1
|
Ke X, Cai H, Luo F, Zheng X, Hu Q, Zhou Y, Wang Y, Zhang X, Chen Y, Chen G. TRPC4 Mediates Trigeminal Neuropathic Pain via Ca 2+-ERK/P38-ATF2 Pathway in the Trigeminal Ganglion of Mice. CNS Neurosci Ther 2025; 31:e70368. [PMID: 40202077 PMCID: PMC11979714 DOI: 10.1111/cns.70368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 02/19/2025] [Accepted: 03/18/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Trigeminal neuropathic pain (TNP) is a debilitating condition characterized by chronic facial pain, yet its underlying mechanisms remain incompletely understood. Transient Receptor Potential Canonical 4 (TRPC4) has been reported to promote the development of abnormal pain or pain hypersensitivity in neuropathic pain. However, the specific contribution of TRPC4 to TNP pathogenesis remains unclear. AIM This study aimed to investigate the role of TRPC4 in a mouse model of trigeminal neuropathic pain induced by chronic constriction of the unilateral infraorbital nerve (CION). METHODS Adult male/female mice were subjected to either CION surgery or sham surgery. Behavioral assays were conducted to assess facial pain-like responses over a 28-day period. TRPC4 distribution in the trigeminal ganglion (TG) was evaluated using Immunofluorescence. TRPC4 inhibitor ML204 and agonist Englerin A were employed to evaluate the impact of TRPC4 on facial pain-like behaviors. A TRPC4-overexpressing HEK293 cell model was conducted via plasmid transfection. To assess the function of TRPC4, we employed cellular calcium imaging technology to investigate the effects of modulating TRPC4 function by analyzing dynamic changes in intracellular calcium ion concentrations in primary trigeminal ganglion neurons and HEK293 cells. Trpc4 shRNA was used to specifically knock down TRPC4 in the trigeminal ganglion. Western blot analysis was used to assess the activation of ERK, P38, and ATF2 signaling pathways. RESULTS Mice subjected to CION exhibited persistent facial pain-like behaviors and a significant increase in TRPC4 expression in TG neurons. Trpc4 shRNA or pharmacological inhibition with ML204 attenuated CION-induced pain behaviors, while activation of TRPC4 with Englerin A induced pain-like responses in naive mice. Calcium imaging revealed that both Englerin A and TRPC4 overexpression elevated intracellular Ca²2+ levels in TG neurons and HEK293 cells. This Ca²2+ influx triggered the activation of ERK and P38, leading to enhanced ATF2 activation. Downregulation of TRPC4 in the TG reduced ERK/P38 phosphorylation and ATF2 expression and activation. CONCLUSION This study provides the first evidence that TRPC4 plays a critical role in CION-induced trigeminal neuropathic pain by promoting the activation of the downstream transcription factor ATF2 via the Ca²2+-ERK/P38 pathway.
Collapse
Affiliation(s)
- Xinlong Ke
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of MedicineZhejiang UniversityZhejiangHangzhouChina
| | - Huajing Cai
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of MedicineZhejiang UniversityZhejiangHangzhouChina
| | - Fangla Luo
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of MedicineZhejiang UniversityZhejiangHangzhouChina
| | - Xing Zheng
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of MedicineZhejiang UniversityZhejiangHangzhouChina
| | - Qian Hu
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of MedicineZhejiang UniversityZhejiangHangzhouChina
| | - Youfa Zhou
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of MedicineZhejiang UniversityZhejiangHangzhouChina
| | - Yongjie Wang
- School of PharmacyHangzhou Normal UniversityZhejiangHangzhouChina
| | - Xiangnan Zhang
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Key Laboratory of Medical Neurobiology of the Ministry of Health of ChinaZhejiang UniversityZhejiangHangzhouChina
| | - Yeru Chen
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of MedicineZhejiang UniversityZhejiangHangzhouChina
- Provincial Key Laboratory of Precise Diagnosis and Treatment of Abdominal Infection, Sir Run Run Shaw Hospital, School of MedicineZhejiang UniversityZhejiangHangzhouChina
| | - Gang Chen
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of MedicineZhejiang UniversityZhejiangHangzhouChina
- Provincial Key Laboratory of Precise Diagnosis and Treatment of Abdominal Infection, Sir Run Run Shaw Hospital, School of MedicineZhejiang UniversityZhejiangHangzhouChina
| |
Collapse
|
2
|
Southey BR, Sunderland GR, Gomez AN, Bhamidi S, Rodriguez-Zas SL. Incidence of alternative splicing associated with sex and opioid effects in the axon guidance pathway. Gene 2025; 942:149215. [PMID: 39756548 PMCID: PMC11863264 DOI: 10.1016/j.gene.2025.149215] [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: 09/12/2024] [Revised: 12/19/2024] [Accepted: 01/02/2025] [Indexed: 01/07/2025]
Abstract
The alternative splicing of a gene results in distinct transcript isoforms that can result in proteins that differ in function. Alternative splicing processes are prevalent in the brain, have varying incidence across brain regions, and can present sexual dimorphism. Exposure to opiates and other substances of abuse can also alter the type and incidence of the splicing process and the relative abundance of the isoforms produced. The disruption of alternative splicing patterns associated with sex differences and morphine exposure in the prefrontal cortex of a pig model was studied. The numbers of genes presenting one or more significant (FDR-adjusted p-value < 0.05) alternative splicing events were 933 and 1,368 genes when comparing females relative to males and morphine- relative to saline-treated animals, respectively. The sex-dependent opioid effect was most extreme in the contrast between morphine- versus saline-treated males with 1,934 significantly differentially spliced genes. The most frequent and significant alternative splicing type was skipped exon (∼56 % event), followed by retained intron (∼15 % events). The pathways encompassing a significant number of differentially spliced genes included axon guidance, glutamatergic synapses, circadian rhythm, and lysine degradation. Genes in these pathways included ROBO1, SEMA6C, GRIN3A, GRM2, ARNTL, CLOCK, HYKK, and DOT1L. Transcription factors ETV7 and DMAP1 presented a significant number of differentially spliced target genes. The distribution of the genes presenting differential alternative splicing in the axon guidance and circadian rhythm pathways indicates that this regulatory mechanism impacts hubs and peripheral genes. The identification of sexual dimorphism in the effect of morphine across multiple pathways confirms the necessity to explore the effects of drugs of abuse within sex. Altogether, our findings advance the understanding of the response to factors that can impact the activity of excitatory synapses by modulating transcriptional mechanisms that support the plasticity of the prefrontal cortex.
