1
|
Sawada S, Hitomi S, Hayashi Y, Shinozuka H, Yonehara Y, Iwata K, Shinoda M. Role of trigeminal ganglion satellite glial cells in masseter muscle pain hypersensitivity. J Oral Biosci 2025; 67:100596. [PMID: 39672375 DOI: 10.1016/j.job.2024.100596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 12/15/2024]
Abstract
OBJECTIVES The underlying mechanism of masseter muscle pain hypersensitivity by sustained masseter muscle contraction (SMMC) is not well understood. This study aimed to examine whether the activation of satellite glial cells in the trigeminal ganglion (TG) contributes to masseter muscle pain hypersensitivity induced by SMMC. METHODS Electrodes were placed on the masseter muscle fascia of rats to induce strong contractions, by daily electrical stimulation. Pain sensitivity in the masseter muscle was measured and the activation level of satellite glial cells in the TG was examined. The localization of P2Y12 and the effects of P2Y12 receptor inhibition on SMMC-induced pain hypersensitivity were evaluated. The amount of tumor necrosis factor alpha (TNF-α) and TNF-α receptor localization were determined in the TG. RESULTS SMMC induced masseter muscle pain hypersensitivity and activation of satellite glial cells. P2Y12 receptors were expressed in satellite glial cells and masseter muscle pain hypersensitivity was suppressed by intra-TG P2Y12 receptor antagonism. TG neurons innervating the sustained-contracted masseter muscle expressed TNF-α receptor and SMMC increased TNF-α levels in TG. CONCLUSION SMMC-induced activation of satellite glial cells though the P2Y12 receptor signaling may contribute to masseter muscle pain hypersensitivity via the TNF-α signaling pathway.
Collapse
Affiliation(s)
- Sho Sawada
- Department of Oral and Maxillofacial surgery, Nihon University School of Dentistry, 1-8-13 Kandasurugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
| | - Suzuro Hitomi
- Department of Physiology, Nihon University School of Dentistry, 1-8-13 Kandasurugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
| | - Yoshinori Hayashi
- Department of Physiology, Nihon University School of Dentistry, 1-8-13 Kandasurugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
| | - Hirotaka Shinozuka
- Department of Physiology, Nihon University School of Dentistry, 1-8-13 Kandasurugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
| | - Yoshiyuki Yonehara
- Department of Oral and Maxillofacial surgery, Nihon University School of Dentistry, 1-8-13 Kandasurugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
| | - Koichi Iwata
- Department of Physiology, Nihon University School of Dentistry, 1-8-13 Kandasurugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
| | - Masamichi Shinoda
- Department of Physiology, Nihon University School of Dentistry, 1-8-13 Kandasurugadai, Chiyoda-ku, Tokyo, 101-8310, Japan.
| |
Collapse
|
2
|
Kong Y, Pan T, Liu B, Kuss M, Krishnan MA, Alimi OA, Shi W, Duan B. Double-Layer Microneedle Patch Loaded with HA-PBA-QCT for Management of Paclitaxel-Induced Peripheral Neuropathic Pain. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2409748. [PMID: 39888259 PMCID: PMC11855232 DOI: 10.1002/smll.202409748] [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: 10/20/2024] [Revised: 12/24/2024] [Indexed: 02/01/2025]
Abstract
Chemotherapy-induced neuropathic pain (CINP) is a common adverse effect of antineoplastic drugs, often leading to dose reduction, treatment delays, or cessation of chemotherapy. Chemotherapy agents, like paclitaxel (PTX), damage the somatosensory nervous system by inducing neuroinflammation and oxidative stress, resulting in the sensitization of sensory neurons. Quercetin (QCT), known for its anti-inflammatory, antioxidant, and neuroprotective properties, is investigated for various neurological disorders. This work creates phenylboronic acid-modified hyaluronic acid (HA-PBA) gels with incorporated QCT and fabricates a double-layer microneedle (MN) patch using an HA-PBA-QCT complex in the needles and HA/polyvinyl alcohol (PVA) as the substrate. The crosslinking between PVA and HA-PBA-QCT enables a controlled, sustained release of QCT upon application. This work applies these QCT-loaded microneedle (QMN) patches to the instep skin of PTX-treated mice, which exhibits mechanical allodynia and cold hyperalgesia. Biweekly applications of the QMN patches significantly reduce pain responses. This analgesic effect is associated with the modulation of satellite glial cell activity, decreased macrophage infiltration, and reduced TNF-α and IL-6 levels in dorsal root ganglia (DRGs). Additionally, the treatment improves cellular antioxidant capacity, indicated by upregulated Nrf2 and catalase in DRGs. Overall, these findings suggest that double-layer QMN patches offer long-term anti-inflammatory and antioxidant benefits, potentially alleviating CINP in patients.
