1
|
Mikhalitskaya EV, Vyalova NM, Bokhan NA, Ivanova SA. Alcohol-Induced Activation of Chemokine System and Neuroinflammation Development. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:1889-1903. [PMID: 39647818 DOI: 10.1134/s0006297924110038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 09/27/2024] [Accepted: 09/30/2024] [Indexed: 12/10/2024]
Abstract
Chemokines are immunoregulatory proteins with pleiotropic functions involved in neuromodulation, neurogenesis, and neurotransmission. The way chemokines affect the CNS plays an important role in modulating various conditions that could have negative impact on CNS functions, including development of alcohol use disorders. In this review, we analyzed the literature data available on the problem of chemokine participation in pathogenesis, clinical presentation, and remission of alcohol use disorders both in animal models and in the study of patients with alcoholism. The presented information confirms the hypothesis that the alcohol-induced chemokine production could modulate chronic neuroinflammation. Thus, the data summarized and shown in this review are focused on the relevant direction of research in the field of psychiatry, which is in demand by both scientists and clinical specialists.
Collapse
Affiliation(s)
- Ekaterina V Mikhalitskaya
- Mental Health Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, 634014, Russia.
| | - Natalya M Vyalova
- Mental Health Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, 634014, Russia
| | - Nikolay A Bokhan
- Mental Health Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, 634014, Russia
| | - Svetlana A Ivanova
- Mental Health Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, 634014, Russia
| |
Collapse
|
2
|
He B, Wang W, Wen J, Zhang R, Lin W, Guo Y, Xu Y, Huser T, Wei X, Yang Z. Behavioral dynamics of neuroprotective macrophage polarization in neuropathic pain observed by GHz femtosecond laser two-photon excitation microscopy. JOURNAL OF BIOPHOTONICS 2024; 17:e202400026. [PMID: 38453163 DOI: 10.1002/jbio.202400026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 02/21/2024] [Accepted: 02/24/2024] [Indexed: 03/09/2024]
Abstract
Macrophage polarization in neurotoxic (M1) or neuroprotective (M2) phenotypes is known to play a significant role in neuropathic pain, but its behavioral dynamics and underlying mechanism remain largely unknown. Two-photon excitation microscopy (2PEM) is a promising functional imaging tool for investigating the mechanism of cellular behavior, as using near-infrared excitation wavelengths is less subjected to light scattering. However, the higher-order photobleaching effect in 2PEM can seriously hamper its applications to long-term live-cell studies. Here, we demonstrate a GHz femtosecond (fs) 2PEM that enables hours-long live-cell imaging of macrophage behavior with reduced higher-order photobleaching effect-by leveraging the repetition rate of fs pulses according to the fluorescence lifetime of fluorophores. Using this new functional 2PEM platform, we measure the polarization characteristics of macrophages, especially the long-term cellular behavior in efferocytosis, unveiling the dynamic mechanism of neuroprotective macrophage polarization in neuropathic pain. These efforts can create new opportunities for understanding long-term cellular dynamic behavior in neuropathic pain, as well as other neurobiological problems, and thus dissecting the underlying complex pathogenesis.
Collapse
Affiliation(s)
- Bin He
- School of Physics and Optoelectronics; State Key Laboratory of Luminescent Materials and Devices; Guangdong Engineering Technology Research and Development Center of Special Optical Fiber Materials and Devices; Guangdong Provincial Key Laboratory of Fiber Laser Materials and Applied Techniques, South China University of Technology, Guangzhou, China
- Department of Orthodontics, Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Wenlong Wang
- School of Physics and Optoelectronics; State Key Laboratory of Luminescent Materials and Devices; Guangdong Engineering Technology Research and Development Center of Special Optical Fiber Materials and Devices; Guangdong Provincial Key Laboratory of Fiber Laser Materials and Applied Techniques, South China University of Technology, Guangzhou, China
| | - Junpeng Wen
- School of Physics and Optoelectronics; State Key Laboratory of Luminescent Materials and Devices; Guangdong Engineering Technology Research and Development Center of Special Optical Fiber Materials and Devices; Guangdong Provincial Key Laboratory of Fiber Laser Materials and Applied Techniques, South China University of Technology, Guangzhou, China
| | - Runsen Zhang
- School of Physics and Optoelectronics; State Key Laboratory of Luminescent Materials and Devices; Guangdong Engineering Technology Research and Development Center of Special Optical Fiber Materials and Devices; Guangdong Provincial Key Laboratory of Fiber Laser Materials and Applied Techniques, South China University of Technology, Guangzhou, China
| | - Wei Lin
- School of Physics and Optoelectronics; State Key Laboratory of Luminescent Materials and Devices; Guangdong Engineering Technology Research and Development Center of Special Optical Fiber Materials and Devices; Guangdong Provincial Key Laboratory of Fiber Laser Materials and Applied Techniques, South China University of Technology, Guangzhou, China
| | - Yuankai Guo
- School of Physics and Optoelectronics; State Key Laboratory of Luminescent Materials and Devices; Guangdong Engineering Technology Research and Development Center of Special Optical Fiber Materials and Devices; Guangdong Provincial Key Laboratory of Fiber Laser Materials and Applied Techniques, South China University of Technology, Guangzhou, China
| | - Yue Xu
- Department of Orthodontics, Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Thomas Huser
- Biomolecular Photonics, Department of Physics, Bielefeld University, Bielefeld, Germany
| | - Xiaoming Wei
- School of Physics and Optoelectronics; State Key Laboratory of Luminescent Materials and Devices; Guangdong Engineering Technology Research and Development Center of Special Optical Fiber Materials and Devices; Guangdong Provincial Key Laboratory of Fiber Laser Materials and Applied Techniques, South China University of Technology, Guangzhou, China
| | - Zhongmin Yang
- School of Physics and Optoelectronics; State Key Laboratory of Luminescent Materials and Devices; Guangdong Engineering Technology Research and Development Center of Special Optical Fiber Materials and Devices; Guangdong Provincial Key Laboratory of Fiber Laser Materials and Applied Techniques, South China University of Technology, Guangzhou, China
- Research Institute of Future Technology, South China Normal University, Guangzhou, Guangdong, China
| |
Collapse
|
3
|
Yang H, Liu Z, Liu F, Wu H, Huang X, Huang R, Saw PE, Cao M. TET1-Lipid Nanoparticle Encapsulating Morphine for Specific Targeting of Peripheral Nerve for Pain Alleviation. Int J Nanomedicine 2024; 19:4759-4777. [PMID: 38828199 PMCID: PMC11141738 DOI: 10.2147/ijn.s453608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 05/07/2024] [Indexed: 06/05/2024] Open
Abstract
Background Opioids are irreplaceable analgesics owing to the lack of alternative analgesics that offer opioid-like pain relief. However, opioids have many undesirable central side effects. Restricting opioids to peripheral opioid receptors could reduce those effects while maintaining analgesia. Methods To achieve this goal, we developed Tet1-LNP (morphine), a neural-targeting lipid nanoparticle encapsulating morphine that could specifically activate the peripheral opioid receptor in the dorsal root ganglion (DRG) and significantly reduce the side effects caused by the activation of opioid receptors in the brain. Tet1-LNP (morphine) were successfully prepared using the thin-film hydration method. In vitro, Tet1-LNP (morphine) uptake was assessed in differentiated neuron-like PC-12 cells and dorsal root ganglion (DRG) primary cells. The uptake of Tet1-LNP (morphine) in the DRGs and the brain was assessed in vivo. Von Frey filament and Hargreaves tests were used to assess the antinociception of Tet1-LNP (morphine) in the chronic constriction injury (CCI) neuropathic pain model. Morphine concentration in blood and brain were evaluated using ELISA. Results Tet1-LNP (morphine) had an average size of 131 nm. Tet1-LNP (morphine) showed high cellular uptake and targeted DRG in vitro. CCI mice treated with Tet1-LNP (morphine) experienced prolonged analgesia for nearly 32 h compared with 3 h with free morphine (p < 0.0001). Notably, the brain morphine concentration in the Tet1-LNP (morphine) group was eight-fold lower than that in the morphine group (p < 0.0001). Conclusion Our study presents a targeted lipid nanoparticle system for peripheral neural delivery of morphine. We anticipate Tet1-LNP (morphine) will offer a safe formulation for chronic neuropathic pain treatment, and promise further development for clinical applications.