Collapse
Affiliation(s)
- Bruce R Southey
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801 USA
| | - Gloria R Sunderland
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801 USA
| | - Andrea N Gomez
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801 USA
| | - Sreelaya Bhamidi
- Informatics Program, University of Illinois at Urbana-Champaign, Urbana, IL 61820 USA
| | - Sandra L Rodriguez-Zas
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801 USA; Informatics Program, University of Illinois at Urbana-Champaign, Urbana, IL 61820 USA; Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL 61801 USA; Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801 USA; Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801 USA; Department of Statistics, University of Illinois at Urbana-Champaign, Urbana, IL 61820 USA.
| |
Collapse
|
3
|
Silva ÁJC, de Lavor MSL. Nitroxidative Stress, Cell-Signaling Pathways, and Manganese Porphyrins: Therapeutic Potential in Neuropathic Pain. Int J Mol Sci 2025; 26:2050. [PMID: 40076672 PMCID: PMC11900433 DOI: 10.3390/ijms26052050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 02/23/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
Neuropathic pain, a debilitating condition arising from somatosensory system damage, significantly impacts quality of life, leading to anxiety, self-mutilation, and depression. Oxidative and nitrosative stress, an imbalance between reactive oxygen and nitrogen species (ROS/RNS) and antioxidant defenses, plays a crucial role in its pathophysiology. While reactive species are essential for physiological functions, excessive levels can cause cellular component damage, leading to neuronal dysfunction and pain. This review highlights the complex interactions between reactive species, antioxidant systems, cell signaling, and neuropathic pain. We discuss the physiological roles of ROS/RNS and the detrimental effects of oxidative and nitrosative stress. Furthermore, we explore the potential of manganese porphyrins, compounds with antioxidant properties, as promising therapeutic agents to mitigate oxidative stress and alleviate neuropathic pain by targeting key cellular pathways involved in pain. Further research is needed to fully understand their therapeutic potential in managing neuropathic pain in human and non-human animals.
Collapse
Affiliation(s)
| | - Mário Sérgio Lima de Lavor
- Department of Agricultural and Environmental Sciences, State University of Santa Cruz (UESC), Ilhéus 45662-900, BA, Brazil;
| |
Collapse
|
4
|
Ke X, Cai H, Chen Y, Chen G. Exploring the therapeutic potential of TRPC channels in chronic pain: An investigation into their mechanisms, functions, and prospects. Eur J Pharmacol 2025; 987:177206. [PMID: 39672226 DOI: 10.1016/j.ejphar.2024.177206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 12/10/2024] [Accepted: 12/11/2024] [Indexed: 12/15/2024]
Abstract
Transient Receptor Potential Canonical (TRPC) channels have received more attention in recent years for their role of in the pathophysiology of chronic pain. These non-selective cation channels, which are predominantly present on cell membranes, play a pivotal role in regulating both physiological and pathological processes. Research advances have shown the critical role of TRPC channels in a variety of chronic pain, including neuropathic, inflammatory, and visceral pain. Activation of TRPC channels increases neuronal excitability, amplifying and prolonging pain signals. Moreover, these channels collaborate with other ion channels and receptors to form complexes that augment the transmission and perception of pain. As research advances, our understanding of TRPC channels' regulation mechanisms and signaling pathways improves. An expanding variety of TRPC modulators has been identified as promising therapeutic agents for chronic pain, opening up novel treatment options. Nevertheless, the diversity and complexity of TRPC channels present challenges in drug development, highlighting the importance of full understanding of their unique properties and activities. This review aims to provide a thorough evaluation of recent breakthrough in TRPC channels research related to chronic pain, with a focus on their mechanisms, functions, and prospective therapeutic application. By integrating existing research findings, we seek to bring new viewpoints and approaches for chronic pain management.
Collapse
Affiliation(s)
- Xinlong Ke
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Huajing Cai
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Yeru Chen
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China.
| | - Gang Chen
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China.
| |
Collapse
|
5
|
Khare P, Chand J, Ptakova A, Liguori R, Ferrazzi F, Bishnoi M, Vlachova V, Zimmermann K. The TRPC5 receptor as pharmacological target for pain and metabolic disease. Pharmacol Ther 2024; 263:108727. [PMID: 39384022 DOI: 10.1016/j.pharmthera.2024.108727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 09/11/2024] [Accepted: 10/03/2024] [Indexed: 10/11/2024]
Abstract
The transient receptor potential canonical (TRPC) channels are a group of highly homologous nonselective cation channels from the larger TRP channel family. They have the ability to form homo- and heteromers with varying degrees of calcium (Ca2+) permeability and signalling properties. TRPC5 is the one cold-sensitive among them and likewise facilitates the influx of extracellular Ca2+ into cells to modulate neuronal depolarization and integrate various intracellular signalling pathways. Recent research with cryo-electron microscopy revealed its structure, along with clear insight into downstream signalling and protein-protein interaction sites. Investigations using global and conditional deficient mice revealed the involvement of TRPC5 in metabolic diseases, energy balance, thermosensation and conditions such as osteoarthritis, rheumatoid arthritis, and inflammatory pain including opioid-induced hyperalgesia and hyperalgesia following tooth decay and pulpitis. This review provides an update on recent advances in our understanding of the role of TRPC5 with focus on metabolic diseases and pain.