Collapse
Affiliation(s)
- Yunfan Kong
- Mary & Dick Holland Regenerative Medicine Program University of Nebraska Medical CenterOmahaNE68198USA
- Division of CardiologyDepartment of Internal MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Tianshu Pan
- Mary & Dick Holland Regenerative Medicine Program University of Nebraska Medical CenterOmahaNE68198USA
- Division of CardiologyDepartment of Internal MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Bo Liu
- Mary & Dick Holland Regenerative Medicine Program University of Nebraska Medical CenterOmahaNE68198USA
- Division of CardiologyDepartment of Internal MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Mitchell Kuss
- Mary & Dick Holland Regenerative Medicine Program University of Nebraska Medical CenterOmahaNE68198USA
- Division of CardiologyDepartment of Internal MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Mena A. Krishnan
- Mary & Dick Holland Regenerative Medicine Program University of Nebraska Medical CenterOmahaNE68198USA
- Division of CardiologyDepartment of Internal MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Olawale A. Alimi
- Mary & Dick Holland Regenerative Medicine Program University of Nebraska Medical CenterOmahaNE68198USA
- Division of CardiologyDepartment of Internal MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Wen Shi
- Mary & Dick Holland Regenerative Medicine Program University of Nebraska Medical CenterOmahaNE68198USA
- Division of CardiologyDepartment of Internal MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Bin Duan
- Mary & Dick Holland Regenerative Medicine Program University of Nebraska Medical CenterOmahaNE68198USA
- Division of CardiologyDepartment of Internal MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
- Department of SurgeryCollege of MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
- Department of Mechanical and Materials EngineeringUniversity of Nebraska‐LincolnLincolnNE68588USA
| |
Collapse
|
3
|
Tu C, Wang SC, Dai MX, Lai SQ, Huang ZW, Yu YP, Chen YB, Zeng JH, Wang L, Zhong ZM. Accumulation of advanced oxidative protein products exacerbate satellite glial cells activation and neuropathic pain. Mol Med 2025; 31:25. [PMID: 39865234 PMCID: PMC11765935 DOI: 10.1186/s10020-025-01076-x] [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: 10/21/2024] [Accepted: 01/10/2025] [Indexed: 01/28/2025] Open
Abstract
BACKGROUND Neuropathic pain (NP) is a debilitating condition caused by lesion or dysfunction in the somatosensory nervous system. Accumulation of advanced oxidation protein products (AOPPs) is implicated in mechanical hyperalgesia. However, the effects of AOPPs on NP remain unclear. METHODS A rat model of NP was established by chronic constriction injury (CCI) and employed to evaluate the changes of mechanical withdrawal threshold, thermal and cold withdrawal latency, as well as AOPPs levels. The effects of AOPPs on the activation of satellite glial cells (SGCs) in the dorsal root ganglion (DRG), receptor for advanced glycation end-products (RAGE) expression, and NF-κB signaling pathway activation were also investigated using western blotting, immunofluorescence, and the Fluo4-AM fluorescence probe for calcium signaling. Additionally, oxidative stress levels and inflammatory cytokine production in SGCs, triggered by AOPPs exposure, were measured through the DCFH-DA probe for ROS detection and ELISA kits for cytokine quantification. RESULTS CCI significantly elevated the AOPPs levels in the plasma and sciatic nerve and caused AOPPs accumulation in the DRG. Exogenous AOPPs activated SGCs, increased reactive oxygen species and inflammatory response, upregulated the RAGE, and activated NF-κB signaling. The RAGE inhibitor FPS-ZM1 effectively inhibited AOPPs-induced SGC activation. Additionally, AOPPs intervention worsened CCI-induced hyperalgesia and neuroinflammation in vivo. CONCLUSION These results indicate that AOPPs exacerbate the SGC activation and NP following nerve injury, and AOPPs accumulation might play an important role in the pathogenesis of NP.
Collapse
Affiliation(s)
- Chen Tu
- Department of Spine, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Shi-Cheng Wang
- Division of Spine Surgery, Department of Orthopedics, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou, 510515, People's Republic of China
| | - Meng-Xuan Dai
- Department of Spine, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Si-Qi Lai
- Department of Pathology, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Zhi-Wei Huang
- Division of Spine Surgery, Department of Orthopedics, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou, 510515, People's Republic of China
| | - Yong-Peng Yu
- Division of Spine Surgery, Department of Orthopedics, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou, 510515, People's Republic of China
| | - Yun-Biao Chen
- Division of Spine Surgery, Department of Orthopedics, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou, 510515, People's Republic of China
| | - Ji-Huan Zeng
- Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Liang Wang
- Department of Spine, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China.
| | - Zhao-Ming Zhong
- Division of Spine Surgery, Department of Orthopedics, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou, 510515, People's Republic of China.
| |
Collapse
|
4
|
Thomsen MB, Singh A, Thebeau CN, Gao VD, Schulze NF, Avraham O, Yang SX, Koneru S, Geier SS, Landon SM, Pelea A, Cavalli V, Geisler S. Macrophage depletion restores the DRG microenvironment and prevents axon degeneration in bortezomib-induced neuropathy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.22.634362. [PMID: 39896673 PMCID: PMC11785175 DOI: 10.1101/2025.01.22.634362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Peripheral neuropathy is a common and debilitating side effect of the chemotherapeutic bortezomib (BTZ). To explore the mechanisms underlying BTZ-induced neuropathy (BIPN), we developed a mouse model that replicates the route of administration and approximates the prolonged BTZ exposure experienced by patients. We find that male mice treated with BTZ experience more severe sensorimotor dysfunction and axon loss compared to females and observed similar results when analyzing human data. Using single cell RNA-sequencing, we reveal that BTZ significantly alters the dorsal root ganglia (DRG) microenvironment in mice, producing pronounced sex-specific changes in satellite glial cells (SGCs) in males and females and dysregulation of the extracellular matrix (ECM), particularly in males. These changes are accompanied by expansion of macrophages, which is more pronounced in males. We identify four macrophage subtypes in the DRG, including a pro-fibrotic population that is exclusively associated with BIPN. Depletion of macrophages via anti-CSF1R treatment in male mice prevents BTZ-induced SGC activation and aberrant collagen deposition in DRGs, potently preserves peripheral axons, and improves functional outcomes. These findings highlight SGCs, neuroinflammation and dysregulation of the ECM as drivers of sex-specific differences in BIPN and suggest that targeting neuroinflammation is a promising therapeutic strategy to treat this disease.