Collapse
Affiliation(s)
- Hongmei Yang
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People’s Republic of China
| | - Zhongqi Liu
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People’s Republic of China
| | - Fan Liu
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People’s Republic of China
- Department of Anesthesiology, Shenshan Medical Center, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Shanwei, 516600, People’s Republic of China
| | - Haixuan Wu
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People’s Republic of China
| | - Xiaoyan Huang
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People’s Republic of China
| | - Rong Huang
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People’s Republic of China
| | - Phei Er Saw
- Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People’s Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People’s Republic of China
| | - Minghui Cao
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People’s Republic of China
- Department of Anesthesiology, Shenshan Medical Center, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Shanwei, 516600, People’s Republic of China
| |
Collapse
|
4
|
Li S, Tang S, Dai L, Jian Z, Li X. Emodin relieves morphine-stimulated BV2 microglial activation and inflammation through the TLR4/NF-κB/NLRP3 pathway. Neuroreport 2024; 35:518-528. [PMID: 38597275 DOI: 10.1097/wnr.0000000000002034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2024]
Abstract
The objective of this study is to disclose the role of emodin, a natural anthraquinone derivative that has been proposed to suppress microglial activation and inflammation, in morphine tolerance. Here, cell counting kit-8 method assayed the viability of BV2 microglial cells treated by ascending concentrations of emodin. In emodin-pretreated BV2 microglial cells challenged with morphine with or without transfection of toll-like receptor 4 (TLR4) overexpression plasmids, transwell assay measured cell migration. Immunofluorescence staining and western blot detected the expression of microglial markers. Inflammatory levels were subjected to ELISA and western blot. BODIPY 581/591 C11 assay estimated lipid reactive oxygen species activity. Iron assay kit examined total iron content. Western blot tested the expression of ferroptosis- and TLR4/nuclear factor-kappaB (NF-κB)/NOD-like receptor 3 (NLRP3) pathway-associated proteins. Molecular docking predicted the binding affinity of emodin to TLR4. Emodin was noted to obstruct the migration, activation, inflammatory response, and ferroptosis of BV2 microglial cells induced by morphine. In addition, emodin had a high binding affinity with TLR4 and inactivated TLR4/NF-κB/NLRP3 pathway in morphine-challenged BV2 microglial cells. Upregulation of TLR4 partially countervailed the protective role of emodin against morphine-elicited BV2 microglial cell migration, activation, inflammation, and ferroptosis. Accordingly, emodin might target TLR4 and act as an inactivator of TLR4/NF-κB/NLRP3 pathway, thus inhibiting BV2 microglial activation and inflammation to mitigate morphine tolerance.
Collapse
Affiliation(s)
- Shimei Li
- Department of Anesthesiology, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou Province, China
| | | | | | | | | |
Collapse
|
5
|
Goh MPY, Samsul RN, Mohaimin AW, Goh HP, Zaini NH, Kifli N, Ahmad N. The Analgesic Potential of Litsea Species: A Systematic Review. Molecules 2024; 29:2079. [PMID: 38731572 PMCID: PMC11085224 DOI: 10.3390/molecules29092079] [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: 04/06/2024] [Revised: 04/25/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Various plant species from the Litsea genus have been claimed to be beneficial for pain relief. The PRISMA approach was adopted to identify studies that reported analgesic properties of plants from the Litsea genus. Out of 450 records returned, 19 primary studies revealed the analgesic potential of nine Litsea species including (1) Litsea cubeba, (2) Litsea elliptibacea, (3) Litsea japonica, (4) Litsea glutinosa, (5) Litsea glaucescens, (6) Litsea guatemalensis, (7) Litsea lancifolia, (8) Litsea liyuyingi and (9) Litsea monopetala. Six of the species, 1, 3, 4, 7, 8 and 9, demonstrated peripheral antinociceptive properties as they inhibited acetic-acid-induced writhing in animal models. Species 1, 3, 4, 8 and 9 further showed effects via the central analgesic route at the spinal level by increasing the latencies of heat stimulated-nocifensive responses in the tail flick assay. The hot plate assay also revealed the efficacies of 4 and 9 at the supraspinal level. Species 6 was reported to ameliorate hyperalgesia induced via partial sciatic nerve ligation (PSNL). The antinociceptive effects of 1 and 3 were attributed to the regulatory effects of their bioactive compounds on inflammatory mediators. As for 2 and 5, their analgesic effect may be a result of their activity with the 5-hydroxytryptamine 1A receptor (5-HT1AR) which disrupted the pain-stimulating actions of 5-HT. Antinociceptive activities were documented for various major compounds of the Litsea plants. Overall, the findings suggested Litsea species as good sources of antinociceptive compounds that can be further developed to complement or substitute prescription drugs for pain management.
Collapse
Affiliation(s)
- May Poh Yik Goh
- Herbal Research Group, Universiti Brunei Darussalam, Jalan Tungku Link, Gadong, Bandar Seri Begawan BE 1410, Brunei; (M.P.Y.G.); (R.N.S.); (A.W.M.); (N.K.)
- PAP Rashidah Saádatul Bolkiah Institute of Health Sciences, Universiti Brunei Darussalam, Jalan Tungku Link, Gadong, Bandar Seri Begawan BE 1410, Brunei;
| | - Raudhatun Na’emah Samsul
- Herbal Research Group, Universiti Brunei Darussalam, Jalan Tungku Link, Gadong, Bandar Seri Begawan BE 1410, Brunei; (M.P.Y.G.); (R.N.S.); (A.W.M.); (N.K.)
- Environmental and Life Sciences, Faculty of Science, Universiti Brunei Darussalam, Jalan Tungku Link, Gadong, Bandar Seri Begawan BE 1410, Brunei
| | - Amal Widaad Mohaimin
- Herbal Research Group, Universiti Brunei Darussalam, Jalan Tungku Link, Gadong, Bandar Seri Begawan BE 1410, Brunei; (M.P.Y.G.); (R.N.S.); (A.W.M.); (N.K.)
