodegenerative Diseases, 39120 Magdeburg, Germany
| | - Luca Nolte
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Klinik und Hochschulambulanz für Neurologie, Charitéplatz 1, 10117 Berlin, Germany
| | - Petra Huehnchen
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Klinik und Hochschulambulanz für Neurologie, Charitéplatz 1, 10117 Berlin, Germany; Berlin Institute of Health at Charité, Universitätsmedizin Berlin, Anna-Louisa-Karsch Straße 2, 10178 Berlin, Germany; Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, NeuroCure Cluster of Excellence, Charitéplatz 1, 10117 Berlin, Germany
| | - Dieter Beule
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Core Unit Bioinformatics, Charitéplatz 1, 10117 Berlin, Germany; Max-Delbrueck Center for Molecular Medicine, 13125 Berlin, Germany
| | - Harald Stachelscheid
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Stem Cell Core Facility, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Wolfgang Boehmerle
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Klinik und Hochschulambulanz für Neurologie, Charitéplatz 1, 10117 Berlin, Germany; Berlin Institute of Health at Charité, Universitätsmedizin Berlin, Anna-Louisa-Karsch Straße 2, 10178 Berlin, Germany; Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, NeuroCure Cluster of Excellence, Charitéplatz 1, 10117 Berlin, Germany.
| | - Matthias Endres
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Klinik und Hochschulambulanz für Neurologie, Charitéplatz 1, 10117 Berlin, Germany; Berlin Institute of Health at Charité, Universitätsmedizin Berlin, Anna-Louisa-Karsch Straße 2, 10178 Berlin, Germany; Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, NeuroCure Cluster of Excellence, Charitéplatz 1, 10117 Berlin, Germany; Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Center for Stroke Research Berlin, Charitéplatz 1, 10117 Berlin, Germany; German Center for Neurodegenerative Diseases (DZNE), partner site Berlin, Germany; German Center for Cardiovascular Research (DZHK), partner site Berlin, Germany
| |
Collapse
|
10
|
Uchytilova E, Spicarova D, Palecek J. Hypersensitivity Induced by Intrathecal Bradykinin Administration Is Enhanced by N-oleoyldopamine (OLDA) and Prevented by TRPV1 Antagonist. Int J Mol Sci 2021; 22:3712. [PMID: 33918267 PMCID: PMC8038144 DOI: 10.3390/ijms22073712] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 03/29/2021] [Accepted: 03/31/2021] [Indexed: 02/06/2023] Open
Abstract
Transient receptor potential vanilloid 1 (TRPV1) channels contribute to the development of several chronic pain states and represent a possible therapeutic target in many painful disease treatment. Proinflammatory mediator bradykinin (BK) sensitizes TRPV1, whereas noxious peripheral stimulation increases BK level in the spinal cord. Here, we investigated the involvement of spinal TRPV1 in thermal and mechanical hypersensitivity, evoked by intrathecal (i.t.) administration of BK and an endogenous agonist of TRPV1, N-oleoyldopamine (OLDA), using behavioral tests and i.t. catheter implantation, and administration of BK-induced transient thermal and mechanical hyperalgesia and mechanical allodynia. All these hypersensitive states were enhanced by co-administration of a low dose of OLDA (0.42 µg i.t.), which was ineffective only under the control conditions. Intrathecal pretreatment with TRPV1 selective antagonist SB366791 prevented hypersensitivity induced by i.t. co-administration of BK and OLDA. Our results demonstrate that both thermal and mechanical hypersensitivity evoked by co-administration of BK and OLDA is mediated by the activation of spinal TRPV1 channels.
Collapse
Affiliation(s)
- Eva Uchytilova
- Laboratory of Pain Research, Institute of Physiology, Czech Academy of Sciences, Videnska 1083, 14220 Prague, Czech Republic;
- Department of Anaesthesiology, Resuscitation and Critical Care, Institute for Clinical and Experimental Medicine, Videnska 1958/9, 14021 Prague, Czech Republic
| | - Diana Spicarova
- Laboratory of Pain Research, Institute of Physiology, Czech Academy of Sciences, Videnska 1083, 14220 Prague, Czech Republic;
| | - Jiri Palecek
- Laboratory of Pain Research, Institute of Physiology, Czech Academy of Sciences, Videnska 1083, 14220 Prague, Czech Republic;
| |
Collapse
|
11
|
Spinal PAR2 Activation Contributes to Hypersensitivity Induced by Peripheral Inflammation in Rats. Int J Mol Sci 2021; 22:ijms22030991. [PMID: 33498178 PMCID: PMC7863954 DOI: 10.3390/ijms22030991] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/16/2021] [Accepted: 01/17/2021] [Indexed: 11/17/2022] Open
Abstract
The mechanisms of inflammatory pain need to be identified in order to find new superior treatments. Protease-activated receptors 2 (PAR2) and transient receptor potential vanilloid 1 (TRPV1) are highly co-expressed in dorsal root ganglion neurons and implicated in pain development. Here, we examined the role of spinal PAR2 in hyperalgesia and the modulation of synaptic transmission in carrageenan-induced peripheral inflammation, using intrathecal (i.t.) treatment in the behavioral experiments and recordings of spontaneous, miniature and dorsal root stimulation-evoked excitatory postsynaptic currents (sEPSCs, mEPSCs and eEPSCs) in spinal cord slices. Intrathecal PAR2-activating peptide (AP) administration aggravated the carrageenan-induced thermal hyperalgesia, and this was prevented by a TRPV1 antagonist (SB 366791) and staurosporine i.t. pretreatment. Additionally, the frequency of the mEPSC and sEPSC and the amplitude of the eEPSC recorded from the superficial dorsal horn neurons were enhanced after acute PAR2 AP application, while prevented with SB 366791 or staurosporine pretreatment. PAR2 antagonist application reduced the thermal hyperalgesia and decreased the frequency of mEPSC and sEPSC and the amplitude of eEPSC. Our findings highlight the contribution of spinal PAR2 activation to carrageenan-induced hyperalgesia and the importance of dorsal horn PAR2 and TRPV1 receptor interactions in the modulation of nociceptive synaptic transmission.