Collapse
Affiliation(s)
| | - Abhishek Singh
- Department of Neurology, Washington University School of Medicine in St. Louis; St. Louis, USA
| | - Christina N. Thebeau
- Department of Neurology, Washington University School of Medicine in St. Louis; St. Louis, USA
| | - Vivian D. Gao
- Department of Neurology, Washington University School of Medicine in St. Louis; St. Louis, USA
| | - Nicholas F. Schulze
- Department of Neurology, Washington University School of Medicine in St. Louis; St. Louis, USA
| | - Oshri Avraham
- Department of Neuroscience, Washington University School of Medicine in St. Louis; St. Louis, USA
| | - Sarah X. Yang
- Department of Neurology, Washington University School of Medicine in St. Louis; St. Louis, USA
| | - Shriya Koneru
- Department of Neurology, Washington University School of Medicine in St. Louis; St. Louis, USA
| | - Sami S. Geier
- Department of Neurology, Washington University School of Medicine in St. Louis; St. Louis, USA
| | - Shannon M. Landon
- Department of Neurology, Washington University School of Medicine in St. Louis; St. Louis, USA
| | - Aidan Pelea
- Department of Neurology, Washington University School of Medicine in St. Louis; St. Louis, USA
| | - Valeria Cavalli
- Department of Neuroscience, Washington University School of Medicine in St. Louis; St. Louis, USA
- Hope Center for Neurological Diseases, Washington University School of Medicine in St. Louis; St. Louis, USA
- Center of Regenerative Medicine, Washington University School of Medicine in St. Louis; St. Louis, USA
| | - Stefanie Geisler
- Department of Neurology, Washington University School of Medicine in St. Louis; St. Louis, USA
- Hope Center for Neurological Diseases, Washington University School of Medicine in St. Louis; St. Louis, USA
- Siteman Cancer Center; St. Louis, USA
| |
Collapse
|
5
|
Tao Z, Chen Z, Zeng X, Cui J, Quan M. An emerging aspect of cancer neuroscience: A literature review on chemotherapy-induced peripheral neuropathy. Cancer Lett 2024; 611:217433. [PMID: 39736454 DOI: 10.1016/j.canlet.2024.217433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 12/20/2024] [Accepted: 12/24/2024] [Indexed: 01/01/2025]
Abstract
The nervous system governs both ontogeny and oncology. Foundational discoveries have clarified the direct communication of neurotransmitters with tumors and indirect interactions through neural effects on the immune system and the tumor microenvironment. Meantime, the nervous system is susceptible to cancer and its treatment. Chemotherapy-induced peripheral neuropathy (CIPN) is the most common side effects that significantly reduce the efficacy of anti-cancer treatment and patients' quality of life by leading to dose reduction or early cessation of chemotherapy. However, there are no effective strategies to reverse or treat CIPN. A better understanding of the mechanisms is expected to enable the development of the next generation of therapies. Here, we summarize the recent important studies on clinical manifestations, risk factors, prediction, pathogenesis, prevention, and treatment of CIPN. We also provide perspectives and insights regarding the rationales of bidirectional interactions between cancer and the nervous system.
Collapse
Affiliation(s)
- Zhirui Tao
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200123, China
| | - Zhiqin Chen
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200123, China
| | - Xiaochen Zeng
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200123, China
| | - Jiujie Cui
- Department of Oncology and State Key Laboratory of Systems Medicine for Cancer of Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China.
| | - Ming Quan
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200123, China.
| |
Collapse
|
6
|
Li YZ, Ji RR. Gene therapy for chronic pain management. Cell Rep Med 2024; 5:101756. [PMID: 39366385 PMCID: PMC11513853 DOI: 10.1016/j.xcrm.2024.101756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 07/20/2024] [Accepted: 09/09/2024] [Indexed: 10/06/2024]
Abstract
Despite significant advances in identifying molecular targets for chronic pain over the past two decades, many remain difficult to target with traditional methods. Gene therapies such as antisense oligonucleotides (ASOs), RNA interference (RNAi), CRISPR, and virus-based delivery systems have played crucial roles in discovering and validating new pain targets. While there has been a surge in gene therapy-based clinical trials, those focusing on pain as the primary outcome remain uncommon. This review examines various gene therapy strategies, including ASOs, small interfering RNA (siRNAs), optogenetics, chemogenetics, and CRISPR, and their delivery methods targeting primary sensory neurons and non-neuronal cells, including glia and chondrocytes. We also explore emerging gene therapy tools and highlight gene therapy's clinical potential in pain management, including trials targeting pain-related diseases. Advances in single-cell analysis of sensory neurons and non-neuronal cells, along with the development of new delivery tools, are poised to accelerate the application of gene therapy in pain medicine.
Collapse
Affiliation(s)
- Yi-Ze Li
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA; Departments of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA; Departments of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
7
|
Zhang Y, Li T, Wang G, Ma Y. Advancements in Single-Cell RNA Sequencing and Spatial Transcriptomics for Central Nervous System Disease. Cell Mol Neurobiol 2024; 44:65. [PMID: 39387975 PMCID: PMC11467076 DOI: 10.1007/s10571-024-01499-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 09/26/2024] [Indexed: 10/15/2024]
Abstract
The incidence of central nervous system (CNS) disease has persistently increased over the last several years. There is an urgent need for effective methods to improve the cure rates of CNS disease. However, the precise molecular basis underlying the development and progression of major CNS diseases remains elusive. A complete molecular map will contribute to research on CNS disease treatment strategies. Emerging technologies such as single-cell RNA sequencing (scRNA-seq) and Spatial Transcriptomics (ST) are potent tools for exploring the molecular complexity, cell heterogeneity, and functional specificity of CNS disease. scRNA-seq and ST can provide insights into the disease at cellular and spatial transcription levels. This review presents a survey of scRNA-seq and ST studies on CNS diseases, such as chronic neurodegenerative diseases, acute CNS injuries, and others. These studies offer novel perspectives in treating and diagnosing CNS diseases by discovering new cell types or subtypes associated with the disease, proposing new pathophysiological mechanisms, uncovering novel therapeutic targets, and identifying putative biomarkers.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Pharmacy, School of Medicine, Shanghai East Hospital, Tongji University, Shanghai, 200120, China
| | - Teng Li
- Department of Laboratory Medicine, School of Medicine, Shanghai East Hospital, Tongji University, Shanghai, 200120, China
| | - Guangtian Wang
- Teaching Center of Pathogenic Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, Heilongjiang, China.
| | - Yabin Ma
- Department of Pharmacy, School of Medicine, Shanghai East Hospital, Tongji University, Shanghai, 200120, China.