- Environmental and Life Sciences, Faculty of Science, Universiti Brunei Darussalam, Jalan Tungku Link, Gadong, Bandar Seri Begawan BE 1410, Brunei
| | - Hui Poh Goh
- PAP Rashidah Saádatul Bolkiah Institute of Health Sciences, Universiti Brunei Darussalam, Jalan Tungku Link, Gadong, Bandar Seri Begawan BE 1410, Brunei;
| | - Nurul Hazlina Zaini
- UBD Botanical Research Centre, Institute for Biodiversity and Environmental Research, Universiti Brunei Darussalam, Jalan Tungku Link, Gadong, Bandar Seri Begawan BE 1410, Brunei;
| | - Nurolaini Kifli
- Herbal Research Group, Universiti Brunei Darussalam, Jalan Tungku Link, Gadong, Bandar Seri Begawan BE 1410, Brunei; (M.P.Y.G.); (R.N.S.); (A.W.M.); (N.K.)
- PAP Rashidah Saádatul Bolkiah Institute of Health Sciences, Universiti Brunei Darussalam, Jalan Tungku Link, Gadong, Bandar Seri Begawan BE 1410, Brunei;
| | - Norhayati Ahmad
- Herbal Research Group, Universiti Brunei Darussalam, Jalan Tungku Link, Gadong, Bandar Seri Begawan BE 1410, Brunei; (M.P.Y.G.); (R.N.S.); (A.W.M.); (N.K.)
- Environmental and Life Sciences, Faculty of Science, Universiti Brunei Darussalam, Jalan Tungku Link, Gadong, Bandar Seri Begawan BE 1410, Brunei
- UBD Botanical Research Centre, Institute for Biodiversity and Environmental Research, Universiti Brunei Darussalam, Jalan Tungku Link, Gadong, Bandar Seri Begawan BE 1410, Brunei;
| |
Collapse
|
6
|
Bi K, Lei Y, Kong D, Li Y, Fan X, Luo X, Yang J, Wang G, Li X, Xu Y, Luo H. Progress in the study of intestinal microbiota involved in morphine tolerance. Heliyon 2024; 10:e27187. [PMID: 38533077 PMCID: PMC10963202 DOI: 10.1016/j.heliyon.2024.e27187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 01/09/2024] [Accepted: 02/26/2024] [Indexed: 03/28/2024] Open
Abstract
Morphine is a widely used opioid for treatment of pain. The attendant problems including morphine tolerance and morphine dependence pose a major public health challenge. In recent years, there has been increasing interest in the gastrointestinal microbiota in many physiological and pathophysiological processes. The connectivity network between the gut microbiota and the brain is involved in multiple biological systems, and bidirectional communication between them is critical in gastrointestinal tract homeostasis, the central nervous system, and the microbial system. Many research have previously shown that morphine has a variety of effects on the gastrointestinal tract, but none have determined the function of intestinal microbiota in morphine tolerance. This study reviewed the mechanisms of morphine tolerance from the perspective of dysregulation of microbiota-gut-brain axis homeostasis, by summarizing the possible mechanisms originating from the gut that may affect morphine tolerance and the improvement of morphine tolerance through the gut microbiota.
Collapse
Affiliation(s)
- Ke Bi
- Department of Gastrointestinal and Hernia Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
- Yunnan Technological Innovation Centre of Drug Addiction Medicine, Yunnan University, Kunming, 650032, China
| | - Yi Lei
- Department of Gastrointestinal and Hernia Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Deshenyue Kong
- Department of Gastrointestinal and Hernia Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
- Yunnan Technological Innovation Centre of Drug Addiction Medicine, Yunnan University, Kunming, 650032, China
| | - Yuansen Li
- Department of Gastrointestinal and Hernia Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
- Yunnan Technological Innovation Centre of Drug Addiction Medicine, Yunnan University, Kunming, 650032, China
| | - Xuan Fan
- Department of Gastrointestinal and Hernia Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
- Yunnan Technological Innovation Centre of Drug Addiction Medicine, Yunnan University, Kunming, 650032, China
| | - Xiao Luo
- Department of Gastrointestinal and Hernia Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
- Yunnan Technological Innovation Centre of Drug Addiction Medicine, Yunnan University, Kunming, 650032, China
| | - Jiqun Yang
- Third People's Hospital of Kunming City/Drug Rehabilitation Hospital of Kunming City, Kunming, 650041, China
| | - Guangqing Wang
- Drug Rehabilitation Administration of Yunnan Province, Kunming, 650032, China
| | - Xuejun Li
- Drug Rehabilitation Administration of Yunnan Province, Kunming, 650032, China
| | - Yu Xu
- Department of Gastrointestinal and Hernia Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Huayou Luo
- Department of Gastrointestinal and Hernia Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| |
Collapse
|
7
|
Han S, Gao J, Wang Z, Xiao Y, Ge Y, Liang Y, Gao J. Genetically supported causality between gut microbiota, immune cells and morphine tolerance: a two-sample Mendelian randomization study. Front Microbiol 2024; 15:1343763. [PMID: 38389539 PMCID: PMC10882271 DOI: 10.3389/fmicb.2024.1343763] [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: 11/27/2023] [Accepted: 01/29/2024] [Indexed: 02/24/2024] Open
Abstract
Background Previous researches have suggested a significant connection between the gut microbiota/immune cells and morphine tolerance (MT), but there is still uncertainty regarding their causal relationship. Hence, our objective is to inverstigate this causal association and reveal the impact of gut microbiota/immune cells on the risk of developing MT using a two-sample Mendelian randomization (MR) study. Methods We conducted a comprehensive analysis using genome-wide association study (GWAS) summary statistics for gut microbiota, immune cells, and MT. The main approach employed was the inverse variance-weighted (IVW) method in MR. To assess horizontal pleiotropy and remove outlier single-nucleotide polymorphisms (SNPs), we utilized the Mendelian randomization pleiotropy residual sum and outlier (MR-PRESSO) technique as well as MR-Egger regression. Heterogeneity detection was performed using Cochran's Q-test. Additionally, leave-one-out analysis was carried out to determine if any single SNP drove the causal association signals. Finally, we conducted a reverse MR to evaluate the potential of reverse causation. Results We discovered that 6 gut microbial taxa and 16 immune cells were causally related to MT (p < 0.05). Among them, 2 bacterial features and 9 immunophenotypes retained a strong causal relationship with lower risk of MT: genus. Lachnospiraceae NK4A136group (OR: 0.962, 95% CI: 0.940-0.987, p = 0.030), genus. RuminococcaceaeUCG011 (OR: 0.960, 95% CI: 0.946-0.976, p = 0.003), BAFF-R on B cell (OR: 0.972, 95% CI: 0.947-0.998, p = 0.013). Furthermore, 4 bacterial features and 7 immunophenotypes were identified to be significantly associated with MT risk: genus. Flavonifractor (OR: 1.044, 95% CI: 1.017-1.069, p = 0.029), genus. Prevotella9 (OR: 1.054, 95% CI: 1.020-1.090, p = 0.037), B cell % CD3-lymphocyte (OR: 1.976, 95% CI: 1.027-1.129, p = 0.026). The Cochrane's Q test revealed no heterogeneity (p > 0.05). Furthermore, the MR-Egger and MR-PRESSO analyses reveal no instances of horizontal pleiotropy (p > 0.05). Besides, leave-one-out analysis confirmed the robustness of MR results. After adding BMI to the multivariate MR analysis, the gut microbial taxa and immune cells exposure-outcome effect were attenuated. Conclusion Our research confirm the potential link between gut microbiota and immune cells with MT, shedding light on the mechanism by which gut microbiota and immune cells may contribute to MT. These findings lay the groundwork for future investigations into targeted prevention strategies.
