1
|
Chen Y, Liu F, Shi S, Xiao S, Gong X. The Integrated Transcriptome Bioinformatics Analysis of Energy Metabolism-Related Profiles for Dorsal Root Ganglion of Neuropathic Pain. Mol Neurobiol 2025; 62:4149-4171. [PMID: 39406937 DOI: 10.1007/s12035-024-04537-2] [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: 05/28/2024] [Accepted: 10/07/2024] [Indexed: 03/05/2025]
Abstract
Neuropathic pain (NP) is a debilitating disease and is associated with energy metabolism alterations. This study aimed to identify energy metabolism-related differentially expressed genes (EMRDEGs) in NP, construct a diagnostic model, and analyze immune cell infiltration and single-cell gene expression characteristics of NP. GSE89224, GSE123919, and GSE134003 were downloaded from the Gene Expression Omnibus. Differentially expressed genes (DEGs) analysis and an intersection with highly energy metabolism-related modules in weighted gene co-expression network analysis (WGCNA) was performed in GSE89224. Least absolute shrinkage and selection operator (LASSO), random forest, and logistic regression were used for model genes selection. NP samples were divided into high- and low-risk groups and different disease subtypes based on risk score of LASSO algorithm and consensus clustering analysis, respectively. Immune cell composition was estimated in different risk groups and NP subtypes. Datasets 134,003 were performed for identification of single-cell DEGs and functional enrichment. Cell-cell communications and pseudo-time analysis to reveal the expression profile of NP. A total of 38 EMRDEGs were obtained and are majorly enriched in metabolism about glioma and inflammation. LASSO, random forest, and logistic regression identified 6 model genes, which were Itpr1, Gng8, Socs3, Fscn1, Cckbr, and Camk1. The nomogram, based on six model genes, had a good predictive ability, concordance, and diagnostic value. The comparisons between different risk groups and NP subtypes identified important pathways and different immune cells component. The immune infiltration results majorly associated with inflammation and energy metabolism. Single-cell analysis revealed cell-cell communications and cells differentiation characteristics of NP. In conclusion, our results not only elucidate the involvement of energy metabolism in NP but also provides a robust diagnostic tool with six model genes. These findings might give insight into the pathogenesis of NP and provide effective therapeutic regimens for the treatment of NP.
Collapse
Affiliation(s)
- Yongmei Chen
- Department of Laboratory, Xiangyang Central Hospital, Affiliation of Hubei University of Art and Science, Xiangyang City, Hubei, China
| | - Fan Liu
- Institute of Neuroscience, Department of Anesthesiology, Xiangyang Central Hospital, Affiliation of Hubei University of Art and Science, No.136, Jingzhou Street, Xiangcheng District, Xiangyang City, 441000, Hubei, China
| | - Shengnan Shi
- Institute of Neuroscience, Department of Anesthesiology, Xiangyang Central Hospital, Affiliation of Hubei University of Art and Science, No.136, Jingzhou Street, Xiangcheng District, Xiangyang City, 441000, Hubei, China
| | - Shugen Xiao
- Institute of Neuroscience, Department of Anesthesiology, Xiangyang Central Hospital, Affiliation of Hubei University of Art and Science, No.136, Jingzhou Street, Xiangcheng District, Xiangyang City, 441000, Hubei, China
| | - Xingrui Gong
- Institute of Neuroscience, Department of Anesthesiology, Xiangyang Central Hospital, Affiliation of Hubei University of Art and Science, No.136, Jingzhou Street, Xiangcheng District, Xiangyang City, 441000, Hubei, China.
| |
Collapse
|
2
|
Fan CY, McAllister BB, Stokes-Heck S, Harding EK, Pereira de Vasconcelos A, Mah LK, Lima LV, van den Hoogen NJ, Rosen SF, Ham B, Zhang Z, Liu H, Zemp FJ, Burkhard R, Geuking MB, Mahoney DJ, Zamponi GW, Mogil JS, Ousman SS, Trang T. Divergent sex-specific pannexin-1 mechanisms in microglia and T cells underlie neuropathic pain. Neuron 2025; 113:896-911.e9. [PMID: 39892387 DOI: 10.1016/j.neuron.2025.01.005] [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: 05/08/2024] [Revised: 11/25/2024] [Accepted: 01/08/2025] [Indexed: 02/03/2025]
Abstract
Chronic pain is a leading cause of disability, affecting more women than men. Different immune cells contribute to this sexual divergence, but the mechanisms, especially in females, are not well defined. We show that pannexin-1 (Panx1) channels on microglia and T cells differentially cause mechanical allodynia, a debilitating symptom of neuropathic pain. In male rodents, Panx1 drives vascular endothelial growth factor-A (VEGF-A) release from microglia. Cell-specific knockdown of microglial Panx1 or pharmacological blockade of the VEGF receptor attenuated allodynia in nerve-injured males. In females, nerve injury increased spinal CD8+ T cells and leptin levels. Leptin release from female-derived CD8+ T cells was Panx1 dependent, and intrathecal leptin-neutralizing antibody injection sex-specifically reversed allodynia. Adoptive transfer of female-derived CD8+ T cells caused robust allodynia, which was prevented by a leptin-neutralizing antibody or leptin small interfering RNA (siRNA) knockdown. Panx1-targeted approaches may alleviate neuropathic pain in both sexes, while T cell- and leptin-directed treatments could have sex-dependent benefits for women.
Collapse
Affiliation(s)
- Churmy Y Fan
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Canada; Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
| | - Brendan B McAllister
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Canada; Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
| | - Sierra Stokes-Heck
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Canada; Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
| | - Erika K Harding
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Canada; Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada; Department of Clinical Neurosciences, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
| | - Aliny Pereira de Vasconcelos
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Canada; Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
| | - Laura K Mah
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute For Chronic Diseases, University of Calgary, Calgary, Canada
| | - Lucas V Lima
- Departments of Psychology and Anesthesia and Faculty of Dentistry, Alan Edwards Centre for Research on Pain, McGill University, Montreal, Canada
| | - Nynke J van den Hoogen
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Canada; Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
| | - Sarah F Rosen
- Departments of Psychology and Anesthesia and Faculty of Dentistry, Alan Edwards Centre for Research on Pain, McGill University, Montreal, Canada
| | - Boram Ham
- Departments of Psychology and Anesthesia and Faculty of Dentistry, Alan Edwards Centre for Research on Pain, McGill University, Montreal, Canada
| | - Zizhen Zhang
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
| | - Hongrui Liu
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Canada; Annie Charbonneau Cancer Institute, University of Calgary, Calgary, Canada
| | - Franz J Zemp
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Canada; Annie Charbonneau Cancer Institute, University of Calgary, Calgary, Canada; Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada
| | - Regula Burkhard
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute For Chronic Diseases, University of Calgary, Calgary, Canada
| | - Markus B Geuking
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute For Chronic Diseases, University of Calgary, Calgary, Canada
| | - Douglas J Mahoney
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute For Chronic Diseases, University of Calgary, Calgary, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Canada; Annie Charbonneau Cancer Institute, University of Calgary, Calgary, Canada; Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada
| | - Gerald W Zamponi
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada
| | - Jeffrey S Mogil
- Departments of Psychology and Anesthesia and Faculty of Dentistry, Alan Edwards Centre for Research on Pain, McGill University, Montreal, Canada
| | - Shalina S Ousman
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada; Department of Cell Biology & Anatomy, University of Calgary, Calgary, Canada
| | - Tuan Trang
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Canada; Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Canada.
| |
Collapse
|
3
|
Gao XX, Zhang XH, Yu JA. Trends and hotspots in burns-related pain research: A bibliometric analysis. Burns 2025; 51:107345. [PMID: 39793163 DOI: 10.1016/j.burns.2024.107345] [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: 09/27/2024] [Revised: 11/18/2024] [Accepted: 12/03/2024] [Indexed: 01/13/2025]
Abstract
OBJECTIVE The aim of this investigation was to conduct a thorough synthesis of the extant scholarly discourse and to delineate the prevailing global trends in the domain of burn pain, employing a bibliometric analysis. METHODS A bibliometric analysis was performed utilizing the Web of Science Core Collection database. Articles were selected based on titles or abstracts containing keywords associated with burns and pain. Both quantitative and qualitative methodologies were applied to examine the retrieved data, encompassing an analysis of publication trends, research themes, and collaboration networks. RESULTS The number of articles on this topic has been increasing, averaging an annual growth rate of 6.9 % from 1997 to 2023. Contributions have come from 645 institutions across 53 countries, resulting in 446 papers that span areas such as nursing, anesthesia, and immunology. Key journals include Burns, Journal of Burn Care & Research, and Pain. The United States has demonstrated a significant research output in this field, with active international collaboration, notably with Washington University leading in contributions. Patterson DR was the most prolific author in terms of published papers, while Choiniere M was the most frequently co-cited author. The focus of research has shifted from symptom management to exploring pain mechanisms. Current research priorities in burn pain include "quality of life," "music therapy," and "psychological state." Recent analysis has highlighted key areas in neuropathic pain mechanisms, novel analgesic therapies, and specific groups such as pediatric burn patients. Influential studies have advanced our understanding of pathophysiology, while psychological interventions and inflammation are increasingly receiving attention. Emerging topics include non-pharmacological interventions, psychological support, technology in pain assessment and management, quality of life, and personalized pain management. CONCLUSION Research on burn pain is advancing rapidly; however, collaboration among countries and institutions remains limited. Increased cooperation and communication across these entities could significantly advance the field in the future. Future research should prioritize placebo-controlled trials of targeted therapeutic drugs and innovative pain management approaches, with a strong emphasis on patient outcomes and quality of life.
Collapse
Affiliation(s)
- Xin-Xin Gao
- Department of Burn Surgery, The First Hospital of Jilin University, Chaoyang District, 1 Xinmin Street, Changchun City, Jilin Province 130061, China.
| | - Xiu-Hang Zhang
- Department of Burn Surgery, The First Hospital of Jilin University, Chaoyang District, 1 Xinmin Street, Changchun City, Jilin Province 130061, China.
| | - Jia-Ao Yu
- Department of Burn Surgery, The First Hospital of Jilin University, Chaoyang District, 1 Xinmin Street, Changchun City, Jilin Province 130061, China.
| |
Collapse
|
4
|
Gu J, Zhou Z, Xu S, Pan W, Wang J, Liu O, Wang S, Xu J. Topical Application of Nitrate Ameliorates Skin Fibrosis by Regulating ST2 +CD4 + T Cells in Systemic Sclerosis Mouse Model. J Invest Dermatol 2025; 145:346-358.e5. [PMID: 38945439 DOI: 10.1016/j.jid.2024.06.1273] [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: 01/06/2024] [Revised: 06/04/2024] [Accepted: 06/05/2024] [Indexed: 07/02/2024]
Abstract
Systemic sclerosis (SSc) is characterized by intractable multiorgan fibrosis caused by vascular and immune dysfunction. Currently, effective therapeutic options for patients with SSc are limited. Nitrate, an abundant nutrient in the diet, has been demonstrated to be preventative and therapeutic for several diseases. To determine whether nitrate can slow or reverse SSc progression, topical application of nitrate delivered by dissolving microneedles was used to treat a bleomycin-induced dermal fibrosis mouse model. In this study, nitrate considerably attenuated dermal thickness, stiffness, and collagen deposition. Bulk RNA sequencing of skin revealed that Cd4 was a key hub gene in SSc nitrate therapy. In addition, bleomycin-induced cytokines and chemokines were inhibited by nitrate, and CD4+ T cells infiltration markedly declined. Il4, Il6, Il13, and Tgfb expressions in CD4+ T cells isolated from skin biopsies also significantly decreased. Mechanistically, Il1rl1, a type 2 immune response inducer, was markedly repressed in isolated CD4+ T cells and dermal tissues after nitrate treatment. Remarkably, compared with wild-type mice, mice lacking Il1rl1 showed impaired transcriptional profiles after intradermal bleomycin injection. Adoptive transfer of ST2+CD4+ T cells promoted bleomycin-induced Rag2-/- mice dermal fibrosis. Collectively, these findings demonstrate that nitrate targeting ST2+CD4+ T cells is an effective therapeutic option for SSc.
Collapse
Affiliation(s)
- Jianyu Gu
- Salivary Gland Disease Center, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Laboratory of Oral Health and Beijing Stomatological Hospital, Capital Medical University, Beijing, China; Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Zekun Zhou
- Hunan Key Laboratory of Oral Health Research, Hunan 3D Printing Engineering Research Center of Oral Care, Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, China
| | - Shihan Xu
- Salivary Gland Disease Center, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Laboratory of Oral Health and Beijing Stomatological Hospital, Capital Medical University, Beijing, China; Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Wen Pan
- Salivary Gland Disease Center, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Laboratory of Oral Health and Beijing Stomatological Hospital, Capital Medical University, Beijing, China; Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Jinsong Wang
- Salivary Gland Disease Center, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Laboratory of Oral Health and Beijing Stomatological Hospital, Capital Medical University, Beijing, China; Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Ousheng Liu
- Hunan Key Laboratory of Oral Health Research, Hunan 3D Printing Engineering Research Center of Oral Care, Academician Workstation for Oral-maxilofacial and Regenerative Medicine, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, Xiangya School of Stomatology, Central South University, Changsha, China
| | - Songlin Wang
- Salivary Gland Disease Center, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Laboratory of Oral Health and Beijing Stomatological Hospital, Capital Medical University, Beijing, China; Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China; Immunology Research Centre for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing, China; Laboratory of Homeostatic Medicine, School of Medicine, Southern University of Science and Technology, Shenzhen, China; Research Units of Tooth Development and Regeneration, Chinese Academy of Medical Sciences, Beijing, China.
| | - Junji Xu
- Salivary Gland Disease Center, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Laboratory of Oral Health and Beijing Stomatological Hospital, Capital Medical University, Beijing, China; Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China; Immunology Research Centre for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing, China; Department of Periodontics, Beijing Stomatological Hospital, Capital Medical University School of Stomatology, Beijing, China.
| |
Collapse
|
5
|
Li N, Li M, Jin S, Yu J, Wei H, Wang W, Ma S, Jiang Y, Liu Q, Yao H. Animal models of cisplatin-induced neuropathic pain. Animal Model Exp Med 2025. [PMID: 39854055 DOI: 10.1002/ame2.12548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 12/18/2024] [Indexed: 01/26/2025] Open
Abstract
Cisplatin chemotherapy has been used as the main treatment for different types of cancer. However, cisplatin chemotherapy-induced peripheral neuropathic pain (CIPNP) seriously affects the treatment process and quality of life of patients. In addition, it impacts the underlying mechanism and prevention and treatment strategies, indicating that drug selection and efficacy evaluation need to be further investigated. Furthermore, an animal model that is more consistent with the pathological mechanism needs to be developed. In this study, we describe and discuss the methods of developing and detecting CIPNP models in rats and mice induced by cisplatin chemotherapy. The aim was to improve the modeling rate and develop animal models that are more consistent with the developmental pattern of the disease. In addition, the study provides ideal reference animal models for clinical research and drug discovery and development.