| |
Collapse
|
8
|
Santi MD, Zhang M, Asam K, Yu G, Dong PM, Sheehan DH, Aouizerat BE, Thomas CM, Viet CT, Ye Y. Perineural Invasion Is Associated With Function-evoked Pain and Altered Extracellular Matrix in Patients With Head and Neck Squamous Cell Carcinoma. THE JOURNAL OF PAIN 2024; 25:104615. [PMID: 38936749 DOI: 10.1016/j.jpain.2024.104615] [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: 03/22/2024] [Revised: 06/01/2024] [Accepted: 06/19/2024] [Indexed: 06/29/2024]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is painful, and perineural invasion (PNI) has been associated with the worst pain. Pain due to HNSCC is diverse and may vary based on clinicopathological factors. This study aims to characterize different pain patterns linked with PNI, its influence on daily functioning, and gain insights into molecular changes and pathways associated with PNI-related pain in HNSCC patients. We conducted a cross-sectional study across 3 medical centers (n = 114), assessing pain phenotypes and their impact on daily functioning using 2 self-reported pain questionnaires, given to patients prior to their cancer surgery. Furthermore, we conducted RNA-seq analysis utilizing the The Cancer Genome Atlas dataset of HNSCC tumor from patients (n = 192) to identify genes relevant to both PNI and pain. Upon adjusting for demographic and clinicopathological variables using linear regression models, we found that PNI independently predicted function-evoked pain according to the University of Calfornia San Francisco Oral Cancer Pain Questionnaire, as well as the worst pain intensity reported in the Brief Pain Inventory. Distinct pain patterns were observed to be associated with daily activities in varying manners. Our molecular analyses revealed significant disruptions in pathways associated with the extracellular matrix structure and organization. The top differentially expressed genes linked to the extracellular matrix are implicated in cancer development, pain, and neurodegenerative diseases. Our data underscore the importance of properly categorizing pain phenotypes in future studies aiming to uncover mechanistic underpinnings of pain. Additionally, we have compiled a list of genes of interest that could serve as targets for both cancer and cancer pain management. PERSPECTIVE: PNI independently predicts function-evoked pain. Different pain phenotypes affect daily activities differently. We identified a list of candidate genes involved in the extracellular matrix structure and function that can be targeted for both cancer and cancer pain control.
Collapse
Affiliation(s)
- Maria D Santi
- Translational Research Center, College of Dentistry, New York University, New York, New York; Department of Molecular Pathobiology, Pain Research Center, College of Dentistry, New York University, New York, New York
| | - Morgan Zhang
- Translational Research Center, College of Dentistry, New York University, New York, New York; Department of Molecular Pathobiology, Pain Research Center, College of Dentistry, New York University, New York, New York
| | - Kesava Asam
- Translational Research Center, College of Dentistry, New York University, New York, New York
| | - Gary Yu
- Rory Meyers College of Nursing, New York University, New York, New York
| | - Phuong M Dong
- Department of Oral and Maxillofacial Surgery, Loma Linda University School of Dentistry, Loma Linda, California
| | - Delaney H Sheehan
- Department of Otolaryngology - Head & Neck Surgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - Bradley E Aouizerat
- Translational Research Center, College of Dentistry, New York University, New York, New York
| | - Carissa M Thomas
- Department of Otolaryngology - Head & Neck Surgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - Chi T Viet
- Department of Oral and Maxillofacial Surgery, Loma Linda University School of Dentistry, Loma Linda, California
| | - Yi Ye
- Translational Research Center, College of Dentistry, New York University, New York, New York; Department of Molecular Pathobiology, Pain Research Center, College of Dentistry, New York University, New York, New York.
| |
Collapse
|
9
|
Ege E, Briggi D, Vu P, Cheng J, Lin F, Xu J. Targeting dorsal root ganglia for chemotherapy-induced peripheral neuropathy: from bench to bedside. Ther Adv Neurol Disord 2024; 17:17562864241252718. [PMID: 39318973 PMCID: PMC11421407 DOI: 10.1177/17562864241252718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2024] Open
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a debilitating condition affecting an increasing number of cancer survivors worldwide. However, insights into its pathophysiology and availability of effective therapies remain lacking. Dorsal root ganglia (DRG) have been studied as a key component of chemotherapeutic drug toxicity and a potential therapeutic target for CIPN treatment. This comprehensive review aims to synthesize, summarize, and correlate the results of both preclinical and clinical studies relevant to the pathophysiology and management of CIPN in relation to the DRG. Design: Review. A thorough literature search was conducted using the terms 'dorsal root ganglion' and 'chemotherapy-induced peripheral neuropathy', along with appropriate variations. Searched databases included PubMed, EMBASE, Medline, Cochrane Library, Wiley Library, and Web of Science. Inclusion criteria targeted all English language, peer-reviewed original research from the inception of these databases to the present year. Review articles, book chapters, and other nonoriginal publications were excluded. Of 134 relevant studies identified, the majority were preclinical studies elucidating how various chemotherapeutic agents, especially taxanes, disrupt neurotransmission, inflammatory processes, and apoptotic pathways within sensory neurons of DRG. Not only do these effects correlate with the presentation of CIPN, but their disruption has also been shown to reduce CIPN symptoms in preclinical models. However, clinical studies addressing DRG interventions are very limited in number and scope at this time. These results reveal various pathways within DRG that may be effective targets for CIPN treatment. While limited, clinical studies do offer promise in the utility of DRG neuromodulation in managing painful CIPN. In the future, clinical trials are needed to assess interventions aimed at these neuronal and nonneuronal pathological targets to better treat this complex condition.