Collapse
Affiliation(s)
- Shuai Han
- Department of Anesthesiology, Northern Jiangsu People’s Hospital, Clinical Medical School, Yangzhou University, Yangzhou, China
- Yangzhou University Medical College, Yangzhou, China
| | - Jiapei Gao
- Yangzhou University Medical College, Yangzhou, China
| | - Zi Wang
- Department of Anesthesiology, Northern Jiangsu People’s Hospital, Clinical Medical School, Yangzhou University, Yangzhou, China
- Yangzhou University Medical College, Yangzhou, China
| | - Yinggang Xiao
- Department of Anesthesiology, Northern Jiangsu People’s Hospital, Clinical Medical School, Yangzhou University, Yangzhou, China
- Yangzhou University Medical College, Yangzhou, China
| | - Yali Ge
- Department of Anesthesiology, Northern Jiangsu People’s Hospital, Clinical Medical School, Yangzhou University, Yangzhou, China
- Yangzhou University Medical College, Yangzhou, China
| | - Yongxin Liang
- Department of Anesthesiology, Women’s and Children’s Hospital Affiliated to Qingdao University, Qingdao, China
| | - Ju Gao
- Department of Anesthesiology, Northern Jiangsu People’s Hospital, Clinical Medical School, Yangzhou University, Yangzhou, China
- Yangzhou University Medical College, Yangzhou, China
| |
Collapse
|
8
|
Fuller AM, Bharde S, Sikandar S. The mechanisms and management of persistent postsurgical pain. FRONTIERS IN PAIN RESEARCH 2023; 4:1154597. [PMID: 37484030 PMCID: PMC10357043 DOI: 10.3389/fpain.2023.1154597] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 06/16/2023] [Indexed: 07/25/2023] Open
Abstract
An estimated 10%-50% of patients undergoing a surgical intervention will develop persistent postsurgical pain (PPP) lasting more than 3 months despite adequate acute pain management and the availability of minimally invasive procedures. The link between early and late pain outcomes for surgical procedures remains unclear-some patients improve while others develop persistent pain. The elective nature of a surgical procedure offers a unique opportunity for prophylactic or early intervention to prevent the development of PPP and improve our understanding of its associated risk factors, such as pre-operative anxiety and the duration of severe acute postoperative pain. Current perioperative pain management strategies often include opioids, but long-term consumption can lead to tolerance, addiction, opioid-induced hyperalgesia, and death. Pre-clinical models provide the opportunity to dissect mechanisms underpinning the transition from acute to chronic, or persistent, postsurgical pain. This review highlights putative mechanisms of PPP, including sensitisation of peripheral sensory neurons, neuroplasticity in the central nervous system and nociceptive signalling along the neuro-immune axis.
Collapse
|
9
|
Kim MW, Kipnis J. Picking a (neuroimmune) fight against fragile regulation of addiction. Cell 2023; 186:464-466. [PMID: 36693375 PMCID: PMC10205172 DOI: 10.1016/j.cell.2023.01.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 12/30/2022] [Accepted: 01/03/2023] [Indexed: 01/24/2023]
Abstract
T cells and their derived cytokines have been shown to modulate brain function. In this issue of Cell, Zhu, Yan, and colleagues demonstrate that opioid use impacts the crosstalk between the CNS and the peripheral immune system. Regulatory T cell (Treg)-derived IFN-γ signaling translates into synaptic weakening in the nucleus accumbens (NAc) to impart withdrawal-induced behavioral dysfunction.
Collapse
Affiliation(s)
- Min Woo Kim
- Center for Brain Immunology and Glia (BIG), Medical Scientist Training Program, Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO, USA
| | - Jonathan Kipnis
- Center for Brain Immunology and Glia (BIG), Medical Scientist Training Program, Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
10
|
Lee M, Joehanes R, McCartney DL, Kho M, Hüls A, Wyss AB, Liu C, Walker RM, R Kardia SL, Wingo TS, Burkholder A, Ma J, Campbell A, Wingo AP, Huan T, Sikdar S, Keshawarz A, Bennett DA, Smith JA, Evans KL, Levy D, London SJ. Opioid medication use and blood DNA methylation: epigenome-wide association meta-analysis. Epigenomics 2022; 14:1479-1492. [PMID: 36700736 PMCID: PMC9979153 DOI: 10.2217/epi-2022-0353] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 01/13/2023] [Indexed: 01/27/2023] Open
Abstract
Aim: To identify differential methylation related to prescribed opioid use. Methods: This study examined whether blood DNA methylation, measured using Illumina arrays, differs by recent opioid medication use in four population-based cohorts. We meta-analyzed results (282 users; 10,560 nonusers) using inverse-variance weighting. Results: Differential methylation (false discovery rate <0.05) was observed at six CpGs annotated to the following genes: KIAA0226, CPLX2, TDRP, RNF38, TTC23 and GPR179. Integrative epigenomic analyses linked implicated loci to regulatory elements in blood and/or brain. Additionally, 74 CpGs were differentially methylated in males or females. Methylation at significant CpGs correlated with gene expression in blood and/or brain. Conclusion: This study identified DNA methylation related to opioid medication use in general populations. The results could inform the development of blood methylation biomarkers of opioid use.