Collapse
Affiliation(s)
- Ningxin Li
- China Medical University, Shenyang, China
| | - Mingzhu Li
- Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Sheng Yang, China
| | - Shengbo Jin
- Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Jun Yu
- Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | | | - Wenping Wang
- Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Sheng Yang, China
| | - Siyao Ma
- Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Sheng Yang, China
| | - Yuxin Jiang
- Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Qian Liu
- Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Huini Yao
- China Medical University, Shenyang, China
| |
Collapse
|
6
|
Inyang KE, Sim J, Clark KB, Geron M, Monahan K, Evans C, O'Connell P, Laumet S, Peng B, Ma J, Heijnen CJ, Dantzer R, Scherrer G, Kavelaars A, Bernard M, Aldhamen YA, Folger JK, Bavencoffe A, Laumet G. Upregulation of delta opioid receptor by meningeal interleukin-10 prevents relapsing pain. Brain Behav Immun 2025; 123:399-410. [PMID: 39349285 PMCID: PMC11624093 DOI: 10.1016/j.bbi.2024.09.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 09/23/2024] [Accepted: 09/27/2024] [Indexed: 10/02/2024] Open
Abstract
Chronic pain often includes periods of transient amelioration and even remission that alternate with severe relapsing pain. While most research on chronic pain has focused on pain development and maintenance, there is a critical unmet need to better understand the mechanisms that underlie pain remission and relapse. We found that interleukin (IL)-10, a pain resolving cytokine, is produced by resident macrophages in the spinal meninges during remission from pain and signaled to IL-10 receptor-expressing sensory neurons. Using unbiased RNA-sequencing, we identified that IL-10 upregulated expression and antinociceptive activity of δ-opioid receptor (δOR) in the dorsal root ganglion. Genetic or pharmacological inhibition of either IL-10 signaling or δOR triggered relapsing pain. Overall, our findings, from electrophysiology, genetic manipulation, flow cytometry, pharmacology, and behavioral approaches, indicate that remission of pain is not simply a return to the naïve state. Instead, remission is an adapted homeostatic state associated with lasting pain vulnerability resulting from persisting neuroimmune interactions within the nociceptive system. Broadly, this sheds light on the elusive mechanisms underlying recurrence a common aspect across various chronic pain conditions.
Collapse
Affiliation(s)
| | - Jaewon Sim
- Department of Physiology, Michigan State University, East Lansing, MI, USA; Cell and Molecular Biology Graduate Program, Michigan State University, East Lansing, MI, USA
| | - Kimberly B Clark
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Matan Geron
- Department of Cell Biology and Physiology, Department of Pharmacology, UNC Neuroscience Center, University of North Carolina, Chapel Hill, NC, USA
| | - Karli Monahan
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Christine Evans
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Patrick O'Connell
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, USA
| | - Sophie Laumet
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Bo Peng
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jiacheng Ma
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Cobi J Heijnen
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Robert Dantzer
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Grégory Scherrer
- Department of Cell Biology and Physiology, Department of Pharmacology, UNC Neuroscience Center, University of North Carolina, Chapel Hill, NC, USA; New York Stem Cell Foundation - Robertson Investigator, University of North Carolina, Chapel Hill, NC, USA
| | - Annemieke Kavelaars
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Matthew Bernard
- Flow Cytometry Core, Michigan State University, East Lansing, MI, USA
| | - Yasser A Aldhamen
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, USA
| | - Joseph K Folger
- Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - Alexis Bavencoffe
- Department of Integrative Biology and Pharmacology, McGovern Medical School at UTHealth, Houston, TX, USA
| | - Geoffroy Laumet
- Department of Physiology, Michigan State University, East Lansing, MI, USA; Cell and Molecular Biology Graduate Program, Michigan State University, East Lansing, MI, USA; Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
7
|
Hakim S, Jain A, Woolf CJ. Immune drivers of pain resolution and protection. Nat Immunol 2024; 25:2200-2208. [PMID: 39528810 DOI: 10.1038/s41590-024-02002-9] [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: 07/24/2024] [Accepted: 09/23/2024] [Indexed: 11/16/2024]
Abstract
Immune cells are involved in the pathogenesis of pain by directly activating or sensitizing nociceptor sensory neurons. However, because the immune system also has the capacity to self-regulate through anti-inflammatory mechanisms that drive the resolution of inflammation, it might promote pain resolution and prevention. Here, we describe how immune cell-derived cytokines can act directly on sensory neurons to inhibit pain hypersensitivity and how immune-derived endogenous opioids promote analgesia. We also discuss how immune cells support healthy tissue innervation by clearing debris after nerve injury, protecting against axon retraction from target tissues and enhancing regeneration, preventing the development of chronic neuropathic pain. Finally, we review the accumulating evidence that manipulating immune activity positively alters somatosensation, albeit with currently unclear molecular and cellular mechanisms. Exploration of immune-mediated analgesia and pain prevention could, therefore, be important for the development of novel immune therapies for the treatment of clinical pain states.
Collapse
Affiliation(s)
- Sara Hakim
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Aakanksha Jain
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
| | - Clifford J Woolf
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA.
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
8
|
Pan H, Liu CX, Zhu HJ, Zhang GF. Immune cells mediate the effects of gut microbiota on neuropathic pain: a Mendelian randomization study. J Headache Pain 2024; 25:196. [PMID: 39528917 PMCID: PMC11555962 DOI: 10.1186/s10194-024-01906-z] [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: 07/18/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND The gut microbiota may be involved in neuropathic pain. However, the causal association between gut microbiota and neuropathic pain remains unclear. Whether immune cells and inflammatory factors mediate the pathway from gut microbiota to neuropathic pain has not been elucidated. METHODS We obtained the summary data of 412 gut microbiota, 731 immune cells, 91 inflammatory factors, and five types of neuropathic pain (drug-induced neuropathy, postherpetic neuralgia, sciatica, trigeminal neuralgia, and unspecified neuralgia) from large-scale genome-wide association study (GWAS) datasets and the FinnGen database. We used bidirectional Mendelian randomization (MR) analysis to explore the causal association between gut microbiota and neuropathic pain. Additionally, we conducted a mediation analysis to identify whether immune cells and inflammatory factors act as mediators within these causal relationships. RESULTS Our study revealed 30 causal relationships between 26 gut bacterial taxa and five types of neuropathic pain, including four associated with drug-induced neuropathy, six with postherpetic neuralgia, five with sciatica, eight with trigeminal neuralgia, and seven with unspecified neuralgia. Moreover, we identified 35 gut bacterial pathway abundances causally involved in neuropathic pain. The reverse MR analysis showed no evidence of reverse causality from gut microbiota to neuropathic pain. Mediation analysis demonstrated that the immune cell phenotype "HLA-DR++ monocyte % leukocyte" mediated the causal relationship between p_Proteobacteria and sciatica with a mediation proportion of 36.15% (P = 0.038), whereas "CD11c on CD62L+ myeloid dendritic cell" mediated the causal pathway from assimilatory sulfate reduction to trigeminal neuralgia with a mediation proportion of 27.90% (P = 0.041). CONCLUSION This study identified the causal relationships between several specific gut microbiota and various neuropathic pain subtypes. Additionally, two immune cells may act as potential mediators in the pathways from gut microbiota to neuropathic pain.
Collapse
Affiliation(s)
- Hao Pan
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, PR China
| | - Cheng-Xiao Liu
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, PR China
| | - Hui-Juan Zhu
- Department of Anesthesiology, Jinan Seventh People's Hospital, Jinan, 250021, Shandong, PR China
| | - Guang-Fen Zhang
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, PR China.
| |
Collapse
|
9
|
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
|
10
|
Song Q, E S, Zhang Z, Liang Y. Neuroplasticity in the transition from acute to chronic pain. Neurotherapeutics 2024; 21:e00464. [PMID: 39438166 PMCID: PMC11585895 DOI: 10.1016/j.neurot.2024.e00464] [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: 06/21/2024] [Revised: 09/10/2024] [Accepted: 09/27/2024] [Indexed: 10/25/2024] Open
Abstract
Acute pain is a transient sensation that typically serves as part of the body's defense mechanism. However, in certain patients, acute pain can evolve into chronic pain, which persists for months or even longer. Neuroplasticity refers to the capacity for variation and adaptive alterations in the morphology and functionality of neurons and synapses, and it plays a significant role in the transmission and modulation of pain. In this paper, we explore the molecular mechanisms and signaling pathways underlying neuroplasticity during the transition of pain. We also examine the effects of neurotransmitters, inflammatory mediators, and central sensitization on neuroplasticity, as well as the potential of neuroplasticity as a therapeutic strategy for preventing chronic pain. The aims of this article is to clarify the role of neuroplasticity in the transformation from acute pain to chronic pain, with the hope of providing a novel theoretical basis for unraveling the pathogenesis of chronic pain and offering more effective strategies and approaches for its diagnosis and treatment.
Collapse
Affiliation(s)
- Qingbiao Song
- School of Anesthesiology, Shandong Second Medical University, Weifang 261053, China
| | - Sihan E
- School of Anesthesiology, Shandong Second Medical University, Weifang 261053, China
| | - Zhiyu Zhang
- Department of Orthopedics, Affiliated Hospital of Shandong Second Medical University, Weifang 261035, China
| | - Yingxia Liang
- School of Anesthesiology, Shandong Second Medical University, Weifang 261053, China.
| |
Collapse
|
11
|
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
|
12
|
Jain A, Hakim S, Woolf CJ. Immune drivers of physiological and pathological pain. J Exp Med 2024; 221:e20221687. [PMID: 38607420 PMCID: PMC11010323 DOI: 10.1084/jem.20221687] [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: 05/19/2023] [Revised: 09/25/2023] [Accepted: 04/02/2024] [Indexed: 04/13/2024] Open
Abstract
Physiological pain serves as a warning of exposure to danger and prompts us to withdraw from noxious stimuli to prevent tissue damage. Pain can also alert us of an infection or organ dysfunction and aids in locating such malfunction. However, there are instances where pain is purely pathological, such as unresolved pain following an inflammation or injury to the nervous system, and this can be debilitating and persistent. We now appreciate that immune cells are integral to both physiological and pathological pain, and that pain, in consequence, is not strictly a neuronal phenomenon. Here, we discuss recent findings on how immune cells in the skin, nerve, dorsal root ganglia, and spinal cord interact with somatosensory neurons to mediate pain. We also discuss how both innate and adaptive immune cells, by releasing various ligands and mediators, contribute to the initiation, modulation, persistence, or resolution of various modalities of pain. Finally, we propose that the neuroimmune axis is an attractive target for pain treatment, but the challenges in objectively quantifying pain preclinically, variable sex differences in pain presentation, as well as adverse outcomes associated with immune system modulation, all need to be considered in the development of immunotherapies against pain.
Collapse
Affiliation(s)
- Aakanksha Jain
- F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA, USA
| | - Sara Hakim
- F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Clifford J. Woolf
- F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
13
|
Ollodart J, Steele LR, Romero-Sandoval EA, Strowd RE, Shiozawa Y. Contributions of neuroimmune interactions to chemotherapy-induced peripheral neuropathy development and its prevention/therapy. Biochem Pharmacol 2024; 222:116070. [PMID: 38387528 PMCID: PMC10964384 DOI: 10.1016/j.bcp.2024.116070] [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: 09/06/2023] [Revised: 12/04/2023] [Accepted: 02/19/2024] [Indexed: 02/24/2024]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a debilitating sequela that is difficult for both clinicians and cancer patients to manage. Precise mechanisms of CIPN remain elusive and current clinically prescribed therapies for CIPN have limited efficacy. Recent studies have begun investigating the interactions between the peripheral and central nervous systems and the immune system. Understanding these neuroimmune interactions may shift the paradigm of elucidating CIPN mechanisms. Although the contribution of immune cells to CIPN pathogenesis represents a promising area of research, its fully defined mechanisms have not yet been established. Therefore, in this review, we will discuss (i) current shortcoming of CIPN treatments, (ii) the roles of neuroimmune interactions in CIPN development and (iii) potential neuroimmune interaction-targeting treatment strategies for CIPN. Interestingly, monocytes/macrophages in dorsal root ganglia; microglia and astrocytes in spinal cord; mast cells in skin; and Schwann cell near peripheral nerves have been identified as inducers of CIPN behaviors, whereas T cells have been found to contribute to CIPN resolution. Additionally, nerve-resident immune cells have been targeted as prevention and/or therapy for CIPN using traditional herbal medicines, small molecule inhibitors, and intravenous immunoglobulins in a preclinical setting. Overall, unveiling neuroimmune interactions associated with CIPN may ultimately reduce cancer mortality and improve cancer patients' quality of life.
Collapse
Affiliation(s)
- Jenna Ollodart
- Department of Cancer Biology, Wake Forest University School of Medicine, and Atrium Health Wake Forest Baptist Comprehensive Cancer, Winston-Salem, NC, USA
| | - Laiton R Steele
- Department of Cancer Biology, Wake Forest University School of Medicine, and Atrium Health Wake Forest Baptist Comprehensive Cancer, Winston-Salem, NC, USA
| | | | - Roy E Strowd
- Department of Cancer Biology, Wake Forest University School of Medicine, and Atrium Health Wake Forest Baptist Comprehensive Cancer, Winston-Salem, NC, USA
| | - Yusuke Shiozawa
- Department of Cancer Biology, Wake Forest University School of Medicine, and Atrium Health Wake Forest Baptist Comprehensive Cancer, Winston-Salem, NC, USA.
| |
Collapse
|
14
|
Reel JM, Abbadi J, Cox MA. T cells at the interface of neuroimmune communication. J Allergy Clin Immunol 2024; 153:894-903. [PMID: 37952833 PMCID: PMC10999355 DOI: 10.1016/j.jaci.2023.10.026] [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: 09/19/2023] [Revised: 10/12/2023] [Accepted: 10/24/2023] [Indexed: 11/14/2023]
Abstract
The immune system protects the host from infection and works to heal damaged tissue after infection or injury. There is increasing evidence that the immune system and the nervous system work in concert to achieve these goals. The sensory nervous system senses injury, infection, and inflammation, which results in a direct pain signal. Direct activation of peripheral sensory nerves can drive an inflammatory response in the skin. Immune cells express receptors for numerous transmitters released from sensory and autonomic nerves, which allows the nervous system to communicate directly with the immune system. This communication is bidirectional because immune cells can also produce neurotransmitters. Both innate and adaptive immune cells respond to neuronal signaling, but T cells appear to be at the helm of neuroimmune communication.