Collapse
Affiliation(s)
- Eliana Ege
- Department of Physical Medicine and Rehabilitation, Baylor College of Medicine, Houston, TX, USA
| | - Daniel Briggi
- Department of Physical Medicine and Rehabilitation, Baylor College of Medicine, Houston, TX, USA
| | - Peter Vu
- Department of Physical Medicine and Rehabilitation, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jianguo Cheng
- Department of Pain Management, Cleveland Clinic, Cleveland, OH, USA
- Department of Neuroscience, Cleveland Clinic, Cleveland, OH, USA
| | - Feng Lin
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, OH, USA
| | - Jijun Xu
- Department of Pain Management and Inflammation and Immunity, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| |
Collapse
|
10
|
Yuan BT, Li MN, Zhu LP, Xu ML, Gu J, Gao YJ, Ma LJ. TFAP2A is involved in neuropathic pain by regulating Grin1 expression in glial cells of the dorsal root ganglion. Biochem Pharmacol 2024; 227:116427. [PMID: 39009095 DOI: 10.1016/j.bcp.2024.116427] [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: 04/03/2024] [Revised: 06/20/2024] [Accepted: 07/12/2024] [Indexed: 07/17/2024]
Abstract
Neuropathic pain is a highly prevalent and refractory condition, yet its mechanism remains poorly understood. While NR1, the essential subunit of NMDA receptors, has long been recognized for its pivotal role in nociceptive transmission, its involvement in presynaptic stimulation is incompletely elucidated. Transcription factors can regulate the expression of both pro-nociceptive and analgesic factors. Our study shows that transcription factor TFAP2A was up-regulated in the dorsal root ganglion (DRG) neurons, satellite glial cells (SGCs), and Schwann cells following spinal nerve ligation (SNL). Intrathecal injection of siRNA targeting Tfap2a immediately or 7 days after SNL effectively alleviated SNL-induced pain hypersensitivity and reduced Tfap2a expression levels. Bioinformatics analysis revealed that TFAP2A may regulate the expression of the Grin1 gene, which encodes NR1. Dual-luciferase reporter assays confirmed TFAP2A's positive regulation of Grin1 expression. Notably, both Tfap2a and Grin1 were expressed in the primary SGCs and upregulated by lipopolysaccharides. The expression of Grin1 was also down-regulated in the DRG following Tfap2a knockdown. Furthermore, intrathecal injection of siRNA targeting Grin1 immediately or 7 days post-SNL effectively alleviated SNL-induced mechanical allodynia and thermal hyperalgesia. Finally, intrathecal Tfap2a siRNA alleviated SNL-induced neuronal hypersensitivity, and incubation of primary SGCs with Tfap2a siRNA decreased NMDA-induced upregulation of proinflammatory cytokines. Collectively, our study reveals the role of TFAP2A-Grin1 in regulating neuropathic pain in peripheral glia, offering a new strategy for the development of novel analgesics.
Collapse
Affiliation(s)
- Bao-Tong Yuan
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226019, China
| | - Meng-Na Li
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226019, China
| | - Lin-Peng Zhu
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226019, China
| | - Meng-Lin Xu
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226019, China
| | - Jun Gu
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226019, China
| | - Yong-Jing Gao
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226019, China.
| | - Ling-Jie Ma
- Institute of Pain Medicine and Special Environmental Medicine, Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226019, China.
| |
Collapse
|
11
|
Qiao LY. Satellite Glial Cells Bridge Sensory Neuron Crosstalk in Visceral Pain and Cross-Organ Sensitization. J Pharmacol Exp Ther 2024; 390:213-221. [PMID: 38777604 PMCID: PMC11264254 DOI: 10.1124/jpet.123.002061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 05/07/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024] Open
Abstract
Following colonic inflammation, the uninjured bladder afferent neurons are also activated. The mechanisms and pathways underlying this sensory neuron cross-activation (from injured neurons to uninjured neurons) are not fully understood. Colonic and bladder afferent neurons reside in the same spinal segments and are separated by satellite glial cells (SGCs) and extracellular matrix in dorsal root ganglia (DRG). SGCs communicate with sensory neurons in a bidirectional fashion. This review summarizes the differentially regulated genes/proteins in the injured and uninjured DRG neurons and explores the role of SGCs in regulation of sensory neuron crosstalk in visceral cross-organ sensitization. The review also highlights the paracrine pathways in mediating neuron-SGC and SGC-neuron coupling with an emphasis on the neurotrophins and purinergic systems. Finally, I discuss the results from recent RNAseq profiling of SGCs to reveal useful molecular markers for characterization, functional study, and therapeutic targets of SGCs. SIGNIFICANCE STATEMENT: Satellite glial cells (SGCs) are the largest glial subtypes in sensory ganglia and play a critical role in mediating sensory neuron crosstalk, an underlying mechanism in colon-bladder cross-sensitization. Identification of novel and unique molecular markers of SGCs can advance the discovery of therapeutic targets in treatment of chronic pain including visceral pain comorbidity.
Collapse
Affiliation(s)
- Liya Y Qiao
- Department of Physiology and Biophysics, Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| |
Collapse
|
12
|
Bhuiyan SA, Xu M, Yang L, Semizoglou E, Bhatia P, Pantaleo KI, Tochitsky I, Jain A, Erdogan B, Blair S, Cat V, Mwirigi JM, Sankaranarayanan I, Tavares-Ferreira D, Green U, McIlvried LA, Copits BA, Bertels Z, Del Rosario JS, Widman AJ, Slivicki RA, Yi J, Sharif-Naeini R, Woolf CJ, Lennerz JK, Whited JL, Price TJ, Gereau RW, Renthal W. Harmonized cross-species cell atlases of trigeminal and dorsal root ganglia. SCIENCE ADVANCES 2024; 10:eadj9173. [PMID: 38905344 PMCID: PMC11804847 DOI: 10.1126/sciadv.adj9173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 05/16/2024] [Indexed: 06/23/2024]
Abstract
Sensory neurons in the dorsal root ganglion (DRG) and trigeminal ganglion (TG) are specialized to detect and transduce diverse environmental stimuli to the central nervous system. Single-cell RNA sequencing has provided insights into the diversity of sensory ganglia cell types in rodents, nonhuman primates, and humans, but it remains difficult to compare cell types across studies and species. We thus constructed harmonized atlases of the DRG and TG that describe and facilitate comparison of 18 neuronal and 11 non-neuronal cell types across six species and 31 datasets. We then performed single-cell/nucleus RNA sequencing of DRG from both human and the highly regenerative axolotl and found that the harmonized atlas also improves cell type annotation, particularly of sparse neuronal subtypes. We observed that the transcriptomes of sensory neuron subtypes are broadly similar across vertebrates, but the expression of functionally important neuropeptides and channels can vary notably. The resources presented here can guide future studies in comparative transcriptomics, simplify cell-type nomenclature differences across studies, and help prioritize targets for future analgesic development.