Collapse
Affiliation(s)
- Mikyeong Lee
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Roby Joehanes
- Department of Health and Human Services, Framingham Heart Study, National Heart, Lung, and Blood Institute, National Institutes of Health, Framingham, MA 01702, USA
| | - Daniel L McCartney
- Centre for Genomic & Experimental Medicine, Institute of Genetics & Cancer, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh, UK
| | - Minjung Kho
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - Anke Hüls
- Department of Epidemiology & Gangarosa, Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA
| | - Annah B Wyss
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Chunyu Liu
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA 02215, USA
- Framingham Heart Study, Boston University, Framingham, MA 01702, USA
| | - Rosie M Walker
- Centre for Clinical Brain Science, Chancellor's Building, 49 Little France Crescent, Edinburgh Bioquarter, Edinburgh, UK
- School of Psychology, University of Exeter, Exeter, UK
| | - Sharon L R Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - Thomas S Wingo
- Department of Neurology & Human Genetics, Emory University, Atlanta, GA 30322, USA
| | - Adam Burkholder
- Office of Environmental Science Cyberinfrastructure, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Jiantao Ma
- Department of Health and Human Services, Framingham Heart Study, National Heart, Lung, and Blood Institute, National Institutes of Health, Framingham, MA 01702, USA
| | - Archie Campbell
- Centre for Genomic & Experimental Medicine, Institute of Genetics & Cancer, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh, UK
| | - Aliza P Wingo
- Department of Psychiatry & Behavioral Sciences, Emory University, Atlanta, GA 30322, USA
| | - Tianxiao Huan
- Department of Health and Human Services, Framingham Heart Study, National Heart, Lung, and Blood Institute, National Institutes of Health, Framingham, MA 01702, USA
- Department of Ophthalmology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Sinjini Sikdar
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
- Department of Mathematics & Statistics, Old Dominion University, Norfolk, VA 23529, USA
| | - Amena Keshawarz
- Framingham Heart Study, Framingham, MA 01702, USA
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
| | - Jennifer A Smith
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kathryn L Evans
- Centre for Genomic & Experimental Medicine, Institute of Genetics & Cancer, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh, UK
| | - Daniel Levy
- Department of Health and Human Services, Framingham Heart Study, National Heart, Lung, and Blood Institute, National Institutes of Health, Framingham, MA 01702, USA
| | - Stephanie J London
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| |
Collapse
|
11
|
Bongiovanni AR, Zhao P, Inan S, Wiah S, Shekarabi A, Farkas DJ, Watson MN, Wimmer ME, Ruff MR, Rawls SM. Multi-chemokine receptor antagonist RAP-103 inhibits opioid-derived respiratory depression, reduces opioid reinforcement and physical dependence, and normalizes opioid-induced dysregulation of mesolimbic chemokine receptors in rats. Drug Alcohol Depend 2022; 238:109556. [PMID: 35843139 PMCID: PMC9444981 DOI: 10.1016/j.drugalcdep.2022.109556] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/20/2022] [Accepted: 06/29/2022] [Indexed: 11/03/2022]
Abstract
Chemokine-opioid crosstalk is a physiological crossroads for influencing therapeutic and adverse effects of opioids. Activation of chemokine receptors, especially CCR2, CCR5 and CXCR4, reduces opioid-induced analgesia by desensitizing OPRM1 receptors. Chemokine receptor antagonists (CRAs) enhance opioid analgesia, but knowledge about how CRAs impact adverse opioid effects remains limited. We examined effects of RAP-103, a multi-CRA orally active peptide analog of "DAPTA", on opioid-derived dependence, reinforcement, and respiratory depression in male rats and on changes in chemokine and OPRM1 (µ opioid) receptor levels in mesolimbic substrates during opioid abstinence. In rats exposed to chronic morphine (75 mg pellet x 7 d), daily RAP-103 (1 mg/kg, IP) treatment reduced the severity of naloxone-precipitated withdrawal responses. For self-administration (SA) studies, RAP-103 (1 mg/kg, IP) reduced heroin acquisition (0.1 mg/kg/inf) and reinforcing efficacy (assessed by motivation on a progressive-ratio reinforcement schedule) but did not impact sucrose intake. RAP-103 (1-3 mg/kg, IP) also normalized the deficits in oxygen saturation and enhancement of respiratory rate caused by morphine (5 mg/kg, SC) exposure. Abstinence from chronic morphine elicited brain-region specific changes in chemokine receptor protein levels. CCR2 and CXCR4 were increased in the ventral tegmental area (VTA), whereas CCR2 and CCR5 were reduced in the nucleus accumbens (NAC). Effects of RAP-103 (1 mg/kg, IP) were focused in the NAC, where it normalized morphine-induced deficits in CCR2 and CCR5. These results identify CRAs as potential biphasic function opioid signaling modulators to enhance opioid analgesia and inhibit opioid-derived dependence and respiratory depression.
Collapse
Affiliation(s)
- Angela R Bongiovanni
- Department of Psychology and Neuroscience, Temple University, Philadelphia, PA, USA
| | - Pingwei Zhao
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Saadet Inan
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Sonita Wiah
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Aryan Shekarabi
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Daniel J Farkas
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Mia N Watson
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Mathieu E Wimmer
- Department of Psychology and Neuroscience, Temple University, Philadelphia, PA, USA
| | | | - Scott M Rawls
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA; Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.
| |
Collapse
|
12
|
Wang Y, Kang H, Jin M, Wang G, Ma W, Liu Z, Xue Y, Li C. Phenotypic and Transcriptomics Analyses Reveal Underlying Mechanisms in a Mouse Model of Corneal Bee Sting. Toxins (Basel) 2022; 14:toxins14070468. [PMID: 35878206 PMCID: PMC9323056 DOI: 10.3390/toxins14070468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/07/2022] [Accepted: 07/07/2022] [Indexed: 11/23/2022] Open
Abstract
Corneal bee sting (CBS) is one of the most common ocular traumas and can lead to blindness. The ophthalmic manifestations are caused by direct mechanical effects of bee stings, toxic effects, and host immune responses to bee venom (BV); however, the underlying pathogenesis remains unclear. Clinically, topical steroids and antibiotics are routinely used to treat CBS patients but the specific drug targets are unknown; therefore, it is imperative to study the pathological characteristics, injury mechanisms, and therapeutic targets involved in CBS. In the present study, a CBS injury model was successfully established by injecting BV into the corneal stroma of healthy C57BL/6 mice. F-actin staining revealed corneal endothelial cell damage, decreased density, skeletal disorder, and thickened corneal stromal. The terminal-deoxynucleotidyl transferase mediated nick end labeling (TUNEL) assay showed apoptosis of both epithelial and endothelial cells. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis showed that cytokine–cytokine interactions were the most relevant pathway for pathogenesis. Protein–protein interaction (PPI) network analysis showed that IL-1, TNF, and IL-6 were the most relevant nodes. RNA-seq after the application of Tobradex® (0.3% tobramycin and 0.1% dexamethasone) eye ointment showed that Tobradex® not only downregulated relevant inflammatory factors but also reduced corneal pain as well as promoted nerve regeneration by repairing axons. Here, a stable and reliable model of CBS injury was successfully established for the first time, and the pathogenesis of CBS and the therapeutic targets of Tobradex® are discussed. These hub genes are expected to be biomarkers and therapeutic targets for the diagnosis and treatment of CBS.
Collapse
Affiliation(s)
- Yanzi Wang
- Eye Institute & Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen 361102, China; (Y.W.); (H.K.); (M.J.); (Z.L.)
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Honghua Kang
- Eye Institute & Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen 361102, China; (Y.W.); (H.K.); (M.J.); (Z.L.)
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Mengyi Jin
- Eye Institute & Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen 361102, China; (Y.W.); (H.K.); (M.J.); (Z.L.)
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Guoliang Wang
- School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China;
| | - Weifang Ma
- Department of Ophthalmology, No.4 West China Teaching Hospital, Sichuan University, Chengdu 610041, China;
| | - Zhen Liu
- Eye Institute & Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen 361102, China; (Y.W.); (H.K.); (M.J.); (Z.L.)
| | - Yuhua Xue
- School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China;
- Correspondence: (Y.X.); (C.L.); Tel./Fax: +86-592-2189698 (Y.X.)
| | - Cheng Li
- Eye Institute & Affiliated Xiamen Eye Center, School of Medicine, Xiamen University, Xiamen 361102, China; (Y.W.); (H.K.); (M.J.); (Z.L.)