Collapse
|
12
|
Stem Cells from Human Exfoliated Deciduous Teeth Attenuate Trigeminal Neuralgia in Rats. Stem Cells Int 2021; 2021:8819884. [PMID: 33531911 PMCID: PMC7834821 DOI: 10.1155/2021/8819884] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 01/01/2021] [Accepted: 01/07/2021] [Indexed: 12/12/2022] Open
Abstract
Trigeminal neuralgia is an incurable progressive nervous system disease that can last for several months or years. Stem cells from human exfoliated deciduous teeth (SHED) are a candidate source for cell-based therapy. Owing to their neuroprotective and immunomodulatory effects, these neural crest cells have potential roles in mediating chronic pain. In this study, we established a rat model of chronic constriction injury of the infraorbital nerve (CCI-ION) to evaluate the analgesic effect of SHED in neuropathic pain. The effects of local SHED transplantation on inflammatory cell infiltration in the trigeminal nerve were investigated based on hematoxylin and eosin staining. The levels of proinflammatory factors in the injured nerve and transient receptor potential vanilloid type 1 (TRPV1) expression in the trigeminal nerve and ganglion were quantified. The data showed that systemic or local injection of SHED attenuated the sensitivity of rats to mechanical stimuli after nerve injury, and this effect lasted throughout the observation period of 8 weeks. PKH26-labeled SHED were distributed to the ipsilateral trigeminal ganglions 24 and 72 hours after local injection. SHED transplantation at the lesion site led to reduced inflammatory cell infiltration and proinflammatory cytokine levels in the injured nerve and inhibited CCI-ION-induced upregulation of TRPV1 expression in the trigeminal nerve and ganglion in the early phase. Therefore, these results provide preclinical evidence that supports the use of SHED in the treatment of trigeminal neuralgia and potentially other chronic pain conditions.
Collapse
|
13
|
Roh J, Go EJ, Park JW, Kim YH, Park CK. Resolvins: Potent Pain Inhibiting Lipid Mediators via Transient Receptor Potential Regulation. Front Cell Dev Biol 2020; 8:584206. [PMID: 33363143 PMCID: PMC7758237 DOI: 10.3389/fcell.2020.584206] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 11/20/2020] [Indexed: 12/15/2022] Open
Abstract
Chronic pain is a serious condition that occurs in the peripheral nervous system (PNS) and the central nervous system (CNS). It is caused by inflammation or nerve damage that induces the release of inflammatory mediators from immune cells and/or protein kinase activation in neuronal cells. Both nervous systems are closely linked; therefore, inflammation or nerve damage in the PNS can affect the CNS (central sensitization). In this process, nociceptive transient receptor potential (TRP) channel activation and expression are increased. As a result, nociceptive neurons are activated, and pain signals to the brain are amplified and prolonged. In other words, suppressing the onset of pain signals in the PNS can suppress pain signals to the CNS. Resolvins, endogenous lipid mediators generated during the resolution phase of acute inflammation, inhibit nociceptive TRP ion channels and alleviate chronic pain. This paper summarizes the effect of resolvins in chronic pain control and discusses future scientific perspectives. Further study on the effect of resolvins on neuropathic pain will expand the scope of pain research.
Collapse
Affiliation(s)
- Jueun Roh
- Gachon Pain Center and Department of Physiology, College of Medicine, Gachon University, Incheon, South Korea
| | - Eun Jin Go
- Gachon Pain Center and Department of Physiology, College of Medicine, Gachon University, Incheon, South Korea
| | - Jin-Woo Park
- Department of Periodontology, School of Dentistry, Kyungpook National University, Daegu, South Korea
| | - Yong Ho Kim
- Gachon Pain Center and Department of Physiology, College of Medicine, Gachon University, Incheon, South Korea
| | - Chul-Kyu Park
- Gachon Pain Center and Department of Physiology, College of Medicine, Gachon University, Incheon, South Korea
| |
Collapse
|
14
|
O'Reilly ML, Tom VJ. Neuroimmune System as a Driving Force for Plasticity Following CNS Injury. Front Cell Neurosci 2020; 14:187. [PMID: 32792908 PMCID: PMC7390932 DOI: 10.3389/fncel.2020.00187] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 05/29/2020] [Indexed: 12/15/2022] Open
Abstract
Following an injury to the central nervous system (CNS), spontaneous plasticity is observed throughout the neuraxis and affects multiple key circuits. Much of this spontaneous plasticity can elicit beneficial and deleterious functional outcomes, depending on the context of plasticity and circuit affected. Injury-induced activation of the neuroimmune system has been proposed to be a major factor in driving this plasticity, as neuroimmune and inflammatory factors have been shown to influence cellular, synaptic, structural, and anatomical plasticity. Here, we will review the mechanisms through which the neuroimmune system mediates plasticity after CNS injury. Understanding the role of specific neuroimmune factors in driving adaptive and maladaptive plasticity may offer valuable therapeutic insight into how to promote adaptive plasticity and/or diminish maladaptive plasticity, respectively.
Collapse
Affiliation(s)
- Micaela L O'Reilly
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Veronica J Tom
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, PA, United States
| |
Collapse
|
15
|
Kalynovska N, Diallo M, Sotakova-Kasparova D, Palecek J. Losartan attenuates neuroinflammation and neuropathic pain in paclitaxel-induced peripheral neuropathy. J Cell Mol Med 2020; 24:7949-7958. [PMID: 32485058 PMCID: PMC7348151 DOI: 10.1111/jcmm.15427] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 01/15/2020] [Accepted: 05/08/2020] [Indexed: 12/16/2022] Open
Abstract
Paclitaxel-induced peripheral neuropathy (PIPN) is often associated with neuropathic pain and neuroinflammation in the central and peripheral nervous system. Antihypertensive drug losartan, an angiotensin II receptor type 1 (AT1R) blocker, was shown to have anti-inflammatory and neuroprotective effects in disease models, predominantly via activation of peroxisome proliferator-activated receptor gamma (PPARγ). Here, the effect of systemic losartan treatment (100 mg/kg/d) on mechanical allodynia and neuroinflammation was evaluated in rat PIPN model. The expression of pro-inflammatory markers protein and mRNA levels in dorsal root ganglia (DRGs) and spinal cord dorsal horn (SCDH) were measured with Western blot, ELISA and qPCR 10 and 21 days after PIPN induction. Losartan treatment attenuated mechanical allodynia significantly. Paclitaxel induced overexpression of C-C motif chemokine ligand 2 (CCL2), tumour necrosis alpha (TNFα) and interleukin-6 (IL-6) in DRGs, where the presence of macrophages was demonstrated. Neuroinflammatory changes in DRGs were accompanied with glial activation and pro-nociceptive modulators production in SCDH. Losartan significantly attenuated paclitaxel-induced neuroinflammatory changes and induced expression of pro-resolving markers (Arginase 1 and IL-10) indicating a possible shift in macrophage polarization. Considering the safety profile of losartan, acting also as partial PPARγ agonist, it may be considered as a novel treatment strategy for PIPN patients.