Collapse
Affiliation(s)
- Shamsuddin A. Bhuiyan
- Department of Neurology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Mengyi Xu
- Department of Neurology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
- Alan Edwards Center for Research on Pain and Department of Physiology, McGill University, Montreal, QC, H3G 1Y6, Canada
| | - Lite Yang
- Department of Neurology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Evangelia Semizoglou
- Department of Neurology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Parth Bhatia
- Department of Neurology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Katerina I. Pantaleo
- Department of Neurology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Ivan Tochitsky
- F.M. Kirby Neurobiology Center and Department of Neurobiology, Boston Children’s Hospital and Harvard Medical School, 3 Blackfan Cir., Boston, MA 02115, USA
| | - Aakanksha Jain
- F.M. Kirby Neurobiology Center and Department of Neurobiology, Boston Children’s Hospital and Harvard Medical School, 3 Blackfan Cir., Boston, MA 02115, USA
| | - Burcu Erdogan
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Steven Blair
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Victor Cat
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Juliet M. Mwirigi
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX, 75080, USA
| | - Ishwarya Sankaranarayanan
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX, 75080, USA
| | - Diana Tavares-Ferreira
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX, 75080, USA
| | - Ursula Green
- Department of Pathology, Center for Integrated Diagnostics, Massachussetts General Hospital and Havard Medical School, Boston, MA 02114, USA
| | - Lisa A. McIlvried
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Bryan A. Copits
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Zachariah Bertels
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - John S. Del Rosario
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Allie J. Widman
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Richard A. Slivicki
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Jiwon Yi
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Reza Sharif-Naeini
- Alan Edwards Center for Research on Pain and Department of Physiology, McGill University, Montreal, QC, H3G 1Y6, Canada
| | - Clifford J. Woolf
- F.M. Kirby Neurobiology Center and Department of Neurobiology, Boston Children’s Hospital and Harvard Medical School, 3 Blackfan Cir., Boston, MA 02115, USA
| | - Jochen K. Lennerz
- Department of Pathology, Center for Integrated Diagnostics, Massachussetts General Hospital and Havard Medical School, Boston, MA 02114, USA
| | - Jessica L. Whited
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Theodore J. Price
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX, 75080, USA
| | - Robert W. Gereau
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - William Renthal
- Department of Neurology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
13
|
Li X, Prudente AS, Prato V, Guo X, Hao H, Jones F, Figoli S, Mullen P, Wang Y, Tonello R, Lee SH, Shah S, Maffei B, Berta T, Du X, Gamper N. Peripheral gating of mechanosensation by glial diazepam binding inhibitor. J Clin Invest 2024; 134:e176227. [PMID: 38888973 PMCID: PMC11324294 DOI: 10.1172/jci176227] [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: 09/27/2023] [Accepted: 06/13/2024] [Indexed: 06/20/2024] Open
Abstract
We report that diazepam binding inhibitor (DBI) is a glial messenger mediating crosstalk between satellite glial cells (SGCs) and sensory neurons in the dorsal root ganglion (DRG). DBI is highly expressed in SGCs of mice, rats, and humans, but not in sensory neurons or most other DRG-resident cells. Knockdown of DBI results in a robust mechanical hypersensitivity without major effects on other sensory modalities. In vivo overexpression of DBI in SGCs reduces sensitivity to mechanical stimulation and alleviates mechanical allodynia in neuropathic and inflammatory pain models. We further show that DBI acts as an unconventional agonist and positive allosteric modulator at the neuronal GABAA receptors, particularly strongly affecting those with a high-affinity benzodiazepine binding site. Such receptors are selectively expressed by a subpopulation of mechanosensitive DRG neurons, and these are also more enwrapped with DBI-expressing glia, as compared with other DRG neurons, suggesting a mechanism for a specific effect of DBI on mechanosensation. These findings identified a communication mechanism between peripheral neurons and SGCs. This communication modulates pain signaling and can be targeted therapeutically.
Collapse
Affiliation(s)
- Xinmeng Li
- Department of Pharmacology; The Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Arthur Silveira Prudente
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, Ohio, USA
| | - Vincenzo Prato
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| | - Xianchuan Guo
- Department of Pharmacology; The Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Han Hao
- Department of Pharmacology; The Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Frederick Jones
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
- Department of Life Sciences, Manchester Metropolitan University, Manchester, United Kingdom
| | - Sofia Figoli
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| | - Pierce Mullen
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| | - Yujin Wang
- Department of Pharmacology; The Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Raquel Tonello
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, Ohio, USA
| | - Sang Hoon Lee
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, Ohio, USA
| | - Shihab Shah
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| | - Benito Maffei
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| | - Temugin Berta
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, Ohio, USA
| | - Xiaona Du
- Department of Pharmacology; The Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Nikita Gamper
- Department of Pharmacology; The Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
14
|
Hu L, An E, Zhu Z, Cai Y, Ye X, Zhou H, Ge H. Grape seed-derived procyanidins decreases neuropathic pain and nerve regeneration by suppression of toll-like receptor 4-myeloid differentiation factor-88 signaling. Mol Pain 2024; 20:17448069241256466. [PMID: 38716504 PMCID: PMC11110500 DOI: 10.1177/17448069241256466] [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: 03/07/2024] [Revised: 04/15/2024] [Accepted: 04/19/2024] [Indexed: 05/22/2024] Open
Abstract
Background: Recent studies have shown that peripheral nerve regeneration process is closely related to neuropathic pain. Toll-like receptor 4 (TLR4) signaling was involved in different types of pain and nerve regeneration. TLR4 induced the recruitment of myeloid differentiation factor-88 adaptor protein (MyD88) and NF-κB-depended transcriptional process in sensory neurons and glial cells, which produced multiple cytokines and promoted the induction and persistence of pain. Our study aimed to investigate procyanidins's effect on pain and nerve regeneration via TLR4-Myd88 signaling. Methods: Spinal nerve ligation (SNL) model was established to measure the analgesic effect of procyanidins. Anatomical measurement of peripheral nerve regeneration was measured by microscopy and growth associated protein 43 (GAP43) staining. Western blotting and/or immunofluorescent staining were utilized to detect TLR4, myeloid differentiation factor-88 adaptor protein (MyD88), ionized calcium-binding adapter molecule 1 (IBA1) and nuclear factor kappa-B-p65 (NF-κB-p65) expression, as well as the activation of astrocyte and microglia. The antagonist of TLR4 (LPS-RS-Ultra, LRU) were intrathecally administrated to assess the behavioral effects of blocking TLR4 signaling on pain and nerve regeneration. Result: Procyanidins reduced mechanical allodynia, thermal hyperalgesia and significantly suppressed the number of nerve fibers regenerated and the degree of myelination in SNL model. Compared with sham group, TLR4, MyD88, IBA1 and phosphorylation of NF-κB-p65 were upregulated in SNL rats which were reversed by procyanidins administration. Additionally, procyanidins also suppressed activation of spinal astrocytes and glial cells. Conclusion: Suppression of TLR4-MyD88 signaling contributes to the alleviation of neuropathic pain and reduction of nerve regeneration by procyanidins.