Collapse
Affiliation(s)
- Jessica M Reel
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Okla
| | - Jumana Abbadi
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Okla
| | - Maureen A Cox
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Okla; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Okla.
| |
Collapse
|
15
|
Matsuzaka Y, Yashiro R. Unraveling the Immunopathogenesis of Multiple Sclerosis: The Dynamic Dance of Plasmablasts and Pathogenic T Cells. BIOLOGICS 2023; 3:232-252. [DOI: 10.3390/biologics3030013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease of the central nervous system, characterized by multiple lesions occurring temporally and spatially. Additionally, MS is a disease that predominates in the white population. In recent years, there has been a rapid increase in the number of patients, and it often occurs in young people, with an average age of onset of around 30 years old, but it can also occur in children and the elderly. It is more common in women than men, with a male-to-female ratio of approximately 1:3. As the immunopathogenesis of MS, a group of B cells called plasmablasts controls encephalomyelitis via IL-10 production. These IL-10-producing B cells, called regulatory B cells, suppress inflammatory responses in experimental mouse models of autoimmune diseases including MS. Since it has been clarified that these regulatory B cells are plasmablasts, it is expected that the artificial control of plasmablast differentiation will lead to the development of new treatments for MS. Among CD8-positive T cells in the peripheral blood, the proportion of PD-1-positive cells is decreased in MS patients compared with healthy controls. The dysfunction of inhibitory receptors expressed on T cells is known to be the core of MS immunopathology and may be the cause of chronic persistent inflammation. The PD-1+ CD8+ T cells may also serve as indicators that reflect the condition of each patient in other immunological neurological diseases such as MS. Th17 cells also regulate the development of various autoimmune diseases, including MS. Thus, the restoration of weakened immune regulatory functions may be a true disease-modifying treatment. So far, steroids and immunosuppressants have been the mainstream for autoimmune diseases, but the problem is that this kills not only pathogenic T cells, but also lymphocytes, which are necessary for the body. From this understanding of the immune regulation of MS, we can expect the development of therapeutic strategies that target only pathogenic immune cells.
Collapse
Affiliation(s)
- Yasunari Matsuzaka
- Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Ryu Yashiro
- Department of Mycobacteriology, Leprosy Research Center, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| |
Collapse
|
16
|
Zhao J, Huh Y, Bortsov A, Diatchenko L, Ji RR. Immunotherapies in chronic pain through modulation of neuroimmune interactions. Pharmacol Ther 2023; 248:108476. [PMID: 37307899 PMCID: PMC10527194 DOI: 10.1016/j.pharmthera.2023.108476] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/18/2023] [Accepted: 06/06/2023] [Indexed: 06/14/2023]
Abstract
It is generally believed that immune activation can elicit pain through production of inflammatory mediators that can activate nociceptive sensory neurons. Emerging evidence suggests that immune activation may also contribute to the resolution of pain by producing distinct pro-resolution/anti-inflammatory mediators. Recent research into the connection between the immune and nervous systems has opened new avenues for immunotherapy in pain management. This review provides an overview of the most utilized forms of immunotherapies (e.g., biologics) and highlight their potential for immune and neuronal modulation in chronic pain. Specifically, we discuss pain-related immunotherapy mechanisms that target inflammatory cytokine pathways, the PD-L1/PD-1 pathway, and the cGAS/STING pathway. This review also highlights cell-based immunotherapies targeting macrophages, T cells, neutrophils and mesenchymal stromal cells for chronic pain management.
Collapse
Affiliation(s)
- Junli Zhao
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Yul Huh
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA; Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Andrey Bortsov
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Luda Diatchenko
- Alan Edwards Centre for Research on Pain, McGill University, Montréal, QC H3A 0G4, Canada; Faculty of Dental Medicine and Oral Health Sciences, Department of Anesthesia, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC H3A 0G4, Canada
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA; Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA; Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
17
|
Sivanesan E, Sanchez KR, Zhang C, He SQ, Linderoth B, Stephens KE, Raja SN, Guan Y. Spinal Cord Stimulation Increases Chemoefficacy and Prevents Paclitaxel-Induced Pain via CX3CL1. Neuromodulation 2023; 26:938-949. [PMID: 37045646 PMCID: PMC10330336 DOI: 10.1016/j.neurom.2023.03.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 02/19/2023] [Accepted: 03/13/2023] [Indexed: 04/14/2023]
Abstract
INTRODUCTION Despite increasing utilization of spinal cord stimulation (SCS), its effects on chemoefficacy, cancer progression, and chemotherapy-induced peripheral neuropathy (CIPN) pain remain unclear. Up to 30% of adults who are cancer survivors may suffer from CIPN, and there are currently no effective preventative treatments. MATERIALS AND METHODS Through a combination of bioluminescent imaging, behavioral, biochemical, and immunohistochemical approaches, we investigated the role of SCS and paclitaxel (PTX) on tumor growth and PTX-induced peripheral neuropathy (PIPN) pain development in T-cell-deficient male rats (Crl:NIH-Foxn1rnu) with xenograft human non-small cell lung cancer. We hypothesized that SCS can prevent CIPN pain and enhance chemoefficacy partially by modulating macrophages, fractalkine (CX3CL1), and inflammatory cytokines. RESULTS We show that preemptive SCS enhanced the antitumor efficacy of PTX and prevented PIPN pain. Without SCS, rats with and without tumors developed robust PIPN pain-related mechanical hypersensitivity, but only those with tumors developed cold hypersensitivity, suggesting T-cell dependence for different PIPN pain modalities. SCS increased soluble CX3CL1 and macrophages and decreased neuronal and nonneuronal insoluble CX3CL1 expression and inflammation in dorsal root ganglia. CONCLUSION Collectively, our findings suggest that preemptive SCS is a promising strategy to increase chemoefficacy and prevent PIPN pain via CX3CL1-macrophage modulation.
Collapse
Affiliation(s)
- Eellan Sivanesan
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA.
| | - Karla R Sanchez
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Chi Zhang
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Shao-Qiu He
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Bengt Linderoth
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Kimberly E Stephens
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Arkansas Children's Research Institute, Little Rock, AR, USA
| | - Srinivasa N Raja
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Yun Guan
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA; Department of Neurological Surgery, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
18
|
Scott KS, Chelette B, Chidomere C, Phillip West A, Dantzer R. Cisplatin decreases voluntary wheel-running activity but does not impair food-motivated behavior in mice. Brain Behav Immun 2023; 111:169-176. [PMID: 37076053 PMCID: PMC10330347 DOI: 10.1016/j.bbi.2023.04.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 04/12/2023] [Accepted: 04/13/2023] [Indexed: 04/21/2023] Open
Abstract
Cisplatin is a chemotherapeutic agent that is still commonly used to treat solid tumors. However, it has several toxic side effects due in large part to the mitochondrial damage that it induces. As this mitochondrial damage is likely to result in a decrease in the amount of metabolic energy that is available for behavioral activities, it is not surprising that fatigue develops in cancer patients treated with cisplatin. The present preclinical study was initiated to determine whether the detrimental effects of cisplatin were more pronounced on physical effort requiring a lot of energy versus effort that not only requires less energy but also procures energy in the form of food. For this purpose, mice were either trained to run in a wheel or to work for food in various schedules of food reinforcement before being treated with cisplatin. The experiments were carried out only in male mice as we had already reported that sex differences in cisplatin-induced neurotoxicities are minimal. Cisplatin was administered daily for one cycle of five days, or two cycles separated by a five-day rest. As observed in previous experiments, cisplatin drastically reduced voluntary wheel running. In contrast, when cisplatin was administered to food-restricted mice trained to work for a food reward in a progressive ratio schedule or in a fixed-interval schedule, it tended to increase the number of responses emitted to obtain the food rewards. This increase was not associated with any change in the temporal distribution of responses during the interval between two reinforcements in mice submitted to the fixed interval schedule of food reinforcement. When cisplatin was administered to food-restricted mice trained in an effort-based decision-making task in which they had to choose between working for a grain pellet with little effort and working for a preferred chocolate pellet with more effort, it decreased the total number of responses emitted to obtain food rewards. However, this effect was much less marked than the decrease in wheel running induced by cisplatin. The decrease in the effort invested in the procurement of food rewards was not associated with any change in the relative distribution of effort between low reward and high reward during the time course of the test session. These findings show that cisplatin decreases energy-consuming activities but not energy-procuring activities unless they require a choice between options differing in their cost-benefit ratio. Furthermore, they indicate that the physical dimension of fatigue is more likely to develop in cisplatin-treated individuals than the motivational dimension of fatigue.
Collapse
Affiliation(s)
- Kiersten S Scott
- University of Texas MD Anderson Cancer Center, Department of Symptom Research, Houston, TX 77030, USA
| | - Brandon Chelette
- University of Texas MD Anderson Cancer Center, Department of Symptom Research, Houston, TX 77030, USA
| | - Chinenye Chidomere
- University of Texas MD Anderson Cancer Center, Department of Symptom Research, Houston, TX 77030, USA
| | - A Phillip West
- Department of Microbial Pathogenesis and Immunology, School of Medicine, Texas A&M University, Bryan, TX 77807, USA
| | - Robert Dantzer
- University of Texas MD Anderson Cancer Center, Department of Symptom Research, Houston, TX 77030, USA.
| |
Collapse
|
19
|
Inyang KE, Sim J, Clark KB, Matan G, Monahan K, Evans C, Beng P, Ma JV, Heijnen CJ, Dantzer R, Scherrer G, Kavelaars A, Bernard M, Aldhamen Y, Folger JK, Laumet G. Tonic Meningeal Interleukin-10 Upregulates Delta Opioid Receptor to Prevent Relapse to Pain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.08.544200. [PMID: 37333074 PMCID: PMC10274865 DOI: 10.1101/2023.06.08.544200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Chronic pain often alternates between transient remission and relapse of severe pain. While most research on chronic pain has focused on mechanisms maintaining pain, there is a critical unmet need to understand what prevents pain from re-emerging in those who recover from acute pain. We found that interleukin (IL)-10, a pain resolving cytokine, is persistently produced by resident macrophages in the spinal meninges during remission from pain. IL-10 upregulated expression and analgesic activity of δ-opioid receptor (δOR) in the dorsal root ganglion. Genetic or pharmacological inhibition of IL-10 signaling or δOR triggered relapse to pain in both sexes. These data challenge the widespread assumption that remission of pain is simply a return to the naïve state before pain was induced. Instead, our findings strongly suggest a novel concept that: remission is a state of lasting pain vulnerability that results from a long-lasting neuroimmune interactions in the nociceptive system.
Collapse
|
20
|
Scott K, Boukelmoune N, Taniguchi C, West AP, Heijnen CJ, Dantzer R. Resolution of cisplatin-induced fatigue does not require endogenous interleukin-10 in male mice. Behav Brain Res 2023; 444:114381. [PMID: 36870396 PMCID: PMC10029095 DOI: 10.1016/j.bbr.2023.114381] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 03/01/2023] [Accepted: 03/02/2023] [Indexed: 03/06/2023]
Abstract
Based on previous results showing a pivotal role of endogenous interleukin-10 (IL-10) in the recovery from cisplatin-induced peripheral neuropathy, the present experiments were carried out to determine whether this cytokine plays any role in the recovery from cisplatin-induced fatigue in male mice. Fatigue was measured by decreased voluntary wheel running in mice trained to run in a wheel in response to cisplatin. Mice were treated with a monoclonal neutralizing antibody (IL-10na) administered intranasally during the recovery period to neutralize endogenous IL-10. In the first experiment, mice were treated with cisplatin (2.83 mg/kg/day) for five days and IL-10na (12 μg/day for three days) five days later. In the second experiment, they were treated with cisplatin (2.3 mg/kg/day for 5 days twice at a five-day interval) and IL10na (12 μg/day for three days) immediately after the last injection of cisplatin. In both experiments, cisplatin decreased body weight and reduced voluntary wheel running. However, IL-10na did not impair recovery from these effects. These results show that the recovery from the cisplatin-induced decrease in wheel running does not require endogenous IL-10 in contrast to the recovery from cisplatin-induced peripheral neuropathy.
Collapse
Affiliation(s)
- Kiersten Scott
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Nabila Boukelmoune
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Cullen Taniguchi
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - A Phillip West
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M University, Bryan, TX 77087, United States
| | - Cobi J Heijnen
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Robert Dantzer
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
21
|
Kinin B1 and B2 Receptors Contribute to Cisplatin-Induced Painful Peripheral Neuropathy in Male Mice. Pharmaceutics 2023; 15:pharmaceutics15030852. [PMID: 36986713 PMCID: PMC10051506 DOI: 10.3390/pharmaceutics15030852] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 03/02/2023] [Accepted: 03/03/2023] [Indexed: 03/08/2023] Open
Abstract
Cisplatin is the preferential chemotherapeutic drug for highly prevalent solid tumours. However, its clinical efficacy is frequently limited due to neurotoxic effects such as peripheral neuropathy. Chemotherapy-induced peripheral neuropathy is a dose-dependent adverse condition that negatively impacts quality of life, and it may determine dosage limitations or even cancer treatment cessation. Thus, it is urgently necessary to identify pathophysiological mechanisms underlying these painful symptoms. As kinins and their B1 and B2 receptors contribute to the development of chronic painful conditions, including those induced by chemotherapy, the contribution of these receptors to cisplatin-induced peripheral neuropathy was evaluated via pharmacological antagonism and genetic manipulation in male Swiss mice. Cisplatin causes painful symptoms and impaired working and spatial memory. Kinin B1 (DALBK) and B2 (Icatibant) receptor antagonists attenuated some painful parameters. Local administration of kinin B1 and B2 receptor agonists (in sub-nociceptive doses) intensified the cisplatin-induced mechanical nociception attenuated by DALBK and Icatibant, respectively. In addition, antisense oligonucleotides to kinin B1 and B2 receptors reduced cisplatin-induced mechanical allodynia. Thus, kinin B1 and B2 receptors appear to be potential targets for the treatment of cisplatin-induced painful symptoms and may improve patients’ adherence to treatment and their quality of life.