Collapse
Affiliation(s)
- Nataliia Kalynovska
- Department of Functional Morphology, Institute of Physiology, The Czech Academy of Sciences, Prague, Czech Republic
| | - Mickael Diallo
- Department of Functional Morphology, Institute of Physiology, The Czech Academy of Sciences, Prague, Czech Republic
| | - Dita Sotakova-Kasparova
- Department of Functional Morphology, Institute of Physiology, The Czech Academy of Sciences, Prague, Czech Republic
| | - Jiri Palecek
- Department of Functional Morphology, Institute of Physiology, The Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
16
|
Losartan treatment attenuates the development of neuropathic thermal hyperalgesia induced by peripheral nerve injury in rats. Life Sci 2019; 220:147-155. [DOI: 10.1016/j.lfs.2019.02.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 01/25/2019] [Accepted: 02/02/2019] [Indexed: 12/19/2022]
|
17
|
Adamek P, Heles M, Palecek J. Mechanical allodynia and enhanced responses to capsaicin are mediated by PI3K in a paclitaxel model of peripheral neuropathy. Neuropharmacology 2018; 146:163-174. [PMID: 30471295 DOI: 10.1016/j.neuropharm.2018.11.027] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 11/16/2018] [Accepted: 11/19/2018] [Indexed: 12/17/2022]
Abstract
Paclitaxel chemotherapy treatment often leads to neuropathic pain resistant to available analgesic treatments. Recently spinal Toll-like receptor 4 (TLR4) and the transient receptor potential cation channel subfamily V member 1 (TRPV1) were identified to be involved in the pro-nociceptive effect of paclitaxel. The aim of this study was to investigate the role of phosphatidylinositol 3-kinase (PI3K) and serine/threonine kinases in this process, with the use of their antagonists (wortmannin, LY-294002, and staurosporine). The single paclitaxel administration (8 mg/kg i.p.) in mice induced robust mechanical allodynia measured as a reduced threshold to von Frey filament stimulation and generated reduced tachyphylaxis of capsaicin-evoked responses, recorded as changes in mEPSC frequency in patch-clamp recordings of dorsal horn neurons activity in vitro, for up to eight days. Paclitaxel application also induced increased Akt kinase phosphorylation in rat DRG neurons. All these paclitaxel-induced changes were prevented by the wortmannin in vivo pretreatment. Acute co-application of wortmannin or LY-294002 with paclitaxel in spinal cord slices also attenuated the paclitaxel effect on capsaicin-evoked responses. Staurosporine was effective in the acute in vitro experiments and on the first day after the paclitaxel treatment in vivo, but in contrast to wortmannin, it did not have a significant impact later. Our data suggest that the inhibition of PI3K signaling may help alleviate pathological pain syndromes in the paclitaxel-induced neuropathy.
Collapse
Affiliation(s)
- Pavel Adamek
- Department of Functional Morphology, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, Prague, 14220, Czech Republic; Department of Physiology, Faculty of Science, Charles University in Prague, Vinicna 7, Prague, 128 44, Czech Republic
| | - Mario Heles
- Department of Functional Morphology, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, Prague, 14220, Czech Republic; Department of Physiology, Faculty of Science, Charles University in Prague, Vinicna 7, Prague, 128 44, Czech Republic
| | - Jiri Palecek
- Department of Functional Morphology, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, Prague, 14220, Czech Republic.
| |
Collapse
|
18
|
Kalynovska N, Adamek P, Palecek J. TRPV1 receptors contribute to paclitaxel-induced c-Fos expression in spinal cord dorsal horn neurons. Physiol Res 2018; 66:549-552. [PMID: 28730839 DOI: 10.33549/physiolres.933613] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Transient receptor potential vanilloid type 1 (TRPV1) receptors are important in the development of different pathological chronic pain states. Here we examined the role of spinal cord TRPV1 receptors in the mechanisms leading to activation of dorsal horn neurons after paclitaxel (PAC) treatment. PAC is a widely used chemotherapeutic drug that often leads to development of painful neuropathy. Immunohistochemical analysis of c-Fos protein expression in dorsal horn neurons was used as a marker of neuronal activation. Rat spinal cord slices were processed for in vitro incubation with PAC (100 nM) and TRPV1 receptor antagonists (SB366791 and AMG9810; 10 microM). PAC treatment induced significant upregulation of c-Fos nuclear expression in superficial dorsal horn neurons that was diminished by TRPV1 receptor antagonists pre-incubation. These results further substantiated the role of spinal TRPV1 receptors in the development of paclitaxel-induced neuropathic pain and contribute to better understanding of the pathological mechanisms involved.
Collapse
Affiliation(s)
- N Kalynovska
- Department of Functional Morphology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic.
| | | | | |
Collapse
|
19
|
Medelin M, Giacco V, Aldinucci A, Castronovo G, Bonechi E, Sibilla A, Tanturli M, Torcia M, Ballerini L, Cozzolino F, Ballerini C. Bridging pro-inflammatory signals, synaptic transmission and protection in spinal explants in vitro. Mol Brain 2018; 11:3. [PMID: 29334986 PMCID: PMC5769440 DOI: 10.1186/s13041-018-0347-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 01/04/2018] [Indexed: 01/30/2023] Open
Abstract
Multiple sclerosis is characterized by tissue atrophy involving the brain and the spinal cord, where reactive inflammation contributes to the neurodegenerative processes. Recently, the presence of synapse alterations induced by the inflammatory responses was suggested by experimental and clinical observations, in experimental autoimmune encephalomyelitis mouse model and in patients, respectively. Further knowledge on the interplay between pro-inflammatory agents, neuroglia and synaptic dysfunction is crucial to the design of unconventional protective molecules. Here we report the effects, on spinal cord circuits, of a cytokine cocktail that partly mimics the signature of T lymphocytes sub population Th1. In embryonic mouse spinal organ-cultures, containing neuronal cells and neuroglia, cytokines induced inflammatory responses accompanied by a significant increase in spontaneous synaptic activity. We suggest that cytokines specifically altered signal integration in spinal networks by speeding the decay of GABAA responses. This hypothesis is supported by the finding that synapse protection by a non-peptidic NGF mimetic molecule prevented both the changes in the time course of GABA events and in network activity that were left unchanged by the cytokine production from astrocytes and microglia present in the cultured tissue. In conclusion, we developed an important tool for the study of synaptic alterations induced by inflammation, that takes into account the role of neuronal and not neuronal resident cells.