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Xiamen 361102, China
- Correspondence: (Y.X.); (C.L.); Tel./Fax: +86-592-2189698 (Y.X.)
| |
Collapse
|
13
|
Vincenzi M, Milella MS, D’Ottavio G, Caprioli D, Reverte I, Maftei D. Targeting Chemokines and Chemokine GPCRs to Enhance Strong Opioid Efficacy in Neuropathic Pain. Life (Basel) 2022; 12:life12030398. [PMID: 35330149 PMCID: PMC8955776 DOI: 10.3390/life12030398] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/06/2022] [Accepted: 03/07/2022] [Indexed: 12/21/2022] Open
Abstract
Neuropathic pain (NP) originates from an injury or disease of the somatosensory nervous system. This heterogeneous origin and the possible association with other pathologies make the management of NP a real challenge. To date, there are no satisfactory treatments for this type of chronic pain. Even strong opioids, the gold-standard analgesics for nociceptive and cancer pain, display low efficacy and the paradoxical ability to exacerbate pain sensitivity in NP patients. Mounting evidence suggests that chemokine upregulation may be a common mechanism driving NP pathophysiology and chronic opioid use-related consequences (analgesic tolerance and hyperalgesia). Here, we first review preclinical studies on the role of chemokines and chemokine receptors in the development and maintenance of NP. Second, we examine the change in chemokine expression following chronic opioid use and the crosstalk between chemokine and opioid receptors. Then, we examine the effects of inhibiting specific chemokines or chemokine receptors as a strategy to increase opioid efficacy in NP. We conclude that strong opioids, along with drugs that block specific chemokine/chemokine receptor axis, might be the right compromise for a favorable risk/benefit ratio in NP management.
Collapse
Affiliation(s)
- Martina Vincenzi
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, 00185 Rome, Italy;
- Correspondence: (M.V.); (I.R.)
| | - Michele Stanislaw Milella
- Toxicology and Poison Control Center Unit, Department of Emergency, Anesthesia and Critical Care, Policlinico Umberto I Hospital-Sapienza University of Rome, 00161 Rome, Italy;
| | - Ginevra D’Ottavio
- Santa Lucia Foundation (IRCCS Fondazione Santa Lucia), 00143 Rome, Italy; (G.D.); (D.C.)
- Laboratory Affiliated to Institute Pasteur Italia-Fondazione Cenci Bolognetti, Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, 00185 Rome, Italy
| | - Daniele Caprioli
- Santa Lucia Foundation (IRCCS Fondazione Santa Lucia), 00143 Rome, Italy; (G.D.); (D.C.)
- Laboratory Affiliated to Institute Pasteur Italia-Fondazione Cenci Bolognetti, Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, 00185 Rome, Italy
| | - Ingrid Reverte
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, 00185 Rome, Italy;
- Santa Lucia Foundation (IRCCS Fondazione Santa Lucia), 00143 Rome, Italy; (G.D.); (D.C.)
- Correspondence: (M.V.); (I.R.)
| | - Daniela Maftei
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, 00185 Rome, Italy;
- Santa Lucia Foundation (IRCCS Fondazione Santa Lucia), 00143 Rome, Italy; (G.D.); (D.C.)
| |
Collapse
|
14
|
Liu W, Jiang P, Qiu L. Blocking of Caveolin-1 Attenuates Morphine-Induced Inflammation, Hyperalgesia, and Analgesic Tolerance via Inhibiting NLRP3 Inflammasome and ERK/c-JUN Pathway. J Mol Neurosci 2022; 72:1047-1057. [PMID: 35262905 DOI: 10.1007/s12031-022-01989-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 02/23/2022] [Indexed: 12/21/2022]
Abstract
Morphine is generally used to treat chronic pain in clinic. But long-term use of morphine can inevitably induce analgesic tolerance and hyperalgesia. Caveolin-1 is reported to affect morphine-mediated signaling transduction. However, the action mechanism of morphine-induced analgesic tolerance is still unknown. In this study, morphine-induced analgesic tolerance model was established in Sprague-Dawley rats. The effects of Caveolin-1 blocking on neuroinflammation and ERK/c-JUN pathway were then explored. Morphine can remarkably elevate the expression level of Caveolin-1. Based on paw withdrawal latency behavior test, we found that Caveolin-1 blocking can effectively attenuate morphine-induced analgesic tolerance and neuroinflammation. Activation of ERK/c-JUN significantly reversed the above influences caused by Caveolin-1 blocking. Taken together, blocking of Caveolin-1 can attenuate morphine-induced inflammation and analgesic tolerance through inhibiting NLRP3 inflammasome and ERK/c-JUN pathway.
Collapse
Affiliation(s)
- Wenling Liu
- Department of Anestyesiology, HuiZhou Municipal Central Hospital, No. 41, Eling North Road, Huizhou City, Guangdong Province, 516001, China
| | - Peng Jiang
- Department of Anestyesiology, HuiZhou Municipal Central Hospital, No. 41, Eling North Road, Huizhou City, Guangdong Province, 516001, China
| | - Liuji Qiu
- Department of Anestyesiology, HuiZhou Municipal Central Hospital, No. 41, Eling North Road, Huizhou City, Guangdong Province, 516001, China.
| |
Collapse
|
15
|
Ma H, Li M, Pagare PP, Wang H, Nassehi N, Santos EJ, Negus SS, Selley DE, Zhang Y. Novel bivalent ligands carrying potential antinociceptive effects by targeting putative mu opioid receptor and chemokine receptor CXCR4 heterodimers. Bioorg Chem 2022; 120:105641. [PMID: 35093692 PMCID: PMC9187593 DOI: 10.1016/j.bioorg.2022.105641] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 01/12/2022] [Accepted: 01/21/2022] [Indexed: 01/26/2023]
Abstract
The functional interactions between opioid and chemokine receptors have been implicated in the pathological process of chronic pain. Mounting studies have indicated the possibility that a MOR-CXCR4 heterodimer may be involved in nociception and related pharmacologic effects. Herein we have synthesized a series of bivalent ligands containing both MOR agonist and CXCR4 antagonist pharmacophores with an aim to investigate the functional interactions between these two receptors. In vitro studies demonstrated reasonable recognition of designed ligands at both respective receptors. Further antinociceptive testing in mice revealed compound 1a to be the most promising member of this series. Additional molecular modeling studies corroborated the findings observed. Taken together, we identified the first bivalent ligand 1a showing promising antinociceptive effect by targeting putative MOR-CXCR4 heterodimers, which may serve as a novel chemical probe to further develop more potent bivalent ligands with potential application in analgesic therapies for chronic pain management.