Collapse
|
22
|
Abstract
Interactions between the immune and nervous systems are of central importance in neuropathic pain, a common and debilitating form of chronic pain caused by a lesion or disease affecting the somatosensory system. Our understanding of neuroimmune interactions in pain research has advanced considerably. Initially considered as passive bystanders, then as culprits in the pathogenesis of neuropathic pain, immune responses in the nervous system are now established to underpin not only the initiation and progression of pain but also its resolution. Indeed, immune cells and their mediators are well-established promoters of neuroinflammation at each level of the neural pain pathway that contributes to pain hypersensitivity. However, emerging evidence indicates that specific subtypes of immune cells (including antinociceptive macrophages, pain-resolving microglia and T regulatory cells) as well as immunoresolvent molecules and modulators of the gut microbiota-immune system axis can reduce the pain experience and contribute to the resolution of neuropathic pain. This Review provides an overview of the immune mechanisms responsible for the resolution of neuropathic pain, including those involved in innate, adaptive and meningeal immunity as well as interactions with the gut microbiome. Specialized pro-resolving mediators and therapeutic approaches that target these neuroimmune mechanisms are also discussed.
Collapse
|
23
|
Sankaranarayanan I, Tavares-Ferreira D, Mwirigi JM, Mejia GL, Burton MD, Price TJ. Inducible co-stimulatory molecule (ICOS) alleviates paclitaxel-induced neuropathic pain via an IL-10-mediated mechanism in female mice. J Neuroinflammation 2023; 20:32. [PMID: 36774519 PMCID: PMC9922469 DOI: 10.1186/s12974-023-02719-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 02/03/2023] [Indexed: 02/12/2023] Open
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a primary dose-limiting side effect caused by antineoplastic agents, such as paclitaxel. A primary symptom of this neuropathy is pain. Currently, there are no effective treatments for CIPN, which can lead to long-term morbidity in cancer patients and survivors. Neuro-immune interactions occur in CIPN pain and have been implicated both in the development and progression of pain in CIPN and the resolution of pain in CIPN. We investigated the potential role of inducible co-stimulatory molecule (ICOS) in the resolution of CIPN pain-like behaviors in mice. ICOS is an immune checkpoint molecule that is expressed on the surface of activated T cells and promotes proliferation and differentiation of T cells. We found that intrathecal administration of ICOS agonist antibody (ICOSaa) alleviates mechanical hypersensitivity caused by paclitaxel and facilitates the resolution of mechanical hypersensitivity in female mice. Administration of ICOSaa reduced astrogliosis in the spinal cord and satellite cell gliosis in the DRG of mice previously treated with paclitaxel. Mechanistically, ICOSaa intrathecal treatment promoted mechanical hypersensitivity resolution by increasing interleukin 10 (IL-10) expression in the dorsal root ganglion. In line with these observations, blocking IL-10 receptor (IL-10R) activity occluded the effects of ICOSaa treatment on mechanical hypersensitivity in female mice. Suggesting a broader activity in neuropathic pain, ICOSaa also partially resolved mechanical hypersensitivity in the spared nerve injury (SNI) model. Our findings support a model wherein ICOSaa administration induces IL-10 expression to facilitate neuropathic pain relief in female mice. ICOSaa treatment is in clinical development for solid tumors and given our observation of T cells in the human DRG, ICOSaa therapy could be developed for combination chemotherapy-CIPN clinical trials.
Collapse
Affiliation(s)
- Ishwarya Sankaranarayanan
- grid.267323.10000 0001 2151 7939Pain Neurobiology Research Group, University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX 75080 USA
| | - Diana Tavares-Ferreira
- grid.267323.10000 0001 2151 7939Pain Neurobiology Research Group, University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX 75080 USA
| | - Juliet M. Mwirigi
- grid.267323.10000 0001 2151 7939Pain Neurobiology Research Group, University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX 75080 USA
| | - Galo L. Mejia
- grid.267323.10000 0001 2151 7939Pain Neurobiology Research Group, University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX 75080 USA
| | - Michael D. Burton
- grid.267323.10000 0001 2151 7939Neuroimmunology and Behavior Laboratory, Department of Neuroscience, Center for Advanced Pain Studies, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX USA
| | - Theodore J. Price
- grid.267323.10000 0001 2151 7939Pain Neurobiology Research Group, University of Texas at Dallas, 800 W Campbell Rd, Richardson, TX 75080 USA
| |
Collapse
|
24
|
Chelette B, Chidomere CL, Dantzer R. The GDF15-GFRAL axis mediates chemotherapy-induced fatigue in mice. Brain Behav Immun 2023; 108:45-54. [PMID: 36427806 PMCID: PMC9868083 DOI: 10.1016/j.bbi.2022.11.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 11/09/2022] [Accepted: 11/12/2022] [Indexed: 11/25/2022] Open
Abstract
Cancer-related fatigue is defined as a distressing persistent subjective sense of physical, emotional, and/or cognitive tiredness or exhaustion related to cancer or cancer treatment that is not proportional to recent activity and that interferes with usual functioning. This form of fatigue is highly prevalent during cancer treatment and in some patients, it can persist for years after treatment has ended. An understanding of the mechanisms that drive cancer-related fatigue is still lacking, which hampers the identification of effective treatment options. Various chemotherapeutic agents including cisplatin are known to induce mitochondrial dysfunction and this effect is known to mediate chemotherapy-induced peripheral neuropathy and cognitive dysfunction. Mitochondrial dysfunction results in the release of mitokines that act locally and at distance to promote metabolic and behavioral adjustments to this form of cellular stress. One of these mitokines, growth differentiation factor 15 (GDF15) and its receptor, glial cell line-derived neurotrophic factor family receptor α-like (GFRAL), have received special attention in oncology as activation of GFRAL mediates the anorexic response that is responsible for cancer anorexia. The present study was initiated to determine whether GDF15 and GFRAL are involved in cisplatin-induced fatigue. We first tested the ability of cisplatin to increase circulating GDF15 in mice before assessing whether GDF15 can induce behavioral fatigue measured by decreased wheel running in healthy mice and increase behavioral fatigue induced by cisplatin. Mice administered a long acting form of GDF15, mGDF15-fc, decreased their voluntary wheel running activity. When the same treatment was administered to mice receiving cisplatin, it increased the amplitude and duration of cisplatin-induced decrease in wheel running. To determine whether endogenous GDF15 mediates the behavioral fatigue induced by cisplatin, we then administered a neutralizing monoclonal antibody to GFRAL to mice injected with cisplatin. The GFRAL neutralizing antibody mostly prevented cisplatin-induced decrease in wheel running and accelerated recovery. Taken together these findings demonstrate for the first time the role of the GDF15/GFRAL axis in cisplatin-induced behaviors and indicate that this axis could be a promising therapeutic target for the treatment of cancer-related fatigue.
Collapse
Affiliation(s)
- Brandon Chelette
- Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Chinenye L Chidomere
- Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Robert Dantzer
- Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
25
|
Dewaeles E, Carvalho K, Fellah S, Sim J, Boukrout N, Caillierez R, Ramakrishnan H, Van der Hauwaert C, Vijaya Shankara J, Martin N, Massri N, Launay A, Folger JK, de Schutter C, Larrue R, Loison I, Goujon M, Jung M, Le Gras S, Gomez-Murcia V, Faivre E, Lemaire J, Garat A, Beauval N, Maboudou P, Gnemmi V, Gibier JB, Buée L, Abbadie C, Glowacki F, Pottier N, Perrais M, Cunha RA, Annicotte JS, Laumet G, Blum D, Cauffiez C. Istradefylline protects from cisplatin-induced nephrotoxicity and peripheral neuropathy while preserving cisplatin antitumor effects. J Clin Invest 2022; 132:152924. [PMID: 36377661 PMCID: PMC9663157 DOI: 10.1172/jci152924] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 09/13/2022] [Indexed: 11/16/2022] Open
Abstract
Cisplatin is a potent chemotherapeutic drug that is widely used in the treatment of various solid cancers. However, its clinical effectiveness is strongly limited by frequent severe adverse effects, in particular nephrotoxicity and chemotherapy-induced peripheral neuropathy. Thus, there is an urgent medical need to identify novel strategies that limit cisplatin-induced toxicity. In the present study, we show that the FDA-approved adenosine A2A receptor antagonist istradefylline (KW6002) protected from cisplatin-induced nephrotoxicity and neuropathic pain in mice with or without tumors. Moreover, we also demonstrate that the antitumoral properties of cisplatin were not altered by istradefylline in tumor-bearing mice and could even be potentiated. Altogether, our results support the use of istradefylline as a valuable preventive approach for the clinical management of patients undergoing cisplatin treatment.
Collapse
Affiliation(s)
- Edmone Dewaeles
- University of Lille, INSERM, CNRS, CHU Lille, UMR9020-U1277, CANTHER, Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France.,University of Lille, INSERM, CHU Lille, UMR-S1172 LilNCog, Lille Neuroscience and Cognition, Lille, France
| | - Kévin Carvalho
- University of Lille, INSERM, CHU Lille, UMR-S1172 LilNCog, Lille Neuroscience and Cognition, Lille, France.,Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| | - Sandy Fellah
- University of Lille, INSERM, CNRS, CHU Lille, UMR9020-U1277, CANTHER, Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
| | - Jaewon Sim
- Department of Physiology, Michigan State University, East Lansing, Michigan, USA.,Cell and Molecular Biology Graduate program, Michigan State University, East Lansing, Michigan, USA
| | - Nihad Boukrout
- University of Lille, INSERM, CNRS, CHU Lille, UMR9020-U1277, CANTHER, Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
| | - Raphaelle Caillierez
- University of Lille, INSERM, CHU Lille, UMR-S1172 LilNCog, Lille Neuroscience and Cognition, Lille, France.,Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| | | | - Cynthia Van der Hauwaert
- University of Lille, INSERM, CNRS, CHU Lille, UMR9020-U1277, CANTHER, Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France.,CHU Lille, Département de la Recherche en Santé, Lille, France
| | - Jhenkruthi Vijaya Shankara
- University of Lille, INSERM, CHU Lille, UMR-S1172 LilNCog, Lille Neuroscience and Cognition, Lille, France.,Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| | - Nathalie Martin
- University of Lille, INSERM, CNRS, CHU Lille, UMR9020-U1277, CANTHER, Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
| | - Noura Massri
- Department of Physiology, Michigan State University, East Lansing, Michigan, USA.,Cell and Molecular Biology Graduate program, Michigan State University, East Lansing, Michigan, USA
| | - Agathe Launay
- University of Lille, INSERM, CHU Lille, UMR-S1172 LilNCog, Lille Neuroscience and Cognition, Lille, France.,Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| | - Joseph K. Folger
- Department of Physiology, Michigan State University, East Lansing, Michigan, USA
| | - Clémentine de Schutter
- University of Lille, INSERM, CNRS, CHU Lille, UMR9020-U1277, CANTHER, Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
| | - Romain Larrue
- University of Lille, INSERM, CNRS, CHU Lille, UMR9020-U1277, CANTHER, Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France.,CHU Lille, Service de Toxicologie et Génopathies, Lille, France
| | - Ingrid Loison
- University of Lille, INSERM, CNRS, CHU Lille, UMR9020-U1277, CANTHER, Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
| | - Marine Goujon
- University of Lille, INSERM, CNRS, CHU Lille, UMR9020-U1277, CANTHER, Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
| | - Matthieu Jung
- University of Strasbourg, CNRS UMR 7104, INSERM U1258 – GenomEast Platform – IGBMC – Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
| | - Stéphanie Le Gras
- University of Strasbourg, CNRS UMR 7104, INSERM U1258 – GenomEast Platform – IGBMC – Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
| | - Victoria Gomez-Murcia
- University of Lille, INSERM, CHU Lille, UMR-S1172 LilNCog, Lille Neuroscience and Cognition, Lille, France.,Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| | - Emilie Faivre
- University of Lille, INSERM, CHU Lille, UMR-S1172 LilNCog, Lille Neuroscience and Cognition, Lille, France.,Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| | - Julie Lemaire
- University of Lille, INSERM, CNRS, CHU Lille, UMR9020-U1277, CANTHER, Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
| | - Anne Garat
- CHU Lille, Service de Toxicologie et Génopathies, Lille, France.,University of Lille, CHU Lille, Institut Pasteur de Lille, ULR 4483, IMPact de l’Environnement Chimique sur la Santé Humaine (IMPECS), Lille, France
| | - Nicolas Beauval
- CHU Lille, Service de Toxicologie et Génopathies, Lille, France.,University of Lille, CHU Lille, Institut Pasteur de Lille, ULR 4483, IMPact de l’Environnement Chimique sur la Santé Humaine (IMPECS), Lille, France
| | - Patrice Maboudou
- CHU Lille, Service de Biochimie Automatisée, Protéines et Biologie Prédictive, Lille, France
| | - Viviane Gnemmi
- University of Lille, INSERM, CNRS, CHU Lille, UMR9020-U1277, CANTHER, Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France.,CHU Lille, Service d’Anatomopathologie, Lille, France
| | - Jean-Baptiste Gibier
- University of Lille, INSERM, CNRS, CHU Lille, UMR9020-U1277, CANTHER, Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France.,CHU Lille, Service d’Anatomopathologie, Lille, France
| | - Luc Buée
- University of Lille, INSERM, CHU Lille, UMR-S1172 LilNCog, Lille Neuroscience and Cognition, Lille, France.,Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| | - Corinne Abbadie
- University of Lille, INSERM, CNRS, CHU Lille, UMR9020-U1277, CANTHER, Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
| | - Francois Glowacki
- University of Lille, INSERM, CNRS, CHU Lille, UMR9020-U1277, CANTHER, Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France.,CHU Lille, Service de Néphrologie, Lille, France
| | - Nicolas Pottier
- University of Lille, INSERM, CNRS, CHU Lille, UMR9020-U1277, CANTHER, Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France.,CHU Lille, Service de Toxicologie et Génopathies, Lille, France
| | - Michael Perrais
- University of Lille, INSERM, CNRS, CHU Lille, UMR9020-U1277, CANTHER, Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
| | - Rodrigo A. Cunha
- CNC, Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, Faculty of Medicine Building-Polo 1, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Jean-Sébastien Annicotte
- University of Lille, INSERM, CNRS, CHU Lille, Institut Pasteur de Lille, INSERM U1283-UMR8199 – EGID, Lille, France.,University of Lille, INSERM, CHU Lille, Institut Pasteur de Lille, RID-AGE-Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, Lille, France
| | - Geoffroy Laumet
- Department of Physiology, Michigan State University, East Lansing, Michigan, USA
| | - David Blum
- University of Lille, INSERM, CHU Lille, UMR-S1172 LilNCog, Lille Neuroscience and Cognition, Lille, France.,Alzheimer and Tauopathies, LabEx DISTALZ, Lille, France
| | - Christelle Cauffiez
- University of Lille, INSERM, CNRS, CHU Lille, UMR9020-U1277, CANTHER, Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
| |
Collapse
|
26
|
Escudero-Lara A, Cabañero D, Maldonado R. Contribution of CD4+ cells in the emotional alterations induced by endometriosis in mice. Front Behav Neurosci 2022; 16:946975. [PMID: 36311856 PMCID: PMC9596757 DOI: 10.3389/fnbeh.2022.946975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Abstract
Endometriosis is a disease defined by the presence of endometrial tissue in extrauterine locations. This chronic condition is frequently associated with pain and emotional disorders and has been related with altered immune function. However, the specific involvement of immune cells in pain and behavioral symptoms of endometriosis has not been yet elucidated. Here, we implement a mouse model of non-surgical endometriosis in which immunocompetent mice develop abdomino-pelvic hypersensitivity, cognitive deficits, anxiety and depressive-like behaviors. This behavioral phenotype correlates with expression of inflammatory markers in the brain, including the immune cell marker CD4. Depletion of CD4 + cells decreases the anxiety-like behavior of mice subjected to the endometriosis model, whereas abdomino-pelvic hypersensitivity, depressive-like behavior and cognitive deficits remain unaltered. The present data reveal the involvement of the immune response characterized by CD4 + white blood cells in the anxiety-like behavior induced by endometriosis in mice. This model, which recapitulates the symptoms of human endometriosis, may be a useful tool to study the immune mechanisms involved in pain and behavioral alterations associated to endometriosis.