Collapse
Affiliation(s)
- M Medelin
- Department of Life Sciences, University of Trieste, 34127, Trieste, Italy.,International School for Advanced Studies (SISSA/ISAS), 34136, Trieste, Italy
| | - V Giacco
- International School for Advanced Studies (SISSA/ISAS), 34136, Trieste, Italy
| | - A Aldinucci
- Department NEUROFARBA, University of Florence, 50139, Florence, Italy
| | - G Castronovo
- Department of DSBSC, University of Florence, 50134, Florence, Italy
| | - E Bonechi
- Department NEUROFARBA, University of Florence, 50139, Florence, Italy
| | - A Sibilla
- Department NEUROFARBA, University of Florence, 50139, Florence, Italy
| | - M Tanturli
- Department of DSBSC, University of Florence, 50134, Florence, Italy
| | - M Torcia
- Department of DMSC, University of Florence, 50134, Florence, Italy
| | - L Ballerini
- International School for Advanced Studies (SISSA/ISAS), 34136, Trieste, Italy.
| | - F Cozzolino
- Department of DSBSC, University of Florence, 50134, Florence, Italy
| | - C Ballerini
- Department NEUROFARBA, University of Florence, 50139, Florence, Italy.
| |
Collapse
|
20
|
Involvement of Spinal IL-6 Trans-Signaling in the Induction of Hyperexcitability of Deep Dorsal Horn Neurons by Spinal Tumor Necrosis Factor-Alpha. J Neurosci 2017; 36:9782-91. [PMID: 27656018 DOI: 10.1523/jneurosci.4159-15.2016] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 07/21/2016] [Indexed: 12/30/2022] Open
Abstract
UNLABELLED During peripheral inflammation, both spinal TNF-α and IL-6 are released within the spinal cord and support the generation of inflammation-evoked spinal hyperexcitability. However, whether spinal TNF-α and IL-6 act independently in parallel or in a functionally dependent manner has not been investigated. In extracellular recordings from mechanonociceptive deep dorsal horn neurons of normal rats in vivo, we found that spinal application of TNF-α increased spinal neuronal responses to mechanical stimulation of knee and ankle joints. This effect was significantly attenuated by either sgp130, which blocks IL-6 trans-signaling mediated by IL-6 and its soluble receptor IL-6R (sIL-6R); by an antibody to the IL-6 receptor; or by minocycline, which inhibits the microglia. IL-6 was localized in neurons of the spinal cord and, upon peripheral noxious stimulation in the presence of spinal TNF-α, IL-6 was released spinally. Furthermore, TNF-α recruited microglial cells to provide sIL-6R, which can form complexes with IL-6. Spinal application of IL-6 plus sIL-6R, but not of IL-6 alone, enhanced spinal hyperexcitability similar to TNF-α and the inhibition of TNF-α-induced hyperexcitability by minocycline was overcome by coadministration of sIL-6R, showing that sIL-6R is required. Neither minocycline nor the TNF-α-neutralizing compound etanercept inhibited the induction of hyperexcitability by IL-6 plus sIL-6R. Together, these data show that the induction of hyperexcitability of nociceptive deep dorsal horn neurons by TNF-α largely depends on the formation of IL-6/sIL-6R complexes that are downstream of TNF-α and requires the interactions of neurons and microglia orchestrated by TNF-α. SIGNIFICANCE STATEMENT Both spinal TNF-α and IL-6 induce a state of spinal hyperexcitability. We present the novel finding that the full effect of TNF-α on the development of spinal hyperexcitability depends on IL-6 trans-signaling acting downstream of TNF-α. IL-6 trans-signaling requires the formation of complexes of IL-6 and soluble IL-6 receptor. Spinal TNF-α furthers the release of IL-6 from neurons in the spinal cord during peripheral noxious stimulation and recruits microglial cells to provide soluble IL-6 receptor, which can form complexes with IL-6. Therefore, a specific interaction between neurons and microglia is required for the full development of TNF-α-induced hyperexcitability of nociceptive deep horsal horn neurons.
Collapse
|
21
|
Nerandzic V, Mrozkova P, Adamek P, Spicarova D, Nagy I, Palecek J. Peripheral inflammation affects modulation of nociceptive synaptic transmission in the spinal cord induced by N-arachidonoylphosphatidylethanolamine. Br J Pharmacol 2017; 175:2322-2336. [PMID: 28476070 DOI: 10.1111/bph.13849] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 04/07/2017] [Accepted: 04/27/2017] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Endocannabinoids play an important role in modulating spinal nociceptive signalling, crucial for the development of pain. The cannabinoid CB1 receptor and the TRPV1 cation channel are both activated by the endocannabinoid anandamide, a product of biosynthesis from the endogenous lipid precursor N-arachidonoylphosphatidylethanolamine (20:4-NAPE). Here, we report CB1 receptor- and TRPV1-mediated effects of 20:4-NAPE on spinal synaptic transmission in control and inflammatory conditions. EXPERIMENTAL APPROACH Spontaneous (sEPSCs) and dorsal root stimulation-evoked (eEPSCs) excitatory postsynaptic currents from superficial dorsal horn neurons in rat spinal cord slices were assessed. Peripheral inflammation was induced by carrageenan. Anandamide concentration was assessed by mass spectrometry. KEY RESULTS Application of 20:4-NAPE increased anandamide concentration in vitro. 20:4-NAPE (20 μM) decreased sEPSCs frequency and eEPSCs amplitude in control and inflammatory conditions. The inhibitory effect of 20:4-NAPE was sensitive to CB1 receptor antagonist PF514273 (0.2 μM) in both conditions, but to the TRPV1 antagonist SB366791 (10 μM) only after inflammation. After inflammation, 20:4-NAPE increased sEPSCs frequency in the presence of PF514273 and this increase was blocked by SB366791. CONCLUSIONS AND IMPLICATIONS While 20:4-NAPE treatment inhibited the excitatory synaptic transmission in both naive and inflammatory conditions, peripheral inflammation altered the underlying mechanisms. Our data indicate that 20:4-NAPE application induced mainly CB1 receptor-mediated inhibitory effects in naive animals while TRPV1-mediated mechanisms were also involved after inflammation. Increasing anandamide levels for analgesic purposes by applying substrate for its local synthesis may be more effective than systemic anandamide application or inhibition of its degradation. LINKED ARTICLES This article is part of a themed section on Recent Advances in Targeting Ion Channels to Treat Chronic Pain. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.12/issuetoc.