Collapse
Affiliation(s)
- Pragyanshu Khare
- Department of Anesthesiology, Friedrich Alexander Universität Erlangen-Nürnberg, Erlangen, Germany; Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan 333031, India
| | - Jagdish Chand
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan 333031, India
| | - Alexandra Ptakova
- Department of Cellular Neurophysiology, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Renato Liguori
- Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Fulvia Ferrazzi
- Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Mahendra Bishnoi
- TR(i)P for Health Laboratory Centre for Excellence in Functional Foods, Food & Nutrition Biotechnology Division, National Agri-Food Biotechnology Institute, S.A.S Nagar, Sector (Knowledge City), Punjab, India
| | - Viktorie Vlachova
- Department of Cellular Neurophysiology, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Katharina Zimmermann
- Department of Anesthesiology, Friedrich Alexander Universität Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
6
|
Sun Z, Han W, Dou Z, Lu N, Wang X, Wang F, Ma S, Tian Z, Xian H, Liu W, Liu Y, Wu W, Chu W, Guo H, Wang F, Ding H, Liu Y, Tao H, Freichel M, Birnbaumer L, Li Z, Xie R, Wu S, Luo C. TRPC3/6 Channels Mediate Mechanical Pain Hypersensitivity via Enhancement of Nociceptor Excitability and of Spinal Synaptic Transmission. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404342. [PMID: 39340833 PMCID: PMC11600220 DOI: 10.1002/advs.202404342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 09/11/2024] [Indexed: 09/30/2024]
Abstract
Patients with tissue inflammation or injury often experience aberrant mechanical pain hypersensitivity, one of leading symptoms in clinic. Despite this, the molecular mechanisms underlying mechanical distortion are poorly understood. Canonical transient receptor potential (TRPC) channels confer sensitivity to mechanical stimulation. TRPC3 and TRPC6 proteins, coassembling as heterotetrameric channels, are highly expressed in sensory neurons. However, how these channels mediate mechanical pain hypersensitivity has remained elusive. It is shown that in mice and human, TRPC3 and TRPC6 are upregulated in DRG and spinal dorsal horn under pathological states. Double knockout of TRPC3/6 blunts mechanical pain hypersensitivity, largely by decreasing nociceptor hyperexcitability and spinal synaptic potentiation via presynaptic mechanism. In corroboration with this, nociceptor-specific ablation of TRPC3/6 produces comparable pain relief. Mechanistic analysis reveals that upon peripheral inflammation, TRPC3/6 in primary sensory neurons get recruited via released bradykinin acting on B1/B2 receptors, facilitating BDNF secretion from spinal nociceptor terminals, which in turn potentiates synaptic transmission through TRPC3/6 and eventually results in mechanical pain hypersensitivity. Antagonizing TRPC3/6 in DRG relieves mechanical pain hypersensitivity in mice and nociceptor hyperexcitability in human. Thus, TRPC3/6 in nociceptors is crucially involved in pain plasticity and constitutes a promising therapeutic target against mechanical pain hypersensitivity with minor side effects.
Collapse
Affiliation(s)
- Zhi‐Chuan Sun
- Department of NeurobiologySchool of Basic MedicineFourth Military Medical UniversityXi'an710032China
- Department of NeurosurgeryXi'an Daxing HospitalXi'an710016China
| | - Wen‐Juan Han
- Department of NeurobiologySchool of Basic MedicineFourth Military Medical UniversityXi'an710032China
| | - Zhi‐Wei Dou
- Department of NeurobiologySchool of Basic MedicineFourth Military Medical UniversityXi'an710032China
| | - Na Lu
- Department of NeurobiologySchool of Basic MedicineFourth Military Medical UniversityXi'an710032China
- The Assisted Reproduction CenterNorthwest Women and Children's HospitalXi'an710000China
| | - Xu Wang
- Department of NeurobiologySchool of Basic MedicineFourth Military Medical UniversityXi'an710032China
| | - Fu‐Dong Wang
- Department of NeurobiologySchool of Basic MedicineFourth Military Medical UniversityXi'an710032China
| | - Sui‐Bin Ma
- Department of NeurobiologySchool of Basic MedicineFourth Military Medical UniversityXi'an710032China
| | - Zhi‐Cheng Tian
- Department of NeurobiologySchool of Basic MedicineFourth Military Medical UniversityXi'an710032China
| | - Hang Xian
- Department of NeurobiologySchool of Basic MedicineFourth Military Medical UniversityXi'an710032China
- Department of OrthopedicsXijing HospitalFourth Military Medical UniversityXi'an710032China
| | - Wan‐Neng Liu
- Department of NeurobiologySchool of Basic MedicineFourth Military Medical UniversityXi'an710032China
| | - Ying‐Ying Liu
- Department of NeurobiologySchool of Basic MedicineFourth Military Medical UniversityXi'an710032China
| | - Wen‐Bin Wu
- Department of NeurobiologySchool of Basic MedicineFourth Military Medical UniversityXi'an710032China
| | - Wen‐Guang Chu
- Department of NeurobiologySchool of Basic MedicineFourth Military Medical UniversityXi'an710032China
| | - Huan Guo
- Department of NeurobiologySchool of Basic MedicineFourth Military Medical UniversityXi'an710032China
| | - Fei Wang
- Department of NeurobiologySchool of Basic MedicineFourth Military Medical UniversityXi'an710032China
| | - Hui Ding
- Department of NeurobiologySchool of Basic MedicineFourth Military Medical UniversityXi'an710032China
| | - Yuan‐Ying Liu
- Department of NeurobiologySchool of Basic MedicineFourth Military Medical UniversityXi'an710032China
| | - Hui‐Ren Tao
- Department of Orthopedic SurgeryThe University of Hong Kong‐Shenzhen HospitalShenzhenGuangdong518053China
| | - Marc Freichel
- Institute of PharmacologyHeidelberg University69120HeidelbergGermany
| | - Lutz Birnbaumer
- Institute of Biomedical Research (BIOMED)Catholic University of ArgentinaBuenos AiresC1107AVVArgentina
- Signal Transduction LaboratoryNational institute of Environmental Health SciencesResearch Triangle ParkNC27709United States
| | - Zhen‐Zhen Li
- Department of NeurobiologySchool of Basic MedicineFourth Military Medical UniversityXi'an710032China
| | - Rou‐Gang Xie
- Department of NeurobiologySchool of Basic MedicineFourth Military Medical UniversityXi'an710032China
| | - Sheng‐Xi Wu
- Department of NeurobiologySchool of Basic MedicineFourth Military Medical UniversityXi'an710032China
| | - Ceng Luo
- Department of NeurobiologySchool of Basic MedicineFourth Military Medical UniversityXi'an710032China
- Innovation Research InstituteXijing HospitalFourth Military Medical UniversityXi'an710032China
| |
Collapse
|
7
|
Kang H, So I. Unique responses of the fixed stoichiometric TRPC1-TRPC5 concatemer to G proteins. Front Physiol 2024; 15:1392980. [PMID: 39397856 PMCID: PMC11466768 DOI: 10.3389/fphys.2024.1392980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 05/17/2024] [Indexed: 10/15/2024] Open
Abstract