Collapse
Affiliation(s)
- Alejandra Escudero-Lara
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - David Cabañero
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
- David Cabañero,
| | - Rafael Maldonado
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
- *Correspondence: Rafael Maldonado,
| |
Collapse
|
27
|
Su PYP, Zhang L, He L, Zhao N, Guan Z. The Role of Neuro-Immune Interactions in Chronic Pain: Implications for Clinical Practice. J Pain Res 2022; 15:2223-2248. [PMID: 35957964 PMCID: PMC9359791 DOI: 10.2147/jpr.s246883] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 07/19/2022] [Indexed: 11/23/2022] Open
Abstract
Chronic pain remains a public health problem and contributes to the ongoing opioid epidemic. Current pain management therapies still leave many patients with poorly controlled pain, thus new or improved treatments are desperately needed. One major challenge in pain research is the translation of preclinical findings into effective clinical practice. The local neuroimmune interface plays an important role in the initiation and maintenance of chronic pain and is therefore a promising target for novel therapeutic development. Neurons interface with immune and immunocompetent cells in many distinct microenvironments along the nociceptive circuitry. The local neuroimmune interface can modulate the activity and property of the neurons to affect peripheral and central sensitization. In this review, we highlight a specific subset of many neuroimmune interfaces. In the central nervous system, we examine the interface between neurons and microglia, astrocytes, and T lymphocytes. In the periphery, we profile the interface between neurons in the dorsal root ganglion with T lymphocytes, satellite glial cells, and macrophages. To bridge the gap between preclinical research and clinical practice, we review the preclinical studies of each neuroimmune interface, discuss current clinical treatments in pain medicine that may exert its action at the neuroimmune interface, and highlight opportunities for future clinical research efforts.
Collapse
Affiliation(s)
- Po-Yi Paul Su
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, USA
| | - Lingyi Zhang
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, USA
- Department of Anesthesiology, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, People’s Republic of China
| | - Liangliang He
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, USA
- Department of Pain Management, Xuanwu Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Na Zhao
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, USA
| | - Zhonghui Guan
- Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
28
|
Wu J, Li X, Zhang X, Wang W, You X. What role of the cGAS-STING pathway plays in chronic pain? Front Mol Neurosci 2022; 15:963206. [PMID: 35979145 PMCID: PMC9376357 DOI: 10.3389/fnmol.2022.963206] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 07/11/2022] [Indexed: 12/28/2022] Open
Abstract
Chronic pain interferes with daily functioning and is frequently accompanied by depression. Currently, traditional clinic treatments do not produce satisfactory analgesic effects and frequently result in various adverse effects. Pathogen recognition receptors (PRRs) serve as innate cellular sensors of danger signals, sense invading microorganisms, and initiate innate and adaptive immune responses. Among them, cGAS-STING alerts on the presence of both exogenous and endogenous DNA in the cytoplasm, and this pathway has been closely linked to multiple diseases, including auto-inflammation, virus infection, and cancer. An increasing numbers of evidence suggest that cGAS-STING pathway involves in the chronic pain process; however, its role remains controversial. In this narrative review, we summarize the recent findings on the involvement of the cGAS-STING pathway in chronic pain, as well as several possible mechanisms underlying its activation. As a new area of research, this review is unique in considering the cGAS-STING pathway in sensory neurons and glial cells as a part of a broader understanding of pain, including potential mechanisms of inflammation, immunity, apoptosis, and autophagy. It will provide new insight into the treatment of pain in the future.
Collapse
Affiliation(s)
- Jingxiang Wu
- Department of Anesthesiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xin Li
- Department of Anesthesiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
- School of Medicine, Shanghai University, Shanghai, China
| | - Xiaoxuan Zhang
- Department of Anesthesiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
- School of Medicine, Shanghai University, Shanghai, China
| | - Wei Wang
- Department of Anesthesiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xingji You
- School of Medicine, Shanghai University, Shanghai, China
- *Correspondence: Xingji You
| |
Collapse
|
29
|
Valentine T, Hardowar L, Elphick-Ross J, Hulse RP, Paul-Clark M. The Role of Vascular-Immune Interactions in Modulating Chemotherapy Induced Neuropathic Pain. Front Pharmacol 2022; 13:887608. [PMID: 35814225 PMCID: PMC9257211 DOI: 10.3389/fphar.2022.887608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/23/2022] [Indexed: 12/05/2022] Open
Abstract
Chemotherapy causes sensory disturbances in cancer patients that results in neuropathies and pain. As cancer survivorships has dramatically increased over the past 10 years, pain management of these patients is becoming clinically more important. Current analgesic strategies are mainly ineffective and long-term use is associated with severe side effects. The issue being that common analgesic strategies are based on ubiquitous pain mediator pathways, so when applied to clinically diverse neuropathic pain and neurological conditions, are unsuccessful. This is principally due to the lack of understanding of the driving forces that lead to chemotherapy induced neuropathies. It is well documented that chemotherapy causes sensory neurodegeneration through axonal atrophy and intraepidermal fibre degeneration causing alterations in pain perception. Despite the neuropathological alterations associated with chemotherapy-induced neuropathic pain being extensively researched, underlying causes remain elusive. Resent evidence from patient and rodent studies have indicated a prominent inflammatory cell component in the peripheral sensory nervous system in effected areas post chemotherapeutic treatment. This is accompanied by modulation of auxiliary cells of the dorsal root ganglia sensory neurons such as activation of satellite glia and capillary dysfunction. The presence of a neuroinflammatory component was supported by transcriptomic analysis of dorsal root ganglia taken from mice treated with common chemotherapy agents. With key inflammatory mediators identified, having potent immunoregulatory effects that directly influences nociception. We aim to evaluate the current understanding of these immune-neuronal interactions across different cancer therapy drug classes. In the belief this may lead to better pain management approaches for cancer survivors.
Collapse
|
30
|
Mekonnen TW, Darge HF, Tsai HC, Birhan YS, Hanurry EY, Gebrie HT, Chou HY, Lai JY, Lin SZ, Harn HJ, Chen YS. Combination of ovalbumin-coated iron oxide nanoparticles and poly(amidoamine) dendrimer-cisplatin nanocomplex for enhanced anticancer efficacy. Colloids Surf B Biointerfaces 2022; 213:112391. [PMID: 35158218 DOI: 10.1016/j.colsurfb.2022.112391] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 01/10/2022] [Accepted: 02/02/2022] [Indexed: 12/12/2022]
Abstract
Enhancement of drug efficacy is essential in cancer treatment. The immune stimulator ovalbumin (Ova)-coated citric acid (AC-)-stabilized iron oxide nanoparticles (AC-IO-Ova NPs) and enhanced permeability and retention (EPR)-based tumor targeted 4.5 generation poly(amidoamine) dendrimer(4.5GDP)-cisplatin (Cis-pt) nanocomplex (NC) (4.5GDP-Cis-pt NC) were used for enhanced anticancer efficiency. The formations of 4.5GDP-Cis-pt NC, AC-IO, and AC-IO-Ova NPs were examined via FTIR spectroscopy, X-ray diffraction, Raman spectroscopy, and X-ray photoelectron spectroscopy. The conjugation of Cis-pt with 4.5GDP was confirmed using carbon NMR spectroscopy. The tumor-specific 4.5GDP-Cis-pt NC provided 45%and 28% cumulative cisplatin release in 72 h at pH 6.5 and 7.4, respectively. A significant immune response with high TNF-α and IL-6 cytokine secretion was confirmed for the co-incubation of AC-IO-Ova with RAW 264.7 or HaCaT cells. AC-IO-Ova NPs were biocompatible with different cell lines, even at a high concentration (200 µg mL-1). However, AC-IO-Ova NPs mixed with 4.5GDP-Cis-pt NC (Cis-pt at 15 µg mL-1) significantly increased the cytotoxicity against the cancer cells in a dose-dependent manner with the increasing AC-IO-Ova NPs concentrations. The increased anticancer effects may be attributed to the generation of reactive oxygen species (ROS). Moreover, AC-IO-Ova NPs might assist the efficiency of anticancer cells, inducing an innate immune response via M1 macrophage polarization. We provide a novel synergistic chemoimmunotherapeutic strategy to enhance the anticancer efficacy of cisplatin via a chemotherapeutic agent 4.5GDP-Cis-pt NC and induce proinflammatory cytokines stimulating innate immunity through AC-IO-Ova NPs against tumors.
Collapse
Affiliation(s)
- Tefera Worku Mekonnen
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei 106, Taiwan, ROC
| | - Haile Fentahun Darge
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei 106, Taiwan, ROC
| | - Hsieh-Chih Tsai
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei 106, Taiwan, ROC; Advanced Membrane Materials Center, National Taiwan University of Science and Technology, Taipei 106, Taiwan, ROC; R&D Center for Membrane Technology, Chung Yuan Christian University, Chungli, Taoyuan 320, Taiwan, ROC.
| | - Yihenew Simegniew Birhan
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei 106, Taiwan, ROC
| | - Endiries Yibru Hanurry
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei 106, Taiwan, ROC
| | - Hailemichael Tegenu Gebrie
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei 106, Taiwan, ROC
| | - Hsiao-Ying Chou
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei 106, Taiwan, ROC
| | - Juin-Yih Lai
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei 106, Taiwan, ROC; Advanced Membrane Materials Center, National Taiwan University of Science and Technology, Taipei 106, Taiwan, ROC; R&D Center for Membrane Technology, Chung Yuan Christian University, Chungli, Taoyuan 320, Taiwan, ROC; Department of Chemical Engineering & Materials Science, Yuan Ze University, Chungli, Taoyuan 320, Taiwan, ROC
| | - Shinn-Zong Lin
- Department of Neurosurgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan, ROC
| | - Horng-Jyh Harn
- Department of Pathology, Hualien Tzu Chi Hospital, Tzu Chi University, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan, ROC
| | - Yu-Shuan Chen
- Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Taiwan, ROC; Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan, ROC; Tzu Chi University of Science and Technology, Taiwan, ROC.
| |
Collapse
|
31
|
Illias AM, Yu KJ, Hwang SH, Solis J, Zhang H, Velasquez JF, Cata JP, Dougherty PM. Dorsal root ganglion toll-like receptor 4 signaling contributes to oxaliplatin-induced peripheral neuropathy. Pain 2022; 163:923-935. [PMID: 34490849 DOI: 10.1097/j.pain.0000000000002454] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 08/10/2021] [Indexed: 11/26/2022]
Abstract
ABSTRACT Activation of toll-like receptor 4 (TLR4) in the dorsal root ganglion (DRG) and spinal cord contributes to the generation of paclitaxel-related chemotherapy-induced peripheral neuropathy (CIPN). Generalizability of TLR4 signaling in oxaliplatin-induced CIPN was tested here. Mechanical hypersensitivity developed in male SD rats by day 1 after oxaliplatin treatment, reached maximum intensity by day 14, and persisted through day 35. Western blot revealed an increase in TLR4 expression in the DRG of oxaliplatin at days 1 and 7 after oxaliplatin treatment. Cotreatment of rats with the TLR4 antagonist lipopolysaccharide derived from Rhodobacter sphaeroides ultrapure or with the nonspecific immunosuppressive minocycline with oxaliplatin resulted in significantly attenuated hyperalgesia on day 7 and 14 compared with rats that received oxaliplatin plus saline vehicle. Immunostaining of DRGs revealed an increase in the number of neurons expressing TLR4, its canonical downstream signal molecules myeloid differentiation primary response gene 88 (MyD88) and TIR-domain-containing adapter-inducing interferon-β, at both day 7 and day 14 after oxaliplatin treatment. These increases were blocked by cotreatment with either lipopolysaccharide derived from Rhodobacter sphaeroides or minocycline. Double staining showed the localization of TLR4, MyD88, and TIR-domain-containing adapter-inducing interferon-β in subsets of DRG neurons. Finally, there was no significant difference in oxaliplatin-induced mechanical hypersensitivity between male and female rats when observed for 2 weeks. Furthermore, upregulation of TLR4 was detected in both sexes when tested 14 days after treatment with oxaliplatin. These findings suggest that the activation of TLR4 signaling in DRG neurons is a common mechanism in CIPN induced by multiple cancer chemotherapy agents.