Collapse
Affiliation(s)
- Vladimir Nerandzic
- Department of Functional Morphology, Institute of Physiology, The Czech Academy of Sciences, Prague, Czech Republic
| | - Petra Mrozkova
- Department of Functional Morphology, Institute of Physiology, The Czech Academy of Sciences, Prague, Czech Republic
| | - Pavel Adamek
- Department of Functional Morphology, Institute of Physiology, The Czech Academy of Sciences, Prague, Czech Republic.,Faculty of Science, Charles University, Prague, Czech Republic
| | - Diana Spicarova
- Department of Functional Morphology, Institute of Physiology, The Czech Academy of Sciences, Prague, Czech Republic
| | - Istvan Nagy
- Section of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer Imperial College London, Faculty of Medicine, Chelsea and Westminster Hospital, London, UK
| | - Jiri Palecek
- Department of Functional Morphology, Institute of Physiology, The Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
22
|
TRP channels: potential drug target for neuropathic pain. Inflammopharmacology 2016; 24:305-317. [PMID: 27757589 DOI: 10.1007/s10787-016-0288-x] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 10/05/2016] [Indexed: 01/06/2023]
Abstract
Neuropathic pain is a debilitating disease which affects central as well as peripheral nervous system. Transient receptor potential (TRP) channels are ligand-gated ion channels that detect physical and chemical stimuli and promote painful sensations via nociceptor activation. TRP channels have physiological role in the mechanisms controlling several physiological responses like temperature and mechanical sensations, response to painful stimuli, taste, and pheromones. TRP channel family involves six different TRPs (TRPV1, TRPV2, TRPV3, TRPV4, TRPM8, and TRPA1) which are expressed in pain sensing neurons and primary afferent nociceptors. They function as transducers for mechanical, chemical, and thermal stimuli into inward currents, an essential first step for provoking pain sensations. TRP ion channels activated by temperature (thermo TRPs) are important molecular players in acute, inflammatory, and chronic pain states. Different degree of heat activates four TRP channels (TRPV1-4), while cold temperature ranging from affable to painful activate two indistinctly related thermo TRP channels (TRPM8 and TRPA1). Targeting primary afferent nociceptive neurons containing TRP channels that play pivotal role in revealing physical stimuli may be an effective target for the development of successful pharmacotherapeutics for clinical pain syndromes. In this review, we highlighted the potential role of various TRP channels in different types of neuropathic pain. We also discussed the pharmacological activity of naturally and synthetically originated TRP channel modulators for pharmacotherapeutics of nociception and neuropathic pain.
Collapse
|
23
|
Longhi-Balbinot DT, Rossaneis AC, Pinho-Ribeiro FA, Bertozzi MM, Cunha FQ, Alves-Filho JC, Cunha TM, Peron JPS, Miranda KM, Casagrande R, Verri WA. The nitroxyl donor, Angeli's salt, reduces chronic constriction injury-induced neuropathic pain. Chem Biol Interact 2016; 256:1-8. [PMID: 27287419 DOI: 10.1016/j.cbi.2016.06.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 05/27/2016] [Accepted: 06/06/2016] [Indexed: 12/31/2022]
Abstract
Chronic pain is a major health problem worldwide. We have recently demonstrated the analgesic effect of the nitroxyl donor, Angeli's salt (AS) in models of inflammatory pain. In the present study, the acute and chronic analgesic effects of AS was investigated in chronic constriction injury of the sciatic nerve (CCI)-induced neuropathic pain in mice. Acute (7th day after CCI) AS treatment (1 and 3 mg/kg; s.c.) reduced CCI-induced mechanical, but not thermal hyperalgesia. The acute analgesic effect of AS was prevented by treatment with 1H-[1,2, 4]oxadiazolo[4,3,-a]quinoxalin-1-one (ODQ, a soluble guanylate cyclase inhibitor), KT5823 (an inhibitor of protein kinase G [PKG]) or glibenclamide (GLB, an ATP-sensitive potassium channel blocker). Chronic (7-14 days after CCI) treatment with AS (3 mg/kg, s.c.) promoted a sustained reduction of CCI-induced mechanical and thermal hyperalgesia. Acute AS treatment reduced CCI-induced spinal cord allograft inflammatory factor 1 (known as Iba-1), interleukin-1β (IL-1β), and ST2 receptor mRNA expression. Chronic AS treatment reduced CCI-induced spinal cord glial fibrillary acidic protein (GFAP), Iba-1, IL-1β, tumor necrosis factor-α (TNF-α), interleukin-33 (IL-33) and ST2 mRNA expression. Chronic treatment with AS (3 mg/kg, s.c.) did not alter aspartate aminotransferase, alanine aminotransferase, urea or creatinine plasma levels. Together, these results suggest that the acute analgesic effect of AS depends on activating the cGMP/PKG/ATP-sensitive potassium channel signaling pathway. Moreover, chronic AS diminishes CCI-induced mechanical and thermal hyperalgesia by reducing the activation of spinal cord microglia and astrocytes, decreasing TNF-α, IL-1β and IL-33 cytokines expression. This spinal cord immune modulation was more prominent in the chronic treatment with AS. Thus, nitroxyl limits CCI-induced neuropathic pain by reducing spinal cord glial cells activation.
Collapse
Affiliation(s)
- Daniela T Longhi-Balbinot
- Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Rod. Celso Garcia Cid PR 445, Km 380, Cx. Postal 10.011, 86057-970, Londrina, Parana, Brazil
| | - Ana C Rossaneis
- Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Rod. Celso Garcia Cid PR 445, Km 380, Cx. Postal 10.011, 86057-970, Londrina, Parana, Brazil
| | - Felipe A Pinho-Ribeiro
- Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Rod. Celso Garcia Cid PR 445, Km 380, Cx. Postal 10.011, 86057-970, Londrina, Parana, Brazil
| | - Mariana M Bertozzi
- Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Rod. Celso Garcia Cid PR 445, Km 380, Cx. Postal 10.011, 86057-970, Londrina, Parana, Brazil
| | - Fernando Q Cunha
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes, 3900, Ribeirão Preto, 14049-900, São Paulo, Brazil
| | - José C Alves-Filho
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes, 3900, Ribeirão Preto, 14049-900, São Paulo, Brazil
| | - Thiago M Cunha
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes, 3900, Ribeirão Preto, 14049-900, São Paulo, Brazil
| | - Jean P S Peron
- Department of Immunology, Institute of Biomedical Sciences, Ed. Biomédicas IV, University of Sao Paulo, Av. Prof. Dr. Lineu Prestes, 1730, 05508-900, São Paulo, Brazil
| | - Katrina M Miranda
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ, USA
| | - Rubia Casagrande
- Department of Pharmaceutical Sciences, University Hospital (Health Science Centre), Av. Robert Koch, 60, 86038-350, State University of Londrina, Parana, Brazil
| | - Waldiceu A Verri
- Departamento de Ciências Patológicas, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Rod. Celso Garcia Cid PR 445, Km 380, Cx. Postal 10.011, 86057-970, Londrina, Parana, Brazil.