Transient receptor potential canonical (TRPC)5 channel is a non-selective cation channel that plays a significant role in membrane depolarization and calcium influx. TRPC5 not only forms homotetramers itself but also heterotetramers with TRPC1. However, accurately testing and confirming these heterotetrameric channels at specific ratios has proven challenging. Therefore, creating heteromeric concatemers of TRPC5 and TRPC1 with a fixed stoichiometry of 1:1 becomes essential. This study aims to meticulously identify and reaffirm the properties of TRPC5 homomers and heteromers with a 1:1 fixed stoichiometry to determine the optimal ratio for the TRPC1/5 heterotetramer. The overall characteristics were consistent with those of the previous studies, but several specific features were different. The TRPC1-TRPC5 concatemer is activated by Englerin A and GiQL, whereas carbachol alone does not trigger its activation. Additionally, GqQL significantly inhibited the current when co-expressed with the concatemer. Interestingly, carbachol can activate the TRPC1-TRPC5 concatemer in the presence of internal GTPγS, highlighting the influence of intracellular signaling conditions on its activation. Meanwhile, the TRPC5-TRPC5 concatemer is responsive to both carbachol and Englerin A. In conclusion, we provide evidence that the TRPC1-TRPC5 heteromeric concatemer with fixed stoichiometry need specific conditions to respond to carbachol, whereas the TRPC5-TRPC5 homomeric concatemer responds physiologically to carbachol. Additional research may be necessary to ascertain the optimal stoichiometry for the TRPC1-TRPC5 concatemer to enhance its electrophysiological properties.
Collapse
Affiliation(s)
- Hana Kang
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Insuk So
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
- Institute of Human-Environment Interface Biology, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
8
|
Yao C, Fang X, Ru Q, Li W, Li J, Mehsein Z, Tolias KF, Li L. Tiam1-mediated maladaptive plasticity underlying morphine tolerance and hyperalgesia. Brain 2024; 147:2507-2521. [PMID: 38577773 PMCID: PMC11224607 DOI: 10.1093/brain/awae106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 03/16/2024] [Accepted: 03/21/2024] [Indexed: 04/06/2024] Open
Abstract
Opioid pain medications, such as morphine, remain the mainstay for treating severe and chronic pain. Prolonged morphine use, however, triggers analgesic tolerance and hyperalgesia (OIH), which can last for a long period after morphine withdrawal. How morphine induces these detrimental side effects remains unclear. Here, we show that morphine tolerance and OIH are mediated by Tiam1-coordinated synaptic structural and functional plasticity in the spinal nociceptive network. Tiam1 is a Rac1 GTPase guanine nucleotide exchange factor that promotes excitatory synaptogenesis by modulating actin cytoskeletal dynamics. We found that prolonged morphine treatment activated Tiam1 in the spinal dorsal horn and Tiam1 ablation from spinal neurons eliminated morphine antinociceptive tolerance and OIH. At the same time, the pharmacological blockade of Tiam1-Rac1 signalling prevented the development and reserved the established tolerance and OIH. Prolonged morphine treatment increased dendritic spine density and synaptic NMDA receptor activity in spinal dorsal horn neurons, both of which required Tiam1. Furthermore, co-administration of the Tiam1 signalling inhibitor NSC23766 was sufficient to abrogate morphine tolerance in chronic pain management. These findings identify Tiam1-mediated maladaptive plasticity in the spinal nociceptive network as an underlying cause for the development and maintenance of morphine tolerance and OIH and provide a promising therapeutic target to reduce tolerance and prolong morphine use in chronic pain management.
Collapse
Affiliation(s)
- Changqun Yao
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL 35025, USA
| | - Xing Fang
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Qin Ru
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Health and Kinesiology, School of Physical Education, Jianghan University, Wuhan 430056, China
| | - Wei Li
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35025, USA
| | - Jun Li
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL 35025, USA
| | - Zeinab Mehsein
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL 35025, USA
| | - Kimberley F Tolias
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Lingyong Li
- Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, AL 35025, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
9
|
Cao Y, Jiang W, Yan F, Pan Y, Gei L, Lu S, Chen X, Huang Y, Yan Y, Feng Y, Li Q, Zeng W, Xing W, Chen D. Sex differences in PD-L1-induced analgesia in paclitaxel-induced peripheral neuropathy mice depend on TRPV1-based inhibition of CGRP. CNS Neurosci Ther 2024; 30:e14829. [PMID: 38961264 PMCID: PMC11222069 DOI: 10.1111/cns.14829] [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: 02/02/2024] [Revised: 06/13/2024] [Accepted: 06/16/2024] [Indexed: 07/05/2024] Open
Abstract
AIMS Paclitaxel (PTX) is extensively utilized in the management of diverse solid tumors, frequently resulting in paclitaxel-induced peripheral neuropathy (PIPN). The present study aimed to investigate sex differences in the behavioral manifestations and underlying pathogenesis of PIPN and search for clinically efficacious interventions. METHODS Male and female C57BL/6 mice (5-6 weeks and 12 months, weighing 18-30 g) were intraperitoneally (i.p.) administered paclitaxel diluted in saline (NaCl 0.9%) at a dose of 2 mg/kg every other day for a total of 4 injections. Von Frey and hot plate tests were performed before and after administration to confirm the successful establishment of the PIPN model and also to evaluate the pain of PIPN and the analgesic effect of PD-L1. On day 14 after PTX administration, PD-L1 protein (10 ng/pc) was injected into the PIPN via the intrathecal (i.t.) route. To knock down TRPV1 in the spinal cord, adeno-associated virus 9 (AAV9)-Trpv1-RNAi (5 μL, 1 × 1013 vg/mL) was slowly injected via the i.t. route. Four weeks after AAV9 delivery, the downregulation of TRPV1 expression was verified by immunofluorescence staining and Western blotting. The levels of PD-L1, TRPV1 and CGRP were measured via Western blotting, RT-PCR, and immunofluorescence staining. The levels of TNF-α and IL-1β were measured via RT-PCR. RESULTS TRPV1 and CGRP protein and mRNA levels were higher in the spinal cords of control female mice than in those of control male mice. PTX-induced nociceptive behaviors in female PIPN mice were greater than those in male PIPN mice, as indicated by increased expression of TRPV1 and CGRP. The analgesic effects of PD-L1 on mechanical hyperalgesia and thermal sensitivity were significantly greater in female mice than in male mice, with calculated relative therapeutic levels increasing by approximately 2.717-fold and 2.303-fold, respectively. PD-L1 and CGRP were partly co-localized with TRPV1 in the dorsal horn of the mouse spinal cord. The analgesic effect of PD-L1 in PIPN mice was observed to be mediated through the downregulation of TRPV1 and CGRP expression following AAV9-mediated spinal cord specific decreased TRPV1 expression. CONCLUSIONS PTX-induced nociceptive behaviors and the analgesic effect of PD-L1 in PIPN mice were sexually dimorphic, highlighting the significance of incorporating sex as a crucial biological factor in forthcoming mechanistic studies of PIPN and providing insights for potential sex-specific therapeutic approaches.