Collapse
Affiliation(s)
- Hongguang Ma
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, 800 E Leigh Street, Richmond, Virginia 23298, United States
| | - Mengchu Li
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, 800 E Leigh Street, Richmond, Virginia 23298, United States
| | - Piyusha P. Pagare
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, 800 E Leigh Street, Richmond, Virginia 23298, United States
| | - Huiqun Wang
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, 800 E Leigh Street, Richmond, Virginia 23298, United States
| | - Nima Nassehi
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 410 North 12th Street, Richmond, Virginia 23298, United States
| | - Edna J. Santos
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 410 North 12th Street, Richmond, Virginia 23298, United States
| | - S. Stevens Negus
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 410 North 12th Street, Richmond, Virginia 23298, United States
| | - Dana E. Selley
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 410 North 12th Street, Richmond, Virginia 23298, United States
| | - Yan Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, 800 E Leigh Street, Richmond, VA 23298, United States.
| |
Collapse
|
16
|
Wan L, Jia RM, Ji LL, Qin XM, Hu L, Hu F, Han Y, Pan YB, Jiang CY, Liu WT. AMPK-autophagy-mediated inhibition of microRNA-30a-5p alleviates morphine tolerance via SOCS3-dependent neuroinflammation suppression. J Neuroinflammation 2022; 19:25. [PMID: 35093117 PMCID: PMC8800317 DOI: 10.1186/s12974-022-02384-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 01/10/2022] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The development of morphine tolerance is a clinical challenge for managing severe pain. Studies have shown that neuroinflammation is a critical aspect for the development of analgesic tolerance. We found that AMPK-autophagy activation could suppress neuroinflammation and improve morphine tolerance via the upregulation of suppressor of cytokine signaling 3 (SOCS3) by inhibiting the processing and maturation of microRNA-30a-5p. METHODS CD-1 mice were utilized for the tail-flick test to evaluate morphine tolerance. The microglial cell line BV-2 was utilized to investigate the mechanism of AMPK-autophagy-mediated posttranscriptional regulation of SOCS3. Proinflammatory cytokines were measured by western blotting and real-time PCR. The levels of SOCS3 and miRNA-processing enzymes were evaluated by western blotting, real-time PCR and immunofluorescence staining. RESULTS Based on experimental verification, miRNA-30a-5p could negatively regulate SOCS3. The AMPK activators AICAR, resveratrol and metformin downregulated miRNA-30a-5p. We found that AMPK activators specifically inhibited the processing and maturation of miRNA-30a-5p in microglia by degrading DICER and AGO2 via autophagy. Furthermore, a miRNA-30a-5p inhibitor significantly improved morphine tolerance via upregulation of SCOS3 in mice. It markedly increased the level of SOCS3 in the spinal cord of mice and subsequently inhibited morphine-induced phosphorylation of NF-κB p65. In addition, a miRNA-30a-5p inhibitor decreased the levels of IL-1β and TNF-α caused by morphine in microglia. CONCLUSION AMPK-autophagy activation suppresses neuroinflammation and improves morphine tolerance via the upregulation of SOCS3 by inhibiting miRNA-30a-5p.
Collapse
Affiliation(s)
- Li Wan
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Ru-Meng Jia
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Lu-Lu Ji
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Xin-Miao Qin
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Liang Hu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Fan Hu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Yuan Han
- Department of Anesthesiology, Eye & ENT Hospital, Fudan University, Shanghai, 200031, China
| | - Yin-Bing Pan
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Chun-Yi Jiang
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China.
| | - Wen-Tao Liu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China.
| |
Collapse
|
17
|
Joshi M, Nikte SV, Sengupta D. Molecular determinants of GPCR pharmacogenetics: Deconstructing the population variants in β 2-adrenergic receptor. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2022; 128:361-396. [PMID: 35034724 DOI: 10.1016/bs.apcsb.2021.08.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
G protein-coupled receptors (GPCRs) are membrane proteins that play a central role in cell signaling and constitute one of the largest classes of drug targets. The molecular mechanisms underlying GPCR function have been characterized by several experimental and computational methods and provide an understanding of their role in physiology and disease. Population variants arising from nsSNPs affect the native function of GPCRs and have been implicated in differential drug response. In this chapter, we provide an overview on GPCR structure and activation, with a special focus on the β2-adrenergic receptor (β2-AR). First, we discuss the current understanding of the structural and dynamic features of the wildtype receptor. Subsequently, the population variants identified in this receptor from clinical and large-scale genomic studies are described. We show how computational approaches such as bioinformatics tools and molecular dynamics simulations can be used to characterize the variant receptors in comparison to the wildtype receptor. In particular, we discuss three examples of clinically important variants and discuss how the structure and function of these variants differ from the wildtype receptor at a molecular level. Overall, the chapter provides an overview of structure and function of GPCR variants and is a step towards the study of inter-individual differences and personalized medicine.
Collapse
Affiliation(s)
- Manali Joshi
- Bioinformatics Centre, Savitribai Phule Pune University, Pune, India.
| | - Siddhanta V Nikte
- CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Durba Sengupta
- CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India.
| |
Collapse
|
18
|
Rahban M, Danyali S, Zaringhalam J, Manaheji H. Pharmacological blockade of neurokinin1 receptor restricts morphine-induced tolerance and hyperalgesia in the rat. Scand J Pain 2022; 22:193-203. [PMID: 34525274 DOI: 10.1515/sjpain-2021-0052] [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: 03/17/2021] [Accepted: 08/09/2021] [Indexed: 11/15/2022]
Abstract
OBJECTIVES The most notable adverse side effects of chronic morphine administration include tolerance and hyperalgesia. This study investigated the involvement of dorsal root ganglion (DRG) protein kinase Cɛ (PKCɛ) expression during chronic morphine administration and also considered the relationship between DRG PKCɛ expression and the substance P- neurokinin1 receptor (SP- NK1R) activity. METHODS Thirty-six animals were divided into six groups (n=6) in this study. In the morphine and sham groups, rats received 10 µg intrathecal (i.t.) morphine or saline for eight consecutive days, respectively. Behavioral tests were performed on days 1 and 8 before and after the first injections and then 48 h after the last injection (day 10). In the treatment groups, rats received NK1R antagonist (L-732,138, 25 µg) daily, either alone or 10 min before a morphine injection, Sham groups received DMSO alone or 10 min before a morphine injection. Animals were sacrificed on days 8 and 10, and DRG PKCɛ and SP expression were analyzed by western blot and immunohistochemistry techniques, respectively. RESULTS Behavioral tests indicated that tolerance developed following eight days of chronic morphine injection. Hyperalgesia was induced 48 h after the last morphine injection. Expression of SP and PKCɛ in DRG significantly increased in rats that developed morphine tolerance on day 8 and hyperalgesia on day 10, respectively. NK1R antagonist (L-732,138) not only blocked the development of hyperalgesia and the increase of PKCɛ expression but also alleviated morphine tolerance. CONCLUSIONS Our results provide evidence that DRG PKCɛ and SP-NK1R most likely participated in the generation of morphine tolerance and hyperalgesia. Pharmacological inhibition of SP-NK1R activity in the spinal cord suggests a role for NK1R and in restricting some side effects of chronic morphine. All experiments were performed by the National Institute of Health (NIH) Guidelines for the Care and Use of Laboratory Animals (NIH Publication No. 80-23, revised1996) and were approved by the Animal Ethics Committee of Shahid Beheshti University of Medical Sciences, Tehran, Iran (IR.SBMU.MSP.REC.1396.130).