| |
Collapse
|
24
|
TNFα induces co-trafficking of TRPV1/TRPA1 in VAMP1-containing vesicles to the plasmalemma via Munc18-1/syntaxin1/SNAP-25 mediated fusion. Sci Rep 2016; 6:21226. [PMID: 26888187 PMCID: PMC4758037 DOI: 10.1038/srep21226] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 09/16/2015] [Indexed: 11/08/2022] Open
Abstract
Transient receptor potential (TRP) A1 and V1 channels relay sensory signals, yet little is known about their transport to the plasmalemma during inflammation. Herein, TRPA1 and TRPV1 were found on vesicles containing calcitonin gene-related peptide (CGRP), accumulated at sites of exo- and endo-cytosis, and co-localised on fibres and cell bodies of cultured sensory neurons expressing both. A proinflammatory cytokine, TNFα, elevated their surface content, and both resided in close proximity, indicating co-trafficking. Syntaxin 1-interacting protein, Munc18-1, proved necessary for the response to TNFα, and for TRPV1-triggered CGRP release. TNFα-induced surface trafficking of TRPV1 and TRPA1 required a synaptic vesicle membrane protein VAMP1 (but not 2/3), which is essential for CGRP exocytosis from large dense-core vesicles. Inactivation of two proteins on the presynaptic plasma membrane, syntaxin-1 or SNAP-25, by botulinum neurotoxin (BoNT)/C1 or /A inhibited the TNFα-elevated delivery. Accordingly, enhancement by TNFα of Ca(2+) influx through the upregulated surface-expressed TRPV1 and TRPA1 channels was abolished by BoNT/A. Thus, in addition, the neurotoxins' known inhibition of the release of pain transmitters, their therapeutic potential is augmented by lowering the exocytotic delivery of transducing channels and the resultant hyper-sensitisation in inflammation.
Collapse
|
25
|
Boadas-Vaello P, Castany S, Homs J, Álvarez-Pérez B, Deulofeu M, Verdú E. Neuroplasticity of ascending and descending pathways after somatosensory system injury: reviewing knowledge to identify neuropathic pain therapeutic targets. Spinal Cord 2016; 54:330-40. [DOI: 10.1038/sc.2015.225] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 11/25/2015] [Accepted: 11/28/2015] [Indexed: 12/16/2022]
|
26
|
Spicarova D, Nerandzic V, Palecek J. Update on the role of spinal cord TRPV1 receptors in pain modulation. Physiol Res 2014; 63:S225-36. [PMID: 24564662 DOI: 10.33549/physiolres.932713] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
The structure, expression and function of the transient receptor potential vanilloid 1 (TRPV1) receptor were intensively studied since the cloning in 1997 and TRPV1 receptors are now considered to act as transducers and molecular integrators of nociceptive stimuli in the periphery. In contrast, spinal TRPV1 receptors were studied less extensively and their role in pain modulation is still not fully understood. This short review is a follow up on our previous summary in this area (Spicarova and Palecek 2008). The aim was to review preferentially the most recent findings concerning the role of the spinal TRPV1 receptors, published within the last five years. The update is given on the expression and function of the spinal TRPV1 receptors, their activation by endogenous agonists, interaction between the endocannabinoid and endovanillod system and possible role of the spinal TRPV1 receptors in pathological pain states. There is now mounting evidence that TRPV1 receptors may be an important element in modulation of nociceptive information at the spinal cord level and represent an interesting target for analgesic therapy.
Collapse
Affiliation(s)
- D Spicarova
- Department of Functional Morphology, Institute of Physiology Academy of Sciences of the Czech Republic, Prague, Czech Republic.
| | | | | |
Collapse
|
27
|
König C, Zharsky M, Möller C, Schaible HG, Ebersberger A. Involvement of peripheral and spinal tumor necrosis factor α in spinal cord hyperexcitability during knee joint inflammation in rats. Arthritis Rheumatol 2014; 66:599-609. [PMID: 24574220 DOI: 10.1002/art.38271] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Accepted: 11/05/2013] [Indexed: 11/05/2022]
Abstract
OBJECTIVE Tumor necrosis factor α (TNFα) is produced not only in peripheral tissues, but also in the spinal cord. The purpose of this study was to address the potential of peripheral and spinal TNFα to induce and maintain spinal hyperexcitability, which is a hallmark of pain states in the joints during rheumatoid arthritis and osteoarthritis. METHODS In vivo recordings of the responses of spinal cord neurons to nociceptive knee input under normal conditions and in the presence of experimental knee joint inflammation were obtained in anesthetized rats. TNFα, etanercept, or antibodies to TNF receptors were applied to either the knee joint or the spinal cord surface. RESULTS Injection of TNFα into the knee joint cavity increased the responses of spinal cord neurons to mechanical joint stimulation, and injection of etanercept into the knee joint reduced the inflammation-evoked spinal activity. These spinal effects closely mirrored the induction and reduction of peripheral sensitization. Responses to joint stimulation were also enhanced by spinal application of TNFα, and spinal application of either etanercept or anti-TNF receptor type I significantly attenuated the generation of inflammation-evoked spinal hyperexcitability, which is characterized by widespread pain sensitization beyond the inflamed joint. Spinally applied etanercept did not reduce established hyperexcitability in the acute kaolin/carrageenan model. In antigen-induced arthritis, etanercept decreased spinal responses on day 1, but not on day 3. CONCLUSION While peripheral TNFα increases spinal responses to joint stimulation, spinal TNFα supports the generation of the full pattern of spinal hyperexcitability. However, established spinal hyperexcitability may be maintained by downstream mechanisms that are independent of spinal TNFα.
Collapse
Affiliation(s)
- Christian König
- Jena University Hospital and Friedrich Schiller University of Jena, Jena, Germany
| | | | | | | | | |
Collapse
|
28
|
Toxic and drug-induced peripheral neuropathies: updates on causes, mechanisms and management. Curr Opin Neurol 2014; 26:481-8. [PMID: 23995278 DOI: 10.1097/wco.0b013e328364eb07] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW This review discusses publications highlighting current research on toxic, chemotherapy-induced peripheral neuropathies (CIPNs), and drug-induced peripheral neuropathies (DIPNs). RECENT FINDINGS The emphasis in clinical studies is on the early detection and grading of peripheral neuropathies, whereas recent studies in animal models have given insights into molecular mechanisms, with the discovery of novel neuronal, axonal, and Schwann cell targets. Some substances trigger inflammatory changes in the peripheral nerves. Pharmacogenetic techniques are underway to identify genes that may help to predict individuals at higher risk of developing DIPNs. Several papers have been published on chemoprotectants; however, to date, this approach has not been shown effective in clinical trials. SUMMARY Both length and nonlength-dependent neuropathies are encountered, including small-fiber involvement. The introduction of new diagnostic techniques, such as excitability studies, skin laser Doppler flowmetry, and pharmacogenetics, holds promise for early detection and to elucidate underlying mechanisms. New approaches to improve functions and quality of life in CIPN patients are discussed. Apart from developing less neurotoxic anticancer therapies, there is still hope to identify chemoprotective agents (erythropoietin and substances involved in the endocannabinoid system are promising) able to prevent or correct painful CIPNs.