Collapse
Affiliation(s)
- Yan Cao
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouChina
| | - Wenqi Jiang
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouChina
| | - Fang Yan
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouChina
| | - Yuyan Pan
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouChina
| | - Liba Gei
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouChina
- Department of AnesthesiologyPeking University Cancer Hospital (Inner Mongolia Campus)/Affiliated Cancer Hospital of Inner Mongolia Medical University/Inner Mongolia Autonomous Region Cancer HospitalHohhotChina
| | - Simin Lu
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouChina
| | - Xiangnan Chen
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouChina
- Department of AnesthesiologyGuangdong Women and Children HospitalGuangzhouChina
| | - Yang Huang
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouChina
| | - Yan Yan
- Department of AnesthesiologyHuizhou Municipal Central HospitalHuizhouChina
| | - Yan Feng
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouChina
| | - Qiang Li
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouChina
| | - Weian Zeng
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouChina
| | - Wei Xing
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouChina
| | - Dongtai Chen
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for CancerSun Yat‐Sen University Cancer CenterGuangzhouChina
| |
Collapse
|
10
|
Xu S, Wang Y. Transient Receptor Potential Channels: Multiple Modulators of Peripheral Neuropathic Pain in Several Rodent Models. Neurochem Res 2024; 49:872-886. [PMID: 38281247 DOI: 10.1007/s11064-023-04087-4] [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: 10/14/2023] [Revised: 11/22/2023] [Accepted: 12/16/2023] [Indexed: 01/30/2024]
Abstract
Neuropathic pain, a prevalent chronic condition in clinical settings, has attracted widespread societal attention. This condition is characterized by a persistent pain state accompanied by affective and cognitive disruptions, significantly impacting patients' quality of life. However, current clinical therapies fall short of addressing its complexity. Thus, exploring the underlying molecular mechanism of neuropathic pain and identifying potential targets for intervention is highly warranted. The transient receptor potential (TRP) receptors, a class of widely distributed channel proteins, in the nervous system, play a crucial role in sensory signaling, cellular calcium regulation, and developmental influences. TRP ion channels are also responsible for various sensory responses including heat, cold, pain, and stress. This review highlights recent advances in understanding TRPs in various rodent models of neuropathic pain, aiming to uncover potential therapeutic targets for clinical management.
Collapse
Affiliation(s)
- Songchao Xu
- Department of Anesthesiology, Beijing Friendship Hospital, Capital Medical University, No. 95, Yong'an Road, Xicheng District, Beijing, 100050, China
| | - Yun Wang
- Department of Anesthesiology, Beijing Friendship Hospital, Capital Medical University, No. 95, Yong'an Road, Xicheng District, Beijing, 100050, China.
| |
Collapse
|
11
|
Kang H, Kim J, Park CH, Jeong B, So I. Direct modulation of TRPC ion channels by Gα proteins. Front Physiol 2024; 15:1362987. [PMID: 38384797 PMCID: PMC10880550 DOI: 10.3389/fphys.2024.1362987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 01/26/2024] [Indexed: 02/23/2024] Open
Abstract
GPCR-Gi protein pathways are involved in the regulation of vagus muscarinic pathway under physiological conditions and are closely associated with the regulation of internal visceral organs. The muscarinic receptor-operated cationic channel is important in GPCR-Gi protein signal transduction as it decreases heart rate and increases GI rhythm frequency. In the SA node of the heart, acetylcholine binds to the M2 receptor and the released Gβγ activates GIRK (I(K,ACh)) channel, inducing a negative chronotropic action. In gastric smooth muscle, there are two muscarinic acetylcholine receptor (mAChR) subtypes, M2 and M3. M2 receptor activates the muscarinic receptor-operated nonselective cationic current (mIcat, NSCC(ACh)) and induces positive chronotropic effect. Meanwhile, M3 receptor induces hydrolysis of PIP2 and releases DAG and IP3. This IP3 increases intracellular Ca2+ and then leads to contraction of GI smooth muscles. The activation of mIcat is inhibited by anti-Gi/o protein antibodies in GI smooth muscle, indicating the involvement of Gαi/o protein in the activation of mIcat. TRPC4 channel is a molecular candidate for mIcat and can be directly activated by constitutively active Gαi QL proteins. TRPC4 and TRPC5 belong to the same subfamily and both are activated by Gi/o proteins. Initial studies suggested that the binding sites for G protein exist at the rib helix or the CIRB domain of TRPC4/5 channels. However, recent cryo-EM structure showed that IYY58-60 amino acids at ARD of TRPC5 binds with Gi3 protein. Considering the expression of TRPC4/5 in the brain, the direct G protein activation on TRPC4/5 is important in terms of neurophysiology. TRPC4/5 channels are also suggested as a coincidence detector for Gi and Gq pathway as Gq pathway increases intracellular Ca2+ and the increased Ca2+ facilitates the activation of TRPC4/5 channels. More complicated situation would occur when GIRK, KCNQ2/3 (IM) and TRPC4/5 channels are co-activated by stimulation of muscarinic receptors at the acetylcholine-releasing nerve terminals. This review highlights the effects of GPCR-Gi protein pathway, including dopamine, μ-opioid, serotonin, glutamate, GABA, on various oragns, and it emphasizes the importance of considering TRPC4/5 channels as crucial players in the field of neuroscience.