Collapse
Affiliation(s)
- Amina M Illias
- Department of Anesthesiology and Pain Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, TX, United States
- Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Kai-Jie Yu
- Department of Urology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Seon-Hee Hwang
- Department of Anesthesiology and Pain Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, TX, United States
| | - Jacob Solis
- Department of Anesthesiology and Pain Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, TX, United States
| | - Hongmei Zhang
- Department of Anesthesiology and Pain Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, TX, United States
| | - Jose F Velasquez
- Department of Anesthesiology and Perioperative Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, TX, United States
| | - Juan P Cata
- Department of Anesthesiology and Perioperative Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, TX, United States
| | - Patrick M Dougherty
- Department of Anesthesiology and Pain Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
32
|
Hunt MA, Lund H, Delay L, Dos Santos GG, Pham A, Kurtovic Z, Telang A, Lee A, Parvathaneni A, Kussick E, Corr M, Yaksh TL. DRGquant: A new modular AI-based pipeline for 3D analysis of the DRG. J Neurosci Methods 2022; 371:109497. [PMID: 35181343 PMCID: PMC10644910 DOI: 10.1016/j.jneumeth.2022.109497] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 01/17/2022] [Accepted: 02/06/2022] [Indexed: 12/01/2022]
Abstract
BACKGROUND The dorsal root ganglion (DRG) is structurally complex and pivotal to systems processing nociception. Whole mount analysis allows examination of intricate microarchitectural and cellular relationships of the DRG in three-dimensional (3D) space. NEW METHOD We present DRGquant a set of tools and techniques optimized as a pipeline for automated image analysis and reconstruction of cells/structures within the DRG. We have developed an open source software pipeline that utilizes machine learning to identify substructures within the DRG and reliably classify and quantify them. RESULTS Our methods were sufficiently sensitive to isolate, analyze, and classify individual DRG substructures including macrophages. The activation of macrophages was visualized and quantified in the DRG following intrathecal injection of lipopolysaccharide, and in a model of chemotherapy induced peripheral neuropathy. The percent volume of infiltrating macrophages was similar to a commercial source in quantification. Circulating fluorescent dextran was visualized within DRG macrophages using whole mount preparations, which enabled 3D reconstruction of the DRG and DRGquant demonstrated subcellular spatial resolution within individual macrophages. COMPARISON WITH EXISTING METHOD(S) Here we describe a reliable and efficient methodologic pipeline to prepare cleared and whole mount DRG tissue. DRGquant allows automated image analysis without tedious manual gating to reduce bias. The quantitation of DRG macrophages was superior to commercial solutions. CONCLUSIONS Using machine learning to separate signal from noise and identify individual cells, DRGquant enabled us to isolate individual structures or areas of interest within the DRG for a more granular and fine-tuned analysis. Using these 3D techniques, we are better able to appreciate the biology of the DRG under experimental inflammatory conditions.
Collapse
Affiliation(s)
- Matthew A Hunt
- Departments of Anesthesiology and Pharmacology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Department of Physiology, Karolinska Institutet, Stockholm, Sweden.
| | - Harald Lund
- Departments of Anesthesiology and Pharmacology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Department of Physiology, Karolinska Institutet, Stockholm, Sweden.
| | - Lauriane Delay
- Departments of Anesthesiology and Pharmacology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA.
| | - Gilson Goncalves Dos Santos
- Departments of Anesthesiology and Pharmacology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA.
| | - Albert Pham
- Departments of Anesthesiology and Pharmacology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA.
| | - Zerina Kurtovic
- Department of Physiology, Karolinska Institutet, Stockholm, Sweden.
| | - Aditya Telang
- Departments of Anesthesiology and Pharmacology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA.
| | - Adam Lee
- Departments of Anesthesiology and Pharmacology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA.
| | - Akhil Parvathaneni
- Departments of Anesthesiology and Pharmacology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA.
| | - Emily Kussick
- Departments of Anesthesiology and Pharmacology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA.
| | - Maripat Corr
- Department of Medicine, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093, USA.
| | - Tony L Yaksh
- Departments of Anesthesiology and Pharmacology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Departments of Anesthesiology and Pharmacology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA.
| |
Collapse
|
33
|
Goode DJ, Whitaker EE, Mecum NE. Ovariectomy increases paclitaxel-induced mechanical hypersensitivity and reduces anti-inflammatory CD4+ T cells in the dorsal root ganglion of female mice. J Neuroimmunol 2022; 367:577878. [DOI: 10.1016/j.jneuroim.2022.577878] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/01/2022] [Accepted: 04/19/2022] [Indexed: 12/28/2022]
|
34
|
Abstract
We aimed to investigate a sexually dimorphic role of calcitonin gene-related peptide (CGRP) in rodent models of pain. Based on findings in migraine where CGRP has a preferential pain-promoting effect in female rodents, we hypothesized that CGRP antagonists and antibodies would attenuate pain sensitization more efficaciously in female than male mice and rats. In hyperalgesic priming induced by activation of interleukin 6 signaling, CGRP receptor antagonists olcegepant and CGRP8-37 both given intrathecally, blocked, and reversed hyperalgesic priming only in females. A monoclonal antibody against CGRP, given systemically, blocked priming specifically in female rodents but failed to reverse it. In the spared nerve injury model, there was a transient effect of both CGRP antagonists, given intrathecally, on mechanical hypersensitivity in female mice only. Consistent with these findings, intrathecally applied CGRP caused a long-lasting, dose-dependent mechanical hypersensitivity in female mice but more transient effects in males. This CGRP-induced mechanical hypersensitivity was reversed by olcegepant and the KCC2 enhancer CLP257, suggesting a role for anionic plasticity in the dorsal horn in the pain-promoting effects of CGRP in females. In spinal dorsal horn slices, CGRP shifted GABAA reversal potentials to significantly more positive values, but, again, only in female mice. Therefore, CGRP may regulate KCC2 expression and/or activity downstream of CGRP receptors specifically in females. However, KCC2 hypofunction promotes mechanical pain hypersensitivity in both sexes because CLP257 alleviated hyperalgesic priming in male and female mice. We conclude that CGRP promotes pain plasticity in female rodents but has a limited impact in males.SIGNIFICANCE STATEMENT The majority of patients impacted by chronic pain are women. Mechanistic studies in rodents are creating a clear picture that molecular events promoting chronic pain are different in male and female animals. We sought to build on evidence showing that CGRP is a more potent and efficacious promoter of headache in female than in male rodents. To test this, we used hyperalgesic priming and the spared nerve injury neuropathic pain models in mice. Our findings show a clear sex dimorphism wherein CGRP promotes pain in female but not male mice, likely via a centrally mediated mechanism of action. Our work suggests that CGRP receptor antagonists could be tested for efficacy in women for a broader variety of pain conditions.
Collapse
|
35
|
Singh SK, Krukowski K, Laumet GO, Weis D, Alexander JF, Heijnen CJ, Kavelaars A. CD8+ T cell-derived IL-13 increases macrophage IL-10 to resolve neuropathic pain. JCI Insight 2022; 7:154194. [PMID: 35260535 PMCID: PMC8983134 DOI: 10.1172/jci.insight.154194] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 01/26/2022] [Indexed: 01/11/2023] Open
Abstract
Understanding the endogenous mechanisms regulating resolution of pain may identify novel targets for treatment of chronic pain. Resolution of chemotherapy-induced peripheral neuropathy (CIPN) after treatment completion depends on CD8+ T cells and on IL-10 produced by other cells. Using Rag2–/– mice lacking T and B cells and adoptive transfer of Il13–/– CD8+ T cells, we showed that CD8+ T cells producing IL-13 were required for resolution of CIPN. Intrathecal administration of anti–IL-13 delayed resolution of CIPN and reduced IL-10 production by dorsal root ganglion macrophages. Depleting local CD206+ macrophages also delayed resolution of CIPN. In vitro, TIM3+CD8+ T cells cultured with cisplatin, apoptotic cells, or phosphatidylserine liposomes produced IL-13, which induced IL-10 in macrophages. In vivo, resolution of CIPN was delayed by intrathecal administration of anti-TIM3. Resolution was also delayed in Rag2–/– mice reconstituted with Havcr2 (TIM3)–/– CD8+ T cells. Our data indicated that cell damage induced by cisplatin activated TIM3 on CD8+ T cells, leading to increased IL-13 production, which in turn induced macrophage IL-10 production and resolution of CIPN. Development of exogenous activators of the IL-13/IL-10 pain resolution pathway may provide a way to treat the underlying cause of chronic pain.
Collapse
Affiliation(s)
- Susmita K Singh
- Laboratories of Neuroimmunology, Department of Symptom Research, the University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Karen Krukowski
- Laboratories of Neuroimmunology, Department of Symptom Research, the University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Department of Biological Sciences, Knoebel Institute for Healthy Aging, University of Denver, Denver, Colorado, USA
| | - Geoffroy O Laumet
- Laboratories of Neuroimmunology, Department of Symptom Research, the University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Department of Physiology, College of Natural Science, Michigan State University, East Lansing, Michigan, USA
| | - Drew Weis
- Laboratories of Neuroimmunology, Department of Symptom Research, the University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jenolyn F Alexander
- Laboratories of Neuroimmunology, Department of Symptom Research, the University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Cobi J Heijnen
- Laboratories of Neuroimmunology, Department of Symptom Research, the University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Annemieke Kavelaars
- Laboratories of Neuroimmunology, Department of Symptom Research, the University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
36
|
Mecklenburg J, Wangzhou A, Hovhannisyan AH, Barba-Escobedo P, Shein SA, Zou Y, Weldon K, Lai Z, Goffin V, Dussor G, Tumanov AV, Price TJ, Akopian AN. Sex-dependent pain trajectories induced by prolactin require an inflammatory response for pain resolution. Brain Behav Immun 2022; 101:246-263. [PMID: 35065194 PMCID: PMC9173405 DOI: 10.1016/j.bbi.2022.01.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 01/11/2022] [Accepted: 01/16/2022] [Indexed: 11/23/2022] Open
Abstract
Pain development and resolution patterns in many diseases are sex-dependent. This study aimed to develop pain models with sex-dependent resolution trajectories, and identify factors linked to resolution of pain in females and males. Using different intra-plantar (i.pl.) treatment protocols with prolactin (PRL), we established models with distinct, sex-dependent patterns for development and resolution of pain. An acute PRL-evoked pain trajectory, in which hypersensitivity is fully resolved within 1 day, showed substantial transcriptional changes after pain-resolution in female and male hindpaws and in the dorsal root ganglia (DRG). This finding supports the notion that pain resolution is an active process. Prolonged treatment with PRL high dose (1 μg) evoked mechanical hypersensitivity that resolved within 5-7 days in mice of both sexes and exhibited a pro-inflammatory transcriptional response in the hindpaw, but not DRG, at the time point preceding resolution. Flow cytometry analysis linked pro-inflammatory responses in female hindpaws to macrophages/monocytes, especially CD11b+/CD64+/MHCII+ cell accumulation. Prolonged low dose PRL (0.1 μg) treatment caused non-resolving mechanical hypersensitivity only in females. This effect was independent of sensory neuronal PRLR and was associated with a lack of immune response in the hindpaw, although many genes underlying tissue damage were affected. We conclude that different i.pl. PRL treatment protocols generates distinct, sex-specific pain hypersensitivity resolution patterns. PRL-induced pain resolution is preceded by a pro-inflammatory macrophage/monocyte-associated response in the hindpaws of mice of both sexes. On the other hand, the absence of a peripheral inflammatory response creates a permissive condition for PRL-induced pain persistency in females.
Collapse
Affiliation(s)
- Jennifer Mecklenburg
- Department of Endodontics, The School of Dentistry, University of Texas Health Science Center at San Antonio (UTHSCSA), San Antonio, TX 78229, United States
| | - Andi Wangzhou
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas (UTD), Richardson, TX 75080, United States
| | - Anahit H Hovhannisyan
- Department of Endodontics, The School of Dentistry, University of Texas Health Science Center at San Antonio (UTHSCSA), San Antonio, TX 78229, United States
| | - Priscilla Barba-Escobedo
- Department of Endodontics, The School of Dentistry, University of Texas Health Science Center at San Antonio (UTHSCSA), San Antonio, TX 78229, United States
| | - Sergey A Shein
- Departments of Microbiology, Immunology & Molecular Genetics, the School of Medicine, UTHSCSA, San Antonio, TX 78229, United States
| | - Yi Zou
- Molecular Medicine, The School of Medicine, UTHSCSA, San Antonio, TX 78229, United States
| | - Korri Weldon
- Molecular Medicine, The School of Medicine, UTHSCSA, San Antonio, TX 78229, United States
| | - Zhao Lai
- Molecular Medicine, The School of Medicine, UTHSCSA, San Antonio, TX 78229, United States; Greehey Children's Cancer Research Institute, UTHSCSA, United States
| | | | - Gregory Dussor
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas (UTD), Richardson, TX 75080, United States
| | - Alexei V Tumanov
- Departments of Microbiology, Immunology & Molecular Genetics, the School of Medicine, UTHSCSA, San Antonio, TX 78229, United States
| | - Theodore J Price
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas (UTD), Richardson, TX 75080, United States
| | - Armen N Akopian
- Department of Endodontics, The School of Dentistry, University of Texas Health Science Center at San Antonio (UTHSCSA), San Antonio, TX 78229, United States; Departments of Pharmacology, The School of Medicine, UTHSCSA, San Antonio, TX 78229, United States.
| |
Collapse
|
37
|
Targeting the A 3 adenosine receptor to prevent and reverse chemotherapy-induced neurotoxicities in mice. Acta Neuropathol Commun 2022; 10:11. [PMID: 35093182 PMCID: PMC8800287 DOI: 10.1186/s40478-022-01315-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 01/17/2022] [Indexed: 02/07/2023] Open
Abstract
Cisplatin is used to combat solid tumors. However, patients treated with cisplatin often develop cognitive impairments, sensorimotor deficits, and peripheral neuropathy. There is no FDA-approved treatment for these neurotoxicities. We investigated the capacity of a highly selective A3 adenosine receptor (AR) subtype (A3AR) agonist, MRS5980, to prevent and reverse cisplatin-induced neurotoxicities. MRS5980 prevented cisplatin-induced cognitive impairment (decreased executive function and impaired spatial and working memory), sensorimotor deficits, and neuropathic pain (mechanical allodynia and spontaneous pain) in both sexes. At the structural level, MRS5980 prevented the cisplatin-induced reduction in markers of synaptic integrity. In-situ hybridization detected Adora3 mRNA in neurons, microglia, astrocytes and oligodendrocytes. RNAseq analysis identified 164 genes, including genes related to mitochondrial function, of which expression was changed by cisplatin and normalized by MRS5980. Consistently, MRS5980 prevented cisplatin-induced mitochondrial dysfunction and decreased signs of oxidative stress. Transcriptomic analysis showed that the A3AR agonist upregulates genes related to repair pathways including NOTCH1 signaling and chromatin modification in the cortex of cisplatin-treated mice. Importantly, A3AR agonist administration after completion of cisplatin treatment resolved cognitive impairment, neuropathy and sensorimotor deficits. Our results highlight the efficacy of a selective A3AR agonist to prevent and reverse cisplatin-induced neurotoxicities via preventing brain mitochondrial damage and activating repair pathways. An A3AR agonist is already in cancer, clinical trials and our results demonstrate management of neurotoxic side effects of chemotherapy as an additional therapeutic benefit.