Collapse
Affiliation(s)
- Li Hu
- Department of Anesthesiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
- Key Laboratory of Basic Research and Clinical Transformation of Perioperative Precision Anesthesia, Jiaxing, China
| | - Erdan An
- Department of Anesthesiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
- Key Laboratory of Basic Research and Clinical Transformation of Perioperative Precision Anesthesia, Jiaxing, China
| | - ZhiPeng Zhu
- Department of Anesthesiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
- Key Laboratory of Basic Research and Clinical Transformation of Perioperative Precision Anesthesia, Jiaxing, China
| | - Ying Cai
- Department of Anesthesiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
- Key Laboratory of Basic Research and Clinical Transformation of Perioperative Precision Anesthesia, Jiaxing, China
| | - Xiaoyan Ye
- Jiaxing University Medical College, Jiaxing, China
| | - Hongmei Zhou
- Department of Anesthesiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
- Key Laboratory of Basic Research and Clinical Transformation of Perioperative Precision Anesthesia, Jiaxing, China
| | - Hejia Ge
- Department of Pediatrics, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| |
Collapse
|
15
|
Serafini RA, Ramakrishnan A, Shen L, Zachariou V. Desipramine induces anti-inflammatory dorsal root ganglion transcriptional signatures in the murine spared nerve injury model. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2024; 15:100153. [PMID: 38549875 PMCID: PMC10973649 DOI: 10.1016/j.ynpai.2024.100153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 03/19/2024] [Accepted: 03/20/2024] [Indexed: 05/09/2024]
Abstract
Monoamine-targeting antidepressants serve as frontline medications for chronic pain and associated comorbidities. While persistent anti-allodynic properties of antidepressants generally require weeks of treatment, several groups have demonstrated acute analgesic effects within hours of administration, suggesting a role in non-mesocorticolimbic pain processing regions such as the peripheral nervous system. To further explore this possibility, after four weeks of spared nerve injury or sham surgeries, we systemically administered desipramine or saline for an additional three weeks and performed whole transcriptome RNA sequencing on L3-6 dorsal root ganglia. Along with alterations in molecular pathways associated with neuronal activity, we observed a robust immunomodulatory transcriptional signature in the desipramine treated group. Cell subtype deconvolution predicted that these changes were associated with A- and C-fibers. Of note, differentially expressed genes from the dorsal root ganglia of DMI-treated, injured mice were largely unique compared to those from the nucleus accumbens of the same animals. These observations suggest that, under peripheral nerve injury conditions, desipramine induces specific gene expression changes across various regions of the nociceptive circuitry.
Collapse
Affiliation(s)
- Randal A. Serafini
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
- Department of Pharmacology, Physiology & Biophysics, Avedisian and Chobanian School of Medicine at Boston University, Boston, MA 02118, United States
| | - Aarthi Ramakrishnan
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Li Shen
- Department of Pharmacology, Physiology & Biophysics, Avedisian and Chobanian School of Medicine at Boston University, Boston, MA 02118, United States
| | - Venetia Zachariou
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
- Department of Pharmacology, Physiology & Biophysics, Avedisian and Chobanian School of Medicine at Boston University, Boston, MA 02118, United States
| |
Collapse
|
16
|
Tonello R, Davidson S, Berta T. Dissociation and Culture of Adult Mouse Satellite Glial Cells. Bio Protoc 2023; 13:e4906. [PMID: 38156033 PMCID: PMC10751239 DOI: 10.21769/bioprotoc.4906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 10/31/2023] [Accepted: 11/01/2023] [Indexed: 12/30/2023] Open
Abstract
Satellite glial cells (SGCs) are a type of glial cell population that originates from neural crest cells. They ultimately migrate to surround the cell bodies of neurons in the ganglia of the peripheral nervous system. Under physiological conditions, SGCs perform homeostatic functions by modifying the microenvironment around nearby neurons and provide nutrients, structure, and protection. In recent years, they have gained considerable attention due to their involvement in peripheral nerve regeneration and pain. Although methods for culturing neonatal or rat SGCs have long existed, a well-characterized method for dissociating and culturing adult SGCs from mouse tissues has been lacking until recently. This has impeded further studies of their function and the testing of new therapeutics. This protocol provides a detailed description of how to obtain primary cultures of adult SGCs from mouse dorsal root ganglia in approximately two weeks with over 90% cell purity. We also demonstrate cell purity of these cultures using quantitative real-time RT-PCR and their functional integrity using calcium imaging. Key features • Detailed and simplified protocol to dissociate and culture primary satellite glial cells (SGCs) from adult mice. • Cells are dissociated in approximately 2-3 h and cultured for approximately two weeks. • These SGC cultures allow both molecular and functional studies.