Collapse
Affiliation(s)
- Hana Kang
- Department of Physiology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jinhyeong Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Christine Haewon Park
- Department of Physiology, University of California, San Francisco, San Francisco, CA, United States
| | - Byeongseok Jeong
- Department of Physiology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Insuk So
- Department of Physiology, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
12
|
Gei L, Yan Y, Xing W, Li Q, Chen X, Yan F, Wang Y, Cao Y, Jiang W, E R, Luo D, Zhang Y, Zeng W, Chen D. Amiloride alleviates morphine tolerance by suppressing ASIC3-dependent neuroinflammation in the spinal cord. Eur J Pharmacol 2024; 963:176173. [PMID: 37918499 DOI: 10.1016/j.ejphar.2023.176173] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/30/2023] [Accepted: 10/30/2023] [Indexed: 11/04/2023]
Abstract
BACKGROUND The use of morphine in clinical medicine is severely constrained by tolerance. Therefore, it is essential to examine pharmacological therapies that suppress the development of morphine tolerance. Amiloride suppressed the expression of inflammatory cytokines by inhibiting microglial activation. Microglia play a crucial role in the establishment of morphine tolerance. Thus, we anticipated that amiloride might suppress the development of morphine tolerance. During this investigation, we assessed the impact of amiloride on mouse morphine tolerance. METHODS Mice received morphine (10 mg/kg, s.c.) twice daily with intrathecally injected amiloride (0.3 μg/5 μl, 1 μg/5 μl, and 3 μg/5 μl) for nine continuous days. To assess morphine tolerance, mice underwent the tail-flick and hot plate tests. BV-2 cells were used to investigate the mechanism of amiloride. By using Western blotting, real-time PCR, and immunofluorescence labeling methods, the levels of acid-sensing ion channels (ASICs), nuclear factor kappa B (NF-kB) p65, p38 mitogen-activated protein kinase (MAPK) proteins, and neuroinflammation-related cytokines were determined. RESULTS The levels of ASIC3 in the spinal cord were considerably increased after long-term morphine administration. Amiloride was found to delay the development of tolerance to chronic morphine assessed via tail-flick and hot plate tests. Amiloride reduced microglial activation and downregulated the cytokines IL-1β and TNF-a by inhibiting ASIC3 in response to morphine. Furthermore, amiloride reduced p38 MAPK phosphorylation and inhibited NF-κB expression. CONCLUSIONS Amiloride effectively reduces chronic morphine tolerance by suppressing microglial activation caused by morphine by inhibiting ASIC3.
Collapse
Affiliation(s)
- Liba Gei
- Department of Anaesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China; Department of Anaesthesiology, Peking University Cancer Hospital (Inner Mongolia Campus)/Affiliated Cancer Hospital of Inner Mongolia Medical University/Inner Mongolia Autonomous Region Cancer Hospital, Hohhot, 010010, China
| | - Yan Yan
- Department of Anaesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China; Department of Anaesthesiology, Huizhou Municipal Central Hospital, Huizhou, 516001, China
| | - Wei Xing
- Department of Anaesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Qiang Li
- Department of Anaesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Xiangnan Chen
- Department of Anaesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China; Department of Anaesthesiology, Guangdong Women and Children Hospital, Guangzhou, 510060, China
| | - Fang Yan
- Department of Anaesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Yan Wang
- Department of Anaesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Yan Cao
- Department of Anaesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Wenqi Jiang
- Department of Anaesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - RiQi E
- Department of Anaesthesiology, Peking University Cancer Hospital (Inner Mongolia Campus)/Affiliated Cancer Hospital of Inner Mongolia Medical University/Inner Mongolia Autonomous Region Cancer Hospital, Hohhot, 010010, China
| | - DeXing Luo
- Department of Anaesthesiology, Huizhou Municipal Central Hospital, Huizhou, 516001, China
| | - YanHong Zhang
- Department of Anaesthesiology, Peking University Cancer Hospital (Inner Mongolia Campus)/Affiliated Cancer Hospital of Inner Mongolia Medical University/Inner Mongolia Autonomous Region Cancer Hospital, Hohhot, 010010, China
| | - Weian Zeng
- Department of Anaesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
| | - Dongtai Chen
- Department of Anaesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
| |
Collapse
|
13
|
Wang TZ, Wang F, Tian ZC, Li ZZ, Liu WN, Ding H, Xie TT, Cao ZX, Li HT, Sun ZC, Xie RG, Wu SX, Pan ZX, Luo C. Cingulate cGMP-dependent protein kinase I facilitates chronic pain and pain-related anxiety and depression. Pain 2023; 164:2447-2462. [PMID: 37326662 DOI: 10.1097/j.pain.0000000000002952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 04/25/2023] [Indexed: 06/17/2023]
Abstract
ABSTRACT Patients with chronic pain often experience exaggerated pain response and aversive emotion, such as anxiety and depression. Central plasticity in the anterior cingulate cortex (ACC) is assumed to be a critical interface for pain perception and emotion, which has been reported to involve activation of NMDA receptors. Numerous studies have documented the key significance of cGMP-dependent protein kinase I (PKG-I) as a crucial downstream target for the NMDA receptor-NO-cGMP signaling cascade in regulating neuronal plasticity and pain hypersensitivity in specific regions of pain pathway, ie, dorsal root ganglion or spinal dorsal horn. Despite this, whether and how PKG-I in the ACC contributes to cingulate plasticity and comorbidity of chronic pain and aversive emotion has remained elusive. Here, we uncovered a crucial role of cingulate PKG-I in chronic pain and comorbid anxiety and depression. Chronic pain caused by tissue inflammation or nerve injury led to upregulation of PKG-I expression at both mRNA and protein levels in the ACC. Knockdown of ACC-PKG-I relieved pain hypersensitivity as well as pain-associated anxiety and depression. Further mechanistic analysis revealed that PKG-I might act to phosphorylate TRPC3 and TRPC6, leading to enhancement of calcium influx and neuronal hyperexcitability as well as synaptic potentiation, which results in the exaggerated pain response and comorbid anxiety and depression. We believe this study sheds new light on the functional capability of ACC-PKG-I in modulating chronic pain as well as pain-associated anxiety and depression. Hence, cingulate PKG-I may represent a new therapeutic target against chronic pain and pain-related anxiety and depression.