Collapse
|
38
|
D'Agnelli S, Amodeo G, Franchi S, Verduci B, Baciarello M, Panerai AE, Bignami EG, Sacerdote P. Frailty and pain, human studies and animal models. Ageing Res Rev 2022; 73:101515. [PMID: 34813977 DOI: 10.1016/j.arr.2021.101515] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 11/08/2021] [Accepted: 11/11/2021] [Indexed: 11/01/2022]
Abstract
The hypothesis that pain can predispose to frailty development has been recently investigated in several clinical studies suggesting that frailty and pain may share some mechanisms. Both pain and frailty represent important clinical and social problems and both lack a successful treatment. This circumstance is mainly due to the absence of in-depth knowledge of their pathological mechanisms. Evidence of shared pathways between frailty and pain are preliminary. Indeed, many clinical studies are observational and the impact of pain treatment, and relative pain-relief, on frailty onset and progression has never been investigated. Furthermore, preclinical research on this topic has yet to be performed. Specific researches on the pain-frailty relation are needed. In this narrative review, we will attempt to point out the most relevant findings present in both clinical and preclinical literature on the topic, with particular attention to genetics, epigenetics and inflammation, in order to underline the existing gaps and the potential future interventional strategies. The use of pain and frailty animal models discussed in this review might contribute to research in this area.
Collapse
|
39
|
Galvin DA, C M. The role of T-lymphocytes in neuropathic pain initiation, development of chronicity and treatment. Brain Behav Immun Health 2021; 18:100371. [PMID: 34761242 PMCID: PMC8566770 DOI: 10.1016/j.bbih.2021.100371] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 10/04/2021] [Accepted: 10/13/2021] [Indexed: 12/25/2022] Open
Abstract
Ongoing research has strongly suggested the role the immune system plays in the pathogenesis of neuropathic pain. T cells appear to be one of the main regulators of the immune system with many mediators appearing to promote or suppress pain resolution. Limited effective therapies are available for treatment of neuropathic pain. Treatments available appear to modulate specific T cell with altered ratios present 3 months post treatment and parallels clinical improvement. This further supports the neuro-immune basis for neuropathic pain chronicity. Identification of novel immune mediators involved in pain development may suggest new target areas in treatment. Neuroimmunity plays a significant role in neuropathic pain pathogenesis neuropathic pain. Immune mediators contribute to promotion, suppression or resolution of neuropathic pain. Clinical studies in humans are lacking, most research available is pre-clinical or animal-based. Evidence-based therapies for treatment of neuropathic pain demonstrate alteration in T cell phenotype and behavior post therapy.
Collapse
Affiliation(s)
- D A Galvin
- Department of Pain Medicine, Trinity Translational Medicine Institute, St. James's Hospital and Trinity College Dublin, Dublin 8, Ireland.,Hermitage Medical Clinic, Old Lucan Road, Dublin 20, Ireland
| | - McCrory C
- Department of Pain Medicine, Trinity Translational Medicine Institute, St. James's Hospital and Trinity College Dublin, Dublin 8, Ireland.,Hermitage Medical Clinic, Old Lucan Road, Dublin 20, Ireland
| |
Collapse
|
40
|
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Annemieke Kavelaars
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Cobi J Heijnen
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
41
|
Vichaya EG, Ford BG, Moltenkine JM, Taniguchi CM, Phillip West A, Dantzer R. Sex differences in the behavioral and immune responses of mice to tumor growth and cancer therapy. Brain Behav Immun 2021; 98:161-172. [PMID: 34418499 PMCID: PMC8511067 DOI: 10.1016/j.bbi.2021.08.225] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 07/29/2021] [Accepted: 08/14/2021] [Indexed: 10/20/2022] Open
Abstract
There is significant variability in the expression of cancer-related fatigue. Understanding the factors that account for this variation provide insight into the underlying mechanisms. One important, but often overlooked, variable is biological sex. While a few clinical studies have indicated that female patients report higher levels of fatigue, these studies are subject to potential socio-culture reporting biases. Only a limited number of preclinical studies have considered sex differences in animal model of fatigue and few have simultaneously considered both disease- and treatment-related factors. The present series of studies was initiated to address the current knowledge gap on the importance of sex differences in cancer-related fatigue. We selected a murine model of human papilloma virus-positive head and neck cancer based on heterotypic injection of the mEERL95 cell line that grows in both male and female mice and responds to a regimen of cisplatin plus irradiation. We also tested the impact of immunotherapy treatment targeting PD1. Voluntary wheel running was used to evaluate fatigue-like behavior. Male mice grew larger tumors than did female mice and showed more severe fatigue-like behavior. We confirmed that the tumor increased the expression of inflammatory cytokines in the liver, but no sex differences were observed. As a trend toward elevated Cd3 mRNA was observed in female mice, we tested the importance of T cells using female Rag2-/- mice. The Rag2-/- female mice had accelerated tumor growth and more severe fatigue-like behavior. In response to cisplatin alone non-tumor-bearing female mice showed a slower recovery of wheel running activity compared to males. However, in response to chemoradiation and anti-PD1 neutralizing antibody, tumor-bearing female mice showed a better tumor response to therapy than male mice, but no significant sex differences were observed for wheel running. These findings point to different mechanisms underlying tumor- and treatment-induced behavioral fatigue and indicate that the sex factor can intervene to modulate the expression of fatigue-like behavior in particular circumstances.
Collapse
Affiliation(s)
- Elisabeth G Vichaya
- Department of Psychology & Neuroscience, Baylor University, Waco, TX 76798, United States
| | - Bianca G Ford
- Department of Symptom Research, MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Jessica M Moltenkine
- Department of Experimental Radiation Oncology, MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Cullen M Taniguchi
- Department of Experimental Radiation Oncology, MD Anderson Cancer Center, Houston, TX 77030, United States
| | - A Phillip West
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M University, Bryan, TX 77087, United States
| | - Robert Dantzer
- Department of Symptom Research, MD Anderson Cancer Center, Houston, TX 77030, United States.
| |
Collapse
|
42
|
Lassen J, Stürner KH, Gierthmühlen J, Dargvainiene J, Kixmüller D, Leypoldt F, Baron R, Hüllemann P. Protective role of natural killer cells in neuropathic pain conditions. Pain 2021; 162:2366-2375. [PMID: 33769361 DOI: 10.1097/j.pain.0000000000002274] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 03/02/2021] [Indexed: 11/25/2022]
Abstract
ABSTRACT During the past few years, the research of chronic neuropathic pain has focused on neuroinflammation within the central nervous system and its impact on pain chronicity. As part of the ERA-Net NEURON consortium, we aimed to identify immune cell patterns in the cerebrospinal fluid (CSF) of patients with herpes zoster neuralgia and patients with polyneuropathy (PNP), which may contribute to pain chronicity in these neuropathic pain conditions. Cerebrospinal fluid of 41 patients (10 herpes zoster and 31 PNP) was analyzed by flow cytometry identifying lymphocyte subsets: CD4+ (T-helper cells), CD8+ (cytotoxic T cells), CD19+ (B cells), and CD56+ (natural killer [NK]) cells. At baseline and at follow-up, the somatosensory phenotype was assessed with quantitative sensory testing. In addition, the patients answered epidemiological questionnaires and the PainDETECT questionnaire. Immune cell profiles and somatosensory profiles, as well as painDETECT questionnaire scores, were analyzed and correlated to determine specific immune cell patterns, which contribute to chronic pain. We found a negative correlation (P = 0.004, r = -0.596) between the frequency of NK cells and mechanical pain sensitivity (MPS), one of the most relevant quantitative sensory testing markers for central sensitization; a high frequency of NK cells correlated with low MPS. The analysis of the individual follow-up showed a worsening of the pain condition if NK-cell frequency was low. Low NK-cell frequency is associated with signs of central sensitization (MPS), whereas high NK-cell frequency might prevent central sensitization. Therefore, NK cells seem to play a protective role within the neuroinflammatory cascade and may be used as a marker for pain chronicity.
Collapse
Affiliation(s)
- Josephine Lassen
- Division of Neurological Pain Research and Therapy, Department of Neurology, University Hospital Schleswig-Holstein, Kiel Campus, Kiel, Germany
| | - Klarissa Hanja Stürner
- Department of Neurology, University Hospital Schleswig-Holstein, Kiel Campus, Kiel, Germany
| | - Janne Gierthmühlen
- Division of Neurological Pain Research and Therapy, Department of Neurology, University Hospital Schleswig-Holstein, Kiel Campus, Kiel, Germany
| | - Justina Dargvainiene
- Institute of Clinical Chemistry, University Hospital Schleswig-Holstein, Kiel Campus, Kiel, Germany
| | - Dorthe Kixmüller
- Institute of Clinical Chemistry, University Hospital Schleswig-Holstein, Kiel Campus, Kiel, Germany
| | - Frank Leypoldt
- Department of Neurology, University Hospital Schleswig-Holstein, Kiel Campus, Kiel, Germany
- Institute of Clinical Chemistry, University Hospital Schleswig-Holstein, Kiel Campus, Kiel, Germany
| | - Ralf Baron
- Division of Neurological Pain Research and Therapy, Department of Neurology, University Hospital Schleswig-Holstein, Kiel Campus, Kiel, Germany
| | - Philipp Hüllemann
- Division of Neurological Pain Research and Therapy, Department of Neurology, University Hospital Schleswig-Holstein, Kiel Campus, Kiel, Germany
| |
Collapse
|
43
|
Lenert ME, Avona A, Garner KM, Barron LR, Burton MD. Sensory Neurons, Neuroimmunity, and Pain Modulation by Sex Hormones. Endocrinology 2021; 162:bqab109. [PMID: 34049389 PMCID: PMC8237991 DOI: 10.1210/endocr/bqab109] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Indexed: 12/16/2022]
Abstract
The inclusion of women in preclinical pain studies has become more commonplace in the last decade as the National Institutes of Health (NIH) released its "Sex as a Biological Variable" mandate. Presumably, basic researchers have not had a comprehensive understanding about neuroimmune interactions in half of the population and how hormones play a role in this. To date, we have learned that sex hormones contribute to sexual differentiation of the nervous system and sex differences in behavior throughout the lifespan; however, the cycling of sex hormones does not always explain these differences. Here, we highlight recent advances in our understanding of sex differences and how hormones and immune interactions influence sensory neuron activity to contribute to physiology and pain. Neuroimmune mechanisms may be mediated by different cell types in each sex, as the actions of immune cells are sexually dimorphic. Unfortunately, the majority of studies assessing neuronal contributions to immune function have been limited to males, so it is unclear if the mechanisms are similar in females. Finally, pathways that control cellular metabolism, like nuclear receptors, have been shown to play a regulatory role both in pain and inflammation. Overall, communication between the neuroimmune and endocrine systems modulate pain signaling in a sex-dependent manner, but more research is needed to reveal nuances of these mechanisms.
Collapse
Affiliation(s)
- Melissa E Lenert
- Neuroimmunology and Behavior Laboratory, Center for Advanced Pain Studies (CAPS), Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080, USA
| | - Amanda Avona
- Neuroimmunology and Behavior Laboratory, Center for Advanced Pain Studies (CAPS), Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080, USA
| | - Katherine M Garner
- Neuroimmunology and Behavior Laboratory, Center for Advanced Pain Studies (CAPS), Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080, USA
| | - Luz R Barron
- Neuroimmunology and Behavior Laboratory, Center for Advanced Pain Studies (CAPS), Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080, USA
| | - Michael D Burton
- Neuroimmunology and Behavior Laboratory, Center for Advanced Pain Studies (CAPS), Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080, USA
| |
Collapse
|
44
|
Wangzhou A, Paige C, Ray PR, Dussor G, Price TJ. Diversity of Receptor Expression in Central and Peripheral Mouse Neurons Estimated from Single Cell RNA Sequencing. Neuroscience 2021; 463:86-96. [PMID: 33774127 PMCID: PMC8106651 DOI: 10.1016/j.neuroscience.2021.03.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 03/12/2021] [Accepted: 03/16/2021] [Indexed: 12/15/2022]
Abstract
Because somatosensory PNS neurons, in particular nociceptors, are specially tuned to be able to detect a wide variety of both exogenous and endogenous signals, one might assume that these neurons express a greater variety of receptor genes. This assumption has not been formally tested. Because cells detect such signals via cell surface receptors, we sought to formally test the hypothesis that PNS neurons might express a broader array of cell surface receptors than CNS neurons using existing single cell RNA sequencing resources from mouse. We focused our analysis on ion channels, G-protein coupled receptors (GPCRS), receptor tyrosine kinase and cytokine family receptors. In partial support of our hypothesis, we found that mouse PNS somatosensory, sympathetic and enteric neurons and CNS neurons have similar receptor expression diversity in families of receptors examined, with the exception of GPCRs and cytokine receptors which showed greater diversity in the PNS. Surprisingly, these differences were mostly driven by enteric and sympathetic neurons, not by somatosensory neurons or nociceptors. Secondary analysis revealed many receptors that are very specifically expressed in subsets of PNS neurons, including some that are unique among neurons for nociceptors. Finally, we sought to examine specific ligand-receptor interactions between T cells and PNS and CNS neurons. Again, we noted that most interactions between these cells are shared by CNS and PNS neurons despite the fact that T cells only enter the CNS under rare circumstances. Our findings demonstrate that both PNS and CNS neurons express an astonishing array of cell surface receptors and suggest that most neurons are tuned to receive signals from other cells types, in particular immune cells.