Collapse
|
29
|
Tamura R, Nemoto T, Maruta T, Onizuka S, Yanagita T, Wada A, Murakami M, Tsuneyoshi I. Up-regulation of NaV1.7 sodium channels expression by tumor necrosis factor-α in cultured bovine adrenal chromaffin cells and rat dorsal root ganglion neurons. Anesth Analg 2014; 118:318-324. [PMID: 24445633 DOI: 10.1213/ane.0000000000000085] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Tumor necrosis factor-α (TNF-α) is not only a key regulator of inflammatory response but also an important pain modulator. TNF-α enhances both tetrodotoxin-sensitive (TTX-S) and tetrodotoxin-resistant Na channel currents in dorsal root ganglion (DRG) neurons. However, it remains unknown whether TNF-α affects the function and expression of the TTX-S NaV1.7 Na channel, which plays crucial roles in pain generation. METHODS We used cultured bovine adrenal chromaffin cells expressing the NaV1.7 Na channel isoform and compared them with cultured rat DRG neurons. The expression of TNF receptor 1 and 2 (TNFR1 and TNFR2) in adrenal chromaffin cells was studied by Semiquantitative reverse transcription-polymerase chain reaction. The effects of TNF-α on the expression of NaV1.7 were examined with reverse transcription-polymerase chain reaction and Western blot analysis. Results were expressed as mean ± SEM. RESULTS TNFR1 and TNFR2 were expressed in adrenal chromaffin cells, as well as reported in DRG neurons. TNF-α up-regulated NaV1.7 mRNA by 132% ± 9% (N = 5, P = 0.004) in adrenal chromaffin cells, as well as 117% ± 2% (N = 5, P < 0.0001) in DRG neurons. Western blot analysis showed that TNF-α increased NaV1.7 protein up to 166% ± 24% (N = 5, corrected P < 0.0001) in adrenal chromaffin cells, concentration- and time-dependently. CONCLUSIONS TNF-α up-regulated NaV1.7 mRNA in both adrenal chromaffin cells and DRG neurons. In addition, TNF-α up-regulated the protein expression of the TTX-S NaV1.7 channel in adrenal chromaffin cells. Our findings may contribute to understanding the peripheral nociceptive mechanism of TNF-α.
Collapse
Affiliation(s)
- Ryuji Tamura
- From the Departments of Anesthesiology and Intensive Care and Pharmacology, Miyazaki Medical College, University of Miyazaki, Miyazaki, Japan; Department of Anesthesiology, Division of Basic Research, Washington University School of Medicine, St. Louis, Missouri; Department of Sports Health and Welfare, Faculty of Social Welfare, Kyusyu University of Health and Welfare, Miyazaki, Japan
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Spicarova D, Adamek P, Kalynovska N, Mrozkova P, Palecek J. TRPV1 receptor inhibition decreases CCL2-induced hyperalgesia. Neuropharmacology 2014; 81:75-84. [PMID: 24495396 DOI: 10.1016/j.neuropharm.2014.01.041] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2013] [Revised: 01/23/2014] [Accepted: 01/24/2014] [Indexed: 01/03/2023]
Abstract
Modulation of nociceptive synaptic transmission in the spinal cord is implicated in the development and maintenance of several pathological pain states. The chemokine CCL2 (C-C motif ligand 2) was shown to be an important factor in the development of neuropathic pain after peripheral nerve injury. In our experiments we have studied the effect of CCL2 application and TRPV1 (transient receptor potential vanilloid 1) receptor activation on nociceptive signaling and the modulation of synaptic transmission. Intrathecal drug application in behavioral experiments and patch-clamp recordings of spontaneous, miniature and dorsal root stimulation-evoked excitatory postsynaptic currents (sEPSCs, mEPSCs, eEPSCs) from superficial dorsal horn neurons in acute rat spinal cord slices were used. The intrathecal application of CCL2 induced thermal hyperalgesia and mechanical allodynia, while pretreatment with the TRPV1 receptor antagonist SB366791 diminished the thermal but not the mechanical hypersensitivity. Patch-clamp experiments showed an increase of sEPSC and mEPSC (124.5 ± 12.8% and 161.2 ± 17.3%, respectively) frequency in dorsal horn neurons after acute CCL2 application. This CCL2-induced increase was prevented by SB366791 pretreatment (89.4 ± 6.0%, 107.5 ± 14.2%). CCL2 application increased the amplitude of eEPSCs (188.1 ± 32.1%); this increase was significantly lower in experiments with SB366791 pretreatment (120.8 ± 17.2%). Our results demonstrate that the activation of spinal TRPV1 receptors plays an important role in the modulation of nociceptive signaling induced by CCL2 application. The mechanisms of cooperation between the CCL2 activated receptors and TRPV1 receptors on the central branches of primary afferent fibers may be especially important during different pathological pain states and need to be further investigated.
Collapse
Affiliation(s)
- Diana Spicarova
- Department of Functional Morphology, Institute of Physiology vvi, Academy of Sciences of the Czech Republic, Videnska 1083, 142 20 Prague 4, Czech Republic.
| | - Pavel Adamek
- Department of Functional Morphology, Institute of Physiology vvi, Academy of Sciences of the Czech Republic, Videnska 1083, 142 20 Prague 4, Czech Republic.
| | - Nataliia Kalynovska
- Department of Functional Morphology, Institute of Physiology vvi, Academy of Sciences of the Czech Republic, Videnska 1083, 142 20 Prague 4, Czech Republic.
| | - Petra Mrozkova
- Department of Functional Morphology, Institute of Physiology vvi, Academy of Sciences of the Czech Republic, Videnska 1083, 142 20 Prague 4, Czech Republic.
| | - Jiri Palecek
- Department of Functional Morphology, Institute of Physiology vvi, Academy of Sciences of the Czech Republic, Videnska 1083, 142 20 Prague 4, Czech Republic.