Collapse
Affiliation(s)
- Tao-Zhi Wang
- Department of Anesthesiology, The Second Hospital of Jilin University, Jilin University, Changchun, China
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Fei Wang
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
- Medical Experiment Center, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Zhi-Cheng Tian
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhen-Zhen Li
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Wan-Neng Liu
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
- College of Life Sciences, Northwest University, Xi'an, China
| | - Hui Ding
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Ting-Ting Xie
- Department of Anesthesiology, The Second Hospital of Jilin University, Jilin University, Changchun, China
| | - Zi-Xuan Cao
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
- The Twenty-second Squadron of the Sixth Regiment, School of Basal Medicine, Fourth Military Medical University, Xi'an, China
| | - Hai-Tao Li
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
- The Fourteenth Squadron of the Fourth Regiment, School of Basal Medicine, Fourth Military Medical University, Xi'an, China
| | - Zhi-Chuan Sun
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
- Department of Neurosurgery, Xi'an Daxing Hospital, Xi'an, China
| | - Rou-Gang Xie
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Sheng-Xi Wu
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Zhen-Xiang Pan
- Department of Anesthesiology, The Second Hospital of Jilin University, Jilin University, Changchun, China
| | - Ceng Luo
- Department of Neurobiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
14
|
Spinal TRPA1 Contributes to the Mechanical Hypersensitivity Effect Induced by Netrin-1. Int J Mol Sci 2022; 23:ijms23126629. [PMID: 35743067 PMCID: PMC9224357 DOI: 10.3390/ijms23126629] [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: 04/08/2022] [Revised: 06/10/2022] [Accepted: 06/12/2022] [Indexed: 02/06/2023] Open
Abstract
Netrin-1, a chemoattractant expressed by floor plate cells, and one of its receptors (deleted in colorectal cancer) has been associated with pronociceptive actions in a number of pain conditions. Here, we addressed the question of whether spinal TRPC4/C5 or TRPA1 are among the downstream receptors contributing to pronociceptive actions induced by netrin-1. The experiments were performed on rats using a chronic intrathecal catheter for administration of netrin-1 and antagonists of TRPC4/C5 or TRPA1. Pain sensitivity was assessed behaviorally by using mechanical and heat stimuli. Effect on the discharge rate of rostral ventromedial medullary (RVM) pain control neurons was studied in lightly anesthetized animals. Netrin-1, in a dose-related fashion, induced mechanical hypersensitivity that lasted up to three weeks. Netrin-1 had no effect on heat nociception. Mechanical hypersensitivity induced by netrin-1 was attenuated by TRPA1 antagonist Chembridge-5861528 and by the control analgesic compound pregabalin both during the early (first two days) and late (third week) phase of hypersensitivity. TRPC4/C5 antagonist ML-204 had a weak antihypersensitivity effect that was only in the early phase, whereas TRPC4/C5 antagonist HC-070 had no effect on hypersensitivity induced by netrin-1. The discharge rate in pronociceptive ON-like RVM neurons was increased by netrin-1 during the late but not acute phase, whereas netrin-1 had no effect on the discharge rate of antinociceptive RVM OFF-like neurons. The results suggest that spinal TRPA1 receptors and pronociceptive RVM ON-like neurons are involved in the maintenance of submodality-selective pronociceptive actions induced by netrin-1 in the spinal cord.
Collapse
|
15
|
Zergane M, Kuebler WM, Michalick L. Heteromeric TRP Channels in Lung Inflammation. Cells 2021; 10:cells10071654. [PMID: 34359824 PMCID: PMC8307017 DOI: 10.3390/cells10071654] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 06/09/2021] [Accepted: 06/25/2021] [Indexed: 12/15/2022] Open
Abstract
Activation of Transient Receptor Potential (TRP) channels can disrupt endothelial barrier function, as their mediated Ca2+ influx activates the CaM (calmodulin)/MLCK (myosin light chain kinase)-signaling pathway, and thereby rearranges the cytoskeleton, increases endothelial permeability and thus can facilitate activation of inflammatory cells and formation of pulmonary edema. Interestingly, TRP channel subunits can build heterotetramers, whereas heteromeric TRPC1/4, TRPC3/6 and TRPV1/4 are expressed in the lung endothelium and could be targeted as a protective strategy to reduce endothelial permeability in pulmonary inflammation. An update on TRP heteromers and their role in lung inflammation will be provided with this review.
Collapse
Affiliation(s)
- Meryam Zergane
- Institute of Physiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (M.Z.); (L.M.)
| | - Wolfgang M. Kuebler
- Institute of Physiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (M.Z.); (L.M.)
- German Centre for Cardiovascular Research (DZHK), 10785 Berlin, Germany
- German Center for Lung Research (DZL), 35392 Gießen, Germany
- The Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada
- Department of Surgery and Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Correspondence:
| | - Laura Michalick
- Institute of Physiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany; (M.Z.); (L.M.)