Collapse
Affiliation(s)
- Andi Wangzhou
- University of Texas at Dallas, Department of Neuroscience and Center for Advanced Pain Studies, United States
| | - Candler Paige
- University of Texas at Dallas, Department of Neuroscience and Center for Advanced Pain Studies, United States
| | - Pradipta R Ray
- University of Texas at Dallas, Department of Neuroscience and Center for Advanced Pain Studies, United States
| | - Gregory Dussor
- University of Texas at Dallas, Department of Neuroscience and Center for Advanced Pain Studies, United States
| | - Theodore J Price
- University of Texas at Dallas, Department of Neuroscience and Center for Advanced Pain Studies, United States.
| |
Collapse
|
45
|
Agalave NM, Mody PH, Szabo-Pardi TA, Jeong HS, Burton MD. Neuroimmune Consequences of eIF4E Phosphorylation on Chemotherapy-Induced Peripheral Neuropathy. Front Immunol 2021; 12:642420. [PMID: 33912169 PMCID: PMC8071873 DOI: 10.3389/fimmu.2021.642420] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 03/18/2021] [Indexed: 12/17/2022] Open
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a major dose-limiting side effect that occurs in up to 63% of patients and has no known effective treatment. A majority of studies do not effectively assess sex differences in the onset and persistence of CIPN. Here we investigated the onset of CIPN, a point of therapeutic intervention where we may limit, or even prevent the development of CIPN. We hypothesized that cap-dependent translation mechanisms are important in early CIPN development and the bi-directional crosstalk between immune cells and nociceptors plays a complementary role to CIPN establishment and sex differences observed. In this study, we used wild type and eIF4E-mutant mice of both sexes to investigate the role of cap-dependent translation and the contribution of immune cells and nociceptors in the periphery and glia in the spinal cord during paclitaxel-induced peripheral neuropathy. We found that systemically administered paclitaxel induces pain-like behaviors in both sexes, increases helper T-lymphocytes, downregulates cytotoxic T-lymphocytes, and increases mitochondrial dysfunction in dorsal root ganglia neurons; all of which is eIF4E-dependent in both sexes. We identified a robust paclitaxel-induced, eIF4E-dependent increase in spinal astrocyte immunoreactivity in males, but not females. Taken together, our data reveals that cap-dependent translation may be a key pathway that presents relevant therapeutic targets during the early phase of CIPN. By targeting the eIF4E complex, we may reduce or reverse the negative effects associated with chemotherapeutic treatments.
Collapse
Affiliation(s)
- Nilesh M Agalave
- Neuroimmunology and Behavior Laboratory, Department of Neuroscience, School of Behavioral and Brain Sciences, Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, United States
| | - Prapti H Mody
- Neuroimmunology and Behavior Laboratory, Department of Neuroscience, School of Behavioral and Brain Sciences, Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, United States
| | - Thomas A Szabo-Pardi
- Neuroimmunology and Behavior Laboratory, Department of Neuroscience, School of Behavioral and Brain Sciences, Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, United States
| | - Han S Jeong
- Neuroimmunology and Behavior Laboratory, Department of Neuroscience, School of Behavioral and Brain Sciences, Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, United States
| | - Michael D Burton
- Neuroimmunology and Behavior Laboratory, Department of Neuroscience, School of Behavioral and Brain Sciences, Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, United States
| |
Collapse
|
46
|
Kavelaars A, Heijnen CJ. T Cells as Guardians of Pain Resolution. Trends Mol Med 2021; 27:302-313. [PMID: 33431239 PMCID: PMC8005447 DOI: 10.1016/j.molmed.2020.12.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 11/20/2020] [Accepted: 12/10/2020] [Indexed: 01/08/2023]
Abstract
Despite successful research efforts aimed at understanding pain mechanisms, there is still no adequate treatment for many patients suffering from chronic pain. The contribution of neuroinflammation to chronic pain is widely acknowledged. Here, we summarize findings indicating that T cells play a key role in the suppression of pain. An active contribution of the immune system to resolution of pain may explain why immunosuppressive drugs are often not sufficient to control pain. This would also imply that dysregulation of certain immune functions promote transition to chronic pain. Conversely, stimulating the endogenous immune-mediated resolution pathways may provide a potent approach to treat chronic pain.
Collapse
Affiliation(s)
- Annemieke Kavelaars
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, University of Texas, M.D. Anderson Cancer Center, Zayed Building, M.D. Anderson Boulevard, Houston, TX 77030, USA.
| | - Cobi J Heijnen
- Laboratories of Neuroimmunology, Department of Symptom Research, Division of Internal Medicine, University of Texas, M.D. Anderson Cancer Center, Zayed Building, M.D. Anderson Boulevard, Houston, TX 77030, USA.
| |
Collapse
|
47
|
Heussner MJ, Folger JK, Dias C, Massri N, Dahdah A, Vermeer PD, Laumet G. A Novel Syngeneic Immunocompetent Mouse Model of Head and Neck Cancer Pain Independent of Interleukin-1 Signaling. Anesth Analg 2021; 132:1156-1163. [PMID: 33323783 PMCID: PMC7969384 DOI: 10.1213/ane.0000000000005302] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND Pain is one of the first presenting symptoms in patients with head and neck cancer, who often develop chronic and debilitating pain as the disease progresses. Pain is also an important prognostic marker for survival. Unfortunately, patients rarely receive effective pain treatment due to our limited knowledge of the mechanisms underlying head and neck cancer pain (HNCP). Pain is often associated with neuroinflammation and particularly interleukin (IL)-1 signaling. The purpose of this study is to develop a novel syngeneic model of HNCP in immunocompetent mice to examine the contribution of IL-1 signaling. METHODS Male C57BL/6 mice were injected with a murine model of human papillomavirus (HPV+)-induced oropharyngeal squamous cell carcinoma in their right hindlimb to induce tumor growth. Pain sensitivity was measured via von Frey filaments. Spontaneous pain was assessed via the facial grimace scale. IL-1β was measured by quantifying gene expression via quantitative polymerase chain reaction (qPCR) and enzyme-linked immunosorbent assay (ELISA). RESULTS Pain hypersensitivity and spontaneous pain develop quickly after the implantation of tumor cells, a time when tumor volume is still insignificant. Spinal and circulating IL-1β levels are significantly elevated in tumor-bearing mice. Blocking IL-1 signaling either by intrathecal administration of interleukin-1 receptor antagonist (IL-1ra) or by genetic deletion (interleukin-1 receptor knockout [Il1r1-/-]) does not alleviate HNCP. CONCLUSIONS We established the first syngeneic model of HNCP in immunocompetent mice. Unlike inflammatory or nerve-injured pain, HNCP is independent of IL-1 signaling. These findings challenge the common belief that pain results from tissue compression or IL-1 signaling in patients with head and neck cancer.
Collapse
Affiliation(s)
- Matthew J. Heussner
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
- College of Osteopathic Medicine, Michigan State University, East Lansing, MI 48824, USA
| | - Joseph K. Folger
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
| | - Christina Dias
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Noura Massri
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
| | - Albert Dahdah
- Department of Microbiology & Molecular Genetics, Michigan State University, East Lansing, MI 48824, USA
| | - Paola D. Vermeer
- Cancer Biology and Immunotherapies Group, Sanford Research, Sioux Falls, SD 57104, USA
| | - Geoffroy Laumet
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
48
|
Inyang KE, George SR, Laumet G. The µ-δ opioid heteromer masks latent pain sensitization in neuropathic and inflammatory pain in male and female mice. Brain Res 2021; 1756:147298. [PMID: 33516809 DOI: 10.1016/j.brainres.2021.147298] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 01/07/2021] [Accepted: 01/11/2021] [Indexed: 01/16/2023]
Abstract
The episodic nature of chronic pain can be studied in the rodent model of latent pain sensitization. After remission, central sensitization is opposed by activation of opioid receptors. At the behavioral level, latent pain sensitization is unmasked when pain hypersensitivity is reinstated by opioid receptor (OR) antagonism. Previous studies have focused on inflammatory pain and male rodents. Whether latent pain sensitization occurs in models of chemotherapy-induced neuropathic pain in female and male mice is unknown. The first aim of this study was to investigate whether μ- and δ-OR suppress latent pain sensitization in our model of chemotherapy-induced neuropathic pain in both sexes. Mounting evidence suggests that μ-and δ-ORs form a heteromer and that the heteromer modulates pain sensitivity. Potential implications of the μ-δ OR heteromer in latent pain sensitization have not been fully explored due to a lack of tools to effectively modulate the heteromer. To specifically target the μ-δ OR heteromer, we used a specific interfering peptide blocking the heteromerization. The second aim of this study was to investigate whether disruption of the μ-δOR heteromer, after remission, reinstates pain hypersensitivity. After remission from cisplatin-induced neuropathic pain, antagonism of µ-OR and δOR reinstates pain hypersensitivity in both sexes. After remission from cisplatin-induced neuropathic pain and postoperative pain, disruption of the μ-δOR heteromer reinstates pain hypersensitivity in both sexes. Taken together our findings suggest that the μ-δOR heteromer plays a crucial role in remission in various pain models and may represent a novel therapeutic target to prevent the relapse to pain and the transition to chronic pain.
Collapse
Affiliation(s)
| | - Susan R George
- Department of Medicine and Pharmacology, University of Toronto, Toronto, Ontario, Canada
| | - Geoffroy Laumet
- Department of Physiology, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
49
|
Interleukin-10 resolves pain hypersensitivity induced by cisplatin by reversing sensory neuron hyperexcitability. Pain 2021; 161:2344-2352. [PMID: 32427749 DOI: 10.1097/j.pain.0000000000001921] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Understanding the mechanisms that drive transition from acute to chronic pain is essential to identify new therapeutic targets. The importance of endogenous resolution pathways acting as a "brake" to prevent development of chronic pain has been largely ignored. We examined the role of interleukin-10 (IL-10) in resolution of neuropathic pain induced by cisplatin. In search of an underlying mechanism, we studied the effect of cisplatin and IL-10 on spontaneous activity (SA) in dorsal root ganglia neurons. Cisplatin (2 mg/kg daily for 3 days) induced mechanical hypersensitivity that resolved within 3 weeks. In both sexes, resolution of mechanical hypersensitivity was delayed in Il10 mice, in WT mice treated intrathecally with neutralizing anti-IL-10 antibody, and in mice with cell-targeted deletion of IL-10R1 on advillin-positive sensory neurons. Electrophysiologically, small- to medium-sized dorsal root ganglia neurons from cisplatin-treated mice displayed an increase in the incidence of SA. Cisplatin treatment also depolarized the resting membrane potential, and decreased action potential voltage threshold and rheobase, while increasing ongoing activity at -45 mV and the amplitude of depolarizing spontaneous fluctuations. In vitro addition of IL-10 (10 ng/mL) reversed the effect of cisplatin on SA and on the depolarizing spontaneous fluctuation amplitudes, but unexpectedly had little effect on the other electrophysiological parameters affected by cisplatin. Collectively, our findings challenge the prevailing concept that IL-10 resolves pain solely by dampening neuroinflammation and demonstrate in a model of chemotherapy-induced neuropathic pain that endogenous IL-10 prevents transition to chronic pain by binding to IL-10 receptors on sensory neurons to regulate their activity.
Collapse
|
50
|
Boukelmoune N, Laumet G, Tang Y, Ma J, Mahant I, Nijboer C, Benders M, Kavelaars A, Heijnen CJ, Heijnen CJ. Nasal administration of mesenchymal stem cells reverses chemotherapy-induced peripheral neuropathy in mice. Brain Behav Immun 2021; 93:43-54. [PMID: 33316379 PMCID: PMC8826497 DOI: 10.1016/j.bbi.2020.12.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 12/07/2020] [Accepted: 12/08/2020] [Indexed: 12/18/2022] Open
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is one of the most frequently reported adverse effects of cancer treatment. CIPN often persists long after treatment completion and has detrimental effects on patient's quality of life. There are no efficacious FDA-approved drugs for CIPN. We recently demonstrated that nasal administration of mesenchymal stem cells (MSC) reverses the cognitive deficits induced by cisplatin in mice. Here we show that nasal administration of MSC after cisplatin- or paclitaxel treatment- completely reverses signs of established CIPN, including mechanical allodynia, spontaneous pain, and loss of intraepidermal nerve fibers (IENF) in the paw. The resolution of CIPN is associated with normalization of the cisplatin-induced decrease in mitochondrial bioenergetics in DRG neurons. Nasally administered MSC enter rapidly the meninges of the brain, spinal cord and peripheral lymph nodes to promote IL-10 production by macrophages. MSC-mediated resolution of mechanical allodynia, recovery of IENFs and restoration of DRG mitochondrial function critically depends on IL-10 production. MSC from IL-10 knockout animals are not capable of reversing the symptoms of CIPN. Moreover, WT MSC do not reverse CIPN in mice lacking IL-10 receptors on peripheral sensory neurons. In conclusion, only two nasal administrations of MSC fully reverse CIPN and the associated mitochondrial abnormalities via an IL-10 dependent pathway. Since MSC are already applied clinically, we propose that nasal MSC treatment could become a powerful treatment for the large group of patients suffering from neurotoxicities of cancer treatment.
Collapse
Affiliation(s)
- Nabila Boukelmoune
- Laboratories of Neuroimmunology, Department of Symptom Research, The University of Texas MD Anderson Cancer Center, 6565 MD Anderson Blvd, Houston, Texas, 77030, USA
| | - Geoffroy Laumet
- Laboratories of Neuroimmunology, Department of Symptom Research, The University of Texas MD Anderson Cancer Center, 6565 MD Anderson Blvd, Houston, Texas, 77030, USA.,Current affiliation: Department of Physiology, Michigan State University, East Lansing, Michigan, 48824, USA
| | - Yongfu Tang
- Laboratories of Neuroimmunology, Department of Symptom Research, The University of Texas MD Anderson Cancer Center, 6565 MD Anderson Blvd, Houston, Texas, 77030, USA
| | - Jiacheng Ma
- Laboratories of Neuroimmunology, Department of Symptom Research, The University of Texas MD Anderson Cancer Center, 6565 MD Anderson Blvd, Houston, Texas, 77030, USA
| | - Itee Mahant
- Laboratories of Neuroimmunology, Department of Symptom Research, The University of Texas MD Anderson Cancer Center, 6565 MD Anderson Blvd, Houston, Texas, 77030, USA
| | - Cora Nijboer
- Department of Developmental Origins of Disease, Division Woman and Baby, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Manon Benders
- Department of Neonatology, Division Woman and Baby, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Annemieke Kavelaars
- Laboratories of Neuroimmunology, Department of Symptom Research, The University of Texas MD Anderson Cancer Center, 6565 MD Anderson Blvd, Houston, Texas, 77030, USA
| | - Cobi J. Heijnen
- Laboratories of Neuroimmunology, Department of Symptom Research, The University of Texas MD Anderson Cancer Center, 6565 MD Anderson Blvd, Houston, Texas, 77030, USA.,Corresponding author at: Laboratories of Neuroimmunology, Department of Symptom Research, The University of Texas MD Anderson Cancer Center, 6565 MD Anderson Blvd, Z8.5034, Houston, Texas, 77030. (Cobi J. Heijnen)
| | - Cobi J Heijnen
- Laboratories of Neuroimmunology, Department of Symptom Research, The University of Texas MD Anderson Cancer Center, 6565 MD Anderson Blvd, Houston, TX 77030, USA.
| |
Collapse
|