| |
Collapse
|
31
|
Yang CP, Cherng CH, Wu CT, Huang HY, Tao PL, Lee SO, Wong CS. Intrathecal ultra-low dose naloxone enhances the antihyperalgesic effects of morphine and attenuates tumor necrosis factor-α and tumor necrosis factor-α receptor 1 expression in the dorsal horn of rats with partial sciatic nerve transection. Anesth Analg 2014; 117:1493-502. [PMID: 24257399 DOI: 10.1213/ane.0000000000000020] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
BACKGROUND Glutamate homeostasis and microglia activation play an important role in the development and maintenance of neuropathic pain. We designed this investigation to examine whether ultra-low dose naloxone administered alone or in combination with morphine could alter the concentration of the excitatory amino acids (EAAs) glutamate and aspartate, as well as the expression of tumor necrosis factor-α (TNF-α) and its receptors (TNFR1 and TNFR2) in the spinal cord dorsal horn of rats with partial sciatic nerve transection (PST). METHODS Male Wistar rats underwent intrathecal catheter implantation for drug delivery and were divided in 7 groups: sham-operated + saline (sham), PST + saline (S), PST + 15 ng naloxone (n), PST + 15 µg naloxone (N), PST + 10 µg morphine (M), PST + 15 ng naloxone + 10 µg morphine (Mn), PST + 15 µg naloxone + 10 µg morphine (MN). Thermal withdrawal latency and mechanical withdrawal threshold, TNF-α and TNFR expression in the spinal cord and dorsal root ganglia, and EAAs glutamate and aspartate concentration in cerebrospinal fluid dialysates were measured. RESULTS Ten days after PST, rats developed hyperalgesia (P < 0.0001) and allodynia (P < 0.0001), and increased TNF-α (P < 0.0001) and TNFR1 expression (P = 0.0009) were measured in the ipsilateral spinal cord dorsal horn. The antihyperalgesic and antiallodynic effects of morphine (10 μg) were abolished by high-dose naloxone (15 μg; P = 0.0031) but enhanced by ultra-low dose naloxone (15 ng; P = 0.0015), and this was associated with a reduction of TNF-α (P < 0.0001) and TNFR1 (P = 0.0009) expression in the spinal cord dorsal horn and EAAs concentration (glutamate: P = 0.0001; aspartate: P = 0.004) in cerebrospinal fluid dialysate. Analysis of variance (ANOVA) or Student t test with Bonferroni correction were used for statistical analysis. CONCLUSIONS Ultra-low dose naloxone enhances the antihyperalgesia and antiallodynia effects of morphine in PST rats, possibly by reducing TNF-α and TNFR1 expression, and EAAs concentrations in the spinal dorsal horn. Ultra-low dose naloxone may be a useful adjuvant for increasing the analgesic effect of morphine in neuropathic pain conditions.
Collapse
Affiliation(s)
- Chih-Ping Yang
- From the *Division of Anesthesiology, Armed Forces Taoyuan General Hospital, Taoyuan; †Tri-Service General Hospital, ‡Department of Anesthesiology, and §Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei; ‖Division of Mental Health and Addiction Medicine, Institute of Population Health Sciences, National Health Research Institutes, Miaoli County; and ¶Department of Anesthesiology, Cathy General Hospital, Taipei, Taiwan
| | | | | | | | | | | | | |
Collapse
|
32
|
Luo J, Walters ET, Carlton SM, Hu H. Targeting Pain-evoking Transient Receptor Potential Channels for the Treatment of Pain. Curr Neuropharmacol 2014; 11:652-63. [PMID: 24396340 PMCID: PMC3849790 DOI: 10.2174/1570159x113119990040] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2013] [Revised: 06/19/2013] [Accepted: 06/19/2013] [Indexed: 02/06/2023] Open
Abstract
Chronic pain affects billions of lives globally and is a major public health problem in the United States. However, pain management is still a challenging task due to a lack of understanding of the fundamental mechanisms of pain. In the past decades transient receptor potential (TRP) channels have been identified as molecular sensors of tissue damage and inflammation. Activation/sensitization of TRP channels in peripheral nociceptors produces neurogenic inflammation and contributes to both somatic and visceral pain. Pharmacological and genetic studies have affirmed the role of TRP channels in multiple forms of inflammatory and neuropathic pain. Thus pain-evoking TRP channels emerge as promising therapeutic targets for a wide variety of pain and inflammatory conditions.
Collapse
Affiliation(s)
- Jialie Luo
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX 77030
| | - Edgar T Walters
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX 77030
| | - Susan M Carlton
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-1069
| | - Hongzhen Hu
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX 77030
| |
Collapse
|
33
|
Yang Y, Wu H, Yan JQ, Song ZB, Guo QL. Tumor necrosis factor-α inhibits angiotensin II receptor type 1 expression in dorsal root ganglion neurons via β-catenin signaling. Neuroscience 2013; 248:383-91. [DOI: 10.1016/j.neuroscience.2013.06.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Revised: 05/20/2013] [Accepted: 06/13/2013] [Indexed: 01/21/2023]
|
34
|
Li Y, Zhang Y, Liu DB, Liu HY, Hou WG, Dong YS. Curcumin attenuates diabetic neuropathic pain by downregulating TNF-α in a rat model. Int J Med Sci 2013; 10:377-81. [PMID: 23471081 PMCID: PMC3590595 DOI: 10.7150/ijms.5224] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2012] [Accepted: 02/10/2013] [Indexed: 12/22/2022] Open
Abstract
The mechanisms involved in diabetic neuropathic pain are complex and involve peripheral and central pathophysiological phenomena. Proinflammatory tumour necrosis factor α (TNF-α) and TNF-α receptor 1, which are markers of inflammation, contribute to neuropathic pain. The purpose of this experimental study was to evaluate the effect of curcumin on diabetic pain in rats. We tested 24 rats with diabetes induced by a single intraperitoneal injection of streptozotocin and 24 healthy control rats. Twelve rats in each group received 60 mg/kg oral curcumin daily for 28 days, and the other 12 received vehicle. On days 7, 14, 21, and 28, we tested mechanical allodynia with von Frey hairs and thermal hyperalgesia with radiant heat. Markers of inflammation in the spinal cord dorsal horn on day 28 were estimated with a commercial assay and Western blot analysis. Compared to control rats, diabetic rats exhibited increased mean plasma glucose concentration, decreased mean body weight, and significant pain hypersensitivity, as evidenced by decreased paw withdrawal threshold to von Frey hairs and decreased paw withdrawal latency to heat. Curcumin significantly attenuated the diabetes-induced allodynia and hyperalgesia and reduced the expression of both TNF-α and TNF-α receptor 1. Curcumin seems to relieve diabetic hyperalgesia, possibly through an inhibitory action on TNF-α and TNF-α receptor 1.
Collapse
Affiliation(s)
- Yue Li
- Department of air logistics, the 463 Hospital of PLA, Shenyang 110042, PR China
| | | | | | | | | | | |
Collapse
|
35
|
Santello M, Volterra A. TNFα in synaptic function: switching gears. Trends Neurosci 2012; 35:638-47. [DOI: 10.1016/j.tins.2012.06.001] [Citation(s) in RCA: 194] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Revised: 05/18/2012] [Accepted: 06/04/2012] [Indexed: 01/17/2023]
|
36
|
Chiang CY, Sessle BJ, Dostrovsky JO. Role of Astrocytes in Pain. Neurochem Res 2012; 37:2419-31. [DOI: 10.1007/s11064-012-0801-6] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Revised: 04/18/2012] [Accepted: 05/07/2012] [Indexed: 12/18/2022]
|