- German Centre for Cardiovascular Research (DZHK), 10785 Berlin, Germany
| |
Collapse
|
16
|
Santoni A, Mercadante S, Arcuri E. Chronic cancer and non-cancer pain and opioid-induced hyperalgesia share common mechanisms: neuroinflammation and central sensitization. Minerva Anestesiol 2020; 87:210-222. [PMID: 33300326 DOI: 10.23736/s0375-9393.20.14822-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Neuroinflammation, a peculiar form of inflammation that occurs in response to noxious stimuli in peripheral and central nervous system (CNS), consists in altered vascular permeability followed by leukocyte recruitment and activation in the inflamed tissue, release of inflammatory mediators including cytokines and chemokines, and finally in the activation of microglia and astrocytes in the spinal cord and CNS. This phenomenon mediates and even worsen the inflammatory pain in many painful states and is responsible for central sensitization leading to pain chronicity. We describe the major neuroinflammatory mechanisms shared by cancer and non-cancer pain. Particular attention is given to two different chronic inflammatory painful diseases such as the complex regional pain syndrome and the rheumatoid arthritis as prototypes of neuroinflammatory diseases (gliopathies). In addition, we describe the complexity of tumor microenvironment, their main cellular components (tumor cells, tumor infiltrating leukocytes and sensory neurons) and their reciprocal interactions that characterize different forms and intensity of cancer pain. We also hypothesize that one type of cancer pain, the breakthrough pain, can be attributable to receptor-mediated interaction of opioids with tumor cells and intratumoral leukocytes. Surprisingly, long-term opioid treatment shares the same neuroinflammatory potential responsible for the chronicity of both cancer and non-cancer pain; thus, resulting in paradoxical worsening rather than relieving pain. This paradox has upset the world of pain therapy, with neuroinflammation now being a main target of emerging therapies.
Collapse
Affiliation(s)
- Angela Santoni
- Department of Molecular Medicine, Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, Rome, Italy.,IRCCS Neuromed, Pozzilli, Isernia, Italy
| | - Sebastiano Mercadante
- Main Regional Center of Supportive/Palliative Care, La Maddalena Cancer Center, Palermo, Italy
| | - Edoardo Arcuri
- IRCCS Regina Elena Cancer Institute, IFO, Rome, Italy - .,Ars Medica Pain Clinic, Rome, Italy
| |
Collapse
|
17
|
Comparison of Different In Vivo Animal Models of Brachial Plexus Avulsion and Its Application in Pain Study. Neural Plast 2020; 2020:8875915. [PMID: 33273909 PMCID: PMC7676973 DOI: 10.1155/2020/8875915] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 10/26/2020] [Accepted: 10/29/2020] [Indexed: 01/11/2023] Open
Abstract
Brachial plexus injuries (BPIs) are high-energy trauma that can result in serious functional problems in the affected upper extremities, and brachial plexus avulsion (BPA) could be considered the most severe type of them. The booming occurrence rate of BPA brings up devastating impact on patients' life. Complications of muscle atrophy, neuropathic pain, and denervation-associated psychological disorders are major challenges in the treatment of BPA. Animal models of BPA are good vehicles for this kind of research. Full understanding of the current in vivo BPA models, which could be classified into anterior approach avulsion, posterior approach avulsion, and closed approach avulsion groups, could help researchers select the appropriate type of models for their studies. Each group of the BPA model has its distinct merits and demerits. An ideal BPA model that can inherit the advantages and make up for the disadvantages is still required for further exploration.
Collapse
|
18
|
Lavanderos B, Silva I, Cruz P, Orellana-Serradell O, Saldías MP, Cerda O. TRP Channels Regulation of Rho GTPases in Brain Context and Diseases. Front Cell Dev Biol 2020; 8:582975. [PMID: 33240883 PMCID: PMC7683514 DOI: 10.3389/fcell.2020.582975] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 10/05/2020] [Indexed: 12/13/2022] Open
Abstract
Neurological and neuropsychiatric disorders are mediated by several pathophysiological mechanisms, including developmental and degenerative abnormalities caused primarily by disturbances in cell migration, structural plasticity of the synapse, and blood-vessel barrier function. In this context, critical pathways involved in the pathogenesis of these diseases are related to structural, scaffolding, and enzymatic activity-bearing proteins, which participate in Ca2+- and Ras Homologs (Rho) GTPases-mediated signaling. Rho GTPases are GDP/GTP binding proteins that regulate the cytoskeletal structure, cellular protrusion, and migration. These proteins cycle between GTP-bound (active) and GDP-bound (inactive) states due to their intrinsic GTPase activity and their dynamic regulation by GEFs, GAPs, and GDIs. One of the most important upstream inputs that modulate Rho GTPases activity is Ca2+ signaling, positioning ion channels as pivotal molecular entities for Rho GTPases regulation. Multiple non-selective cationic channels belonging to the Transient Receptor Potential (TRP) family participate in cytoskeletal-dependent processes through Ca2+-mediated modulation of Rho GTPases. Moreover, these ion channels have a role in several neuropathological events such as neuronal cell death, brain tumor progression and strokes. Although Rho GTPases-dependent pathways have been extensively studied, how they converge with TRP channels in the development or progression of neuropathologies is poorly understood. Herein, we review recent evidence and insights that link TRP channels activity to downstream Rho GTPase signaling or modulation. Moreover, using the TRIP database, we establish associations between possible mediators of Rho GTPase signaling with TRP ion channels. As such, we propose mechanisms that might explain the TRP-dependent modulation of Rho GTPases as possible pathways participating in the emergence or maintenance of neuropathological conditions.
Collapse
Affiliation(s)
- Boris Lavanderos
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - Ian Silva
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - Pablo Cruz
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - Octavio Orellana-Serradell
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - María Paz Saldías
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile
| | - Oscar Cerda
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Nucleus of Ion Channel-Associated Diseases (MiNICAD), Santiago, Chile.,The Wound Repair, Treatment and Health (WoRTH) Initiative, Santiago, Chile
| |
Collapse
|