Collapse
Affiliation(s)
- Raquel Tonello
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati 45267, USA
- Pain Research Center, Department of Molecular Pathobiology, College of Dentistry, New York, USA
| | - Steve Davidson
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati 45267, USA
| | - Temugin Berta
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati 45267, USA
| |
Collapse
|
17
|
Li X, Prudente AS, Prato V, Guo X, Hao H, Jones F, Figoli S, Mullen P, Wang Y, Tonnello R, Lee SH, Shah S, Maffei B, Berta T, Du X, Gamper N. Peripheral gating of pain by glial endozepine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.20.567848. [PMID: 38045227 PMCID: PMC10690183 DOI: 10.1101/2023.11.20.567848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
We report that diazepam binding inhibitor (DBI) is a glial messenger mediating satellite glia-sensory neuron crosstalk in the dorsal root ganglion (DRG). DBI is highly and specifically expressed in satellite glia cells (SGCs) of mice, rat and human, but not in sensory neurons or other DRG-resident cells. Knockdown of DBI results in a robust mechanical hypersensitivity without significant effects on other sensory modalities. In vivo overexpression of DBI in SGCs reduces sensitivity to mechanical stimulation and alleviates mechanical allodynia in neuropathic and inflammatory pain models. We further show that DBI acts as a partial agonist and positive allosteric modulator at the neuronal GABAA receptors, particularly strongly effecting those with a high-affinity benzodiazepine binding site. Such receptors are selectively expressed by a subpopulation of mechanosensitive DRG neurons and these are also more enwrapped with DBI-expressing glia, as compared to other DRG neurons, suggesting a mechanism for specific effect of DBI on mechanosensation. These findings identified a new, peripheral neuron-glia communication mechanism modulating pain signalling, which can be targeted therapeutically.
Collapse
Affiliation(s)
- Xinmeng Li
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | - Arthur Silveira Prudente
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH, USA
| | - Vincenzo Prato
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, UK
| | - Xianchuan Guo
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | - Han Hao
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | - Frederick Jones
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, UK
- Department of Life Sciences, Manchester Metropolitan University, Manchester, UK
| | - Sofia Figoli
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, UK
| | - Pierce Mullen
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, UK
| | - Yujin Wang
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | - Raquel Tonnello
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH, USA
| | - Sang Hoon Lee
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH, USA
| | - Shihab Shah
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, UK
| | - Benito Maffei
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, UK
| | - Temugin Berta
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH, USA
| | - Xiaona Du
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | - Nikita Gamper
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, UK
| |
Collapse
|
18
|
Mecklenburg J, Shein SA, Malmir M, Hovhannisyan AH, Weldon K, Zou Y, Lai Z, Jin YF, Ruparel S, Tumanov AV, Akopian AN. Transcriptional profiles of non-neuronal and immune cells in mouse trigeminal ganglia. FRONTIERS IN PAIN RESEARCH 2023; 4:1274811. [PMID: 38028432 PMCID: PMC10644122 DOI: 10.3389/fpain.2023.1274811] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 09/29/2023] [Indexed: 12/01/2023] Open
Abstract
Non-neuronal cells constitute 90%-95% of sensory ganglia. These cells, especially glial and immune cells, play critical roles in the modulation of sensory neurons. This study aimed to identify, profile, and summarize the types of trigeminal ganglion (TG) non-neuronal cells in naïve male mice using published and our own data generated by single-cell RNA sequencing, flow cytometry, and immunohistochemistry. TG has five types of non-neuronal cells, namely, glial, fibroblasts, smooth muscle, endothelial, and immune cells. There is an agreement among publications for glial, fibroblasts, smooth muscle, and endothelial cells. Based on gene profiles, glial cells were classified as myelinated and non-myelinated Schwann cells and satellite glial cells. Mpz has dominant expression in Schwann cells, and Fabp7 is specific for SCG. Two types of Col1a2+ fibroblasts located throughout TG were distinguished. TG smooth muscle and endothelial cells in the blood vessels were detected using well-defined markers. Our study reported three types of macrophages (Mph) and four types of neutrophils (Neu) in TG. Mph were located in the neuronal bodies and nerve fibers and were sub-grouped by unique transcriptomic profiles with Ccr2, Cx3cr1, and Iba1 as markers. A comparison of databases showed that type 1 Mph is similar to choroid plexus-low (CPlo) border-associated Mph (BAMs). Type 2 Mph has the highest prediction score with CPhi BAMs, while type 3 Mph is distinct. S100a8+ Neu were located in the dura surrounding TG and were sub-grouped by clustering and expressions of Csf3r, Ly6G, Ngp, Elane, and Mpo. Integrative analysis of published datasets indicated that Neu-1, Neu-2, and Neu-3 are similar to the brain Neu-1 group, while Neu-4 has a resemblance to the monocyte-derived cells. Overall, the generated and summarized datasets on non-neuronal TG cells showed a unique composition of myeloid cell types in TG and could provide essential and fundamental information for studies on cell plasticity, interactomic networks between neurons and non-neuronal cells, and function during a variety of pain conditions in the head and neck regions.
Collapse
Affiliation(s)
- Jennifer Mecklenburg
- Department of Endodontics, School of Dentistry, The University of Texas Health Science Center at San Antonio (UTHSCSA), San Antonio, TX, United States
| | - Sergey A. Shein
- Microbiology, Immunology & Molecular Genetics Departments, School of Medicine, UTHSCSA, San Antonio, TX, United States
| | - Mostafa Malmir
- Department of Electrical and Computer Engineering, the University of Texas at San Antonio, San Antonio, TX, United States
| | - Anahit H. Hovhannisyan
- Department of Endodontics, School of Dentistry, The University of Texas Health Science Center at San Antonio (UTHSCSA), San Antonio, TX, United States
| | - Korri Weldon
- Molecular Medicine, School of Medicine, UTHSCSA, San Antonio, TX, United States
| | - Yi Zou
- Molecular Medicine, School of Medicine, UTHSCSA, San Antonio, TX, United States
| | - Zhao Lai
- Molecular Medicine, School of Medicine, UTHSCSA, San Antonio, TX, United States
- Greehey Children’s Cancer Research Institute, UTHSCSA, San Antonio, TX, United States
| | - Yu-Fang Jin
- Department of Electrical and Computer Engineering, the University of Texas at San Antonio, San Antonio, TX, United States
| | - Shivani Ruparel
- Department of Endodontics, School of Dentistry, The University of Texas Health Science Center at San Antonio (UTHSCSA), San Antonio, TX, United States
| | - Alexei V. Tumanov
- Microbiology, Immunology & Molecular Genetics Departments, School of Medicine, UTHSCSA, San Antonio, TX, United States
| | - Armen N. Akopian
- Department of Endodontics, School of Dentistry, The University of Texas Health Science Center at San Antonio (UTHSCSA), San Antonio, TX, United States
| |
Collapse
|