Collapse
Affiliation(s)
- Mohammad Rahban
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Samira Danyali
- Department of Physiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Jalal Zaringhalam
- Neurophysiology Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Homa Manaheji
- Neurophysiology Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
19
|
Cellular, synaptic, and network effects of chemokines in the central nervous system and their implications to behavior. Pharmacol Rep 2021; 73:1595-1625. [PMID: 34498203 PMCID: PMC8599319 DOI: 10.1007/s43440-021-00323-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 08/15/2021] [Accepted: 08/16/2021] [Indexed: 02/07/2023]
Abstract
Accumulating evidence highlights chemokines as key mediators of the bidirectional crosstalk between neurons and glial cells aimed at preserving brain functioning. The multifaceted role of these immune proteins in the CNS is mirrored by the complexity of the mechanisms underlying its biological function, including biased signaling. Neurons, only in concert with glial cells, are essential players in the modulation of brain homeostatic functions. Yet, attempts to dissect these complex multilevel mechanisms underlying coordination are still lacking. Therefore, the purpose of this review is to summarize the current knowledge about mechanisms underlying chemokine regulation of neuron-glia crosstalk linking molecular, cellular, network, and behavioral levels. Following a brief description of molecular mechanisms by which chemokines interact with their receptors and then summarizing cellular patterns of chemokine expression in the CNS, we next delve into the sequence and mechanisms of chemokine-regulated neuron-glia communication in the context of neuroprotection. We then define the interactions with other neurotransmitters, neuromodulators, and gliotransmitters. Finally, we describe their fine-tuning on the network level and the behavioral relevance of their modulation. We believe that a better understanding of the sequence and nature of events that drive neuro-glial communication holds promise for the development of new treatment strategies that could, in a context- and time-dependent manner, modulate the action of specific chemokines to promote brain repair and reduce the neurological impairment.
Collapse
|
20
|
Zhang L, Roy S. Opioid Modulation of the Gut-Brain Axis in Opioid-Associated Comorbidities. Cold Spring Harb Perspect Med 2021; 11:a040485. [PMID: 32816876 PMCID: PMC8415294 DOI: 10.1101/cshperspect.a040485] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Growing evidence from animal and human studies show that opioids have a major impact on the composition and function of gut microbiota. This leads to disruption in gut permeability and altered microbial metabolites, driving both systemic and neuroinflammation, which in turn impacts central nervous system (CNS) homeostasis. Tolerance and dependence are the major comorbidities associated with prolonged opioid use. Inflammatory mediators and signaling pathways have been implicated in both opioid tolerance and dependence. We provide evidence that targeting the gut microbiome during opioid use through prebiotics, probiotics, antibiotics, and fecal microbial transplantation holds the greatest promise for novel treatments for opioid abuse. Basic research and clinical trials are required to examine what is more efficacious to yield new insights into the role of the gut-brain axis in opioid abuse.
Collapse
Affiliation(s)
- Li Zhang
- Department of Pharmacology, University of Minnesota, Minnesota McGuire Translational Research Facility, Minneapolis, Minnesota 55455, USA
| | - Sabita Roy
- Department of Pharmacology, University of Minnesota, Minnesota McGuire Translational Research Facility, Minneapolis, Minnesota 55455, USA
- Department of Surgery, University of Miami, Miami, Florida 33153, USA
| |
Collapse
|
21
|
Li Y, Bao Y, Zheng H, Qin Y, Hua B. Can Src protein tyrosine kinase inhibitors be combined with opioid analgesics? Src and opioid-induced tolerance, hyperalgesia and addiction. Biomed Pharmacother 2021; 139:111653. [PMID: 34243625 DOI: 10.1016/j.biopha.2021.111653] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/17/2021] [Accepted: 04/21/2021] [Indexed: 10/21/2022] Open
Abstract
The clinical application of opioids may be accompanied by a series of adverse consequences, such as opioid tolerance, opioid-induced hyperalgesia, opioid dependence or addiction. In view of this issue, clinicians are faced with the dilemma of treating various types of pain with or without opioids. In this review, we discuss that Src protein tyrosine kinase plays an important role in these adverse consequences, and Src inhibitors can solve these problems well. Therefore, Src inhibitors have the potential to be used in combination with opioids to achieve synergy. How to combine them together to maximize the analgesic effect while avoiding unnecessary trouble provides a topic for follow-up research.
Collapse
Affiliation(s)
- Yaoyuan Li
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yanju Bao
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Honggang Zheng
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yinggang Qin
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Baojin Hua
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
22
|
Khan F, Mehan A. Addressing opioid tolerance and opioid-induced hypersensitivity: Recent developments and future therapeutic strategies. Pharmacol Res Perspect 2021; 9:e00789. [PMID: 34096178 PMCID: PMC8181203 DOI: 10.1002/prp2.789] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 04/07/2021] [Indexed: 01/07/2023] Open
Abstract
Opioids are a commonly prescribed and efficacious medication for the treatment of chronic pain but major side effects such as addiction, respiratory depression, analgesic tolerance, and paradoxical pain hypersensitivity make them inadequate and unsafe for patients requiring long-term pain management. This review summarizes recent advances in our understanding of the outcomes of chronic opioid administration to lay the foundation for the development of novel pharmacological strategies that attenuate opioid tolerance and hypersensitivity; the two main physiological mechanisms underlying the inadequacies of current therapeutic strategies. We also explore mechanistic similarities between the development of neuropathic pain states, opioid tolerance, and hypersensitivity which may explain opioids' lack of efficacy in certain patients. The findings challenge the current direction of analgesic research in developing non-opioid alternatives and we suggest that improving opioids, rather than replacing them, will be a fruitful avenue for future research.
Collapse
Affiliation(s)
- Faris Khan
- School of Clinical MedicineUniversity of CambridgeCambridgeUK
| | - Aman Mehan
- School of Clinical MedicineUniversity of CambridgeCambridgeUK
| |
Collapse
|
23
|
Ahearn OC, Watson MN, Rawls SM. Chemokines, cytokines and substance use disorders. Drug Alcohol Depend 2021; 220:108511. [PMID: 33465606 PMCID: PMC7889725 DOI: 10.1016/j.drugalcdep.2021.108511] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 11/30/2020] [Accepted: 12/14/2020] [Indexed: 12/18/2022]
Abstract
Efficacious pharmacotherapies for the treatment of substance use disorders need to be expanded and improved. Non-neuronal cells, particularly astrocytes and microglia, have emerged as therapeutic targets for the development of pharmacotherapies to treat dependence and relapse that accompanies chronic drug use. Cytokines and chemokines are neuroimmune factors expressed in neurons, astrocytes, and microglia that demonstrate promising clinical utility as therapeutic targets for substance use disorders. In this review, we describe a role for cytokines and chemokines in the rewarding and reinforcing effects of alcohol, opioids, and psychostimulants. We also discuss emerging cytokine- and chemokine-based therapeutic strategies that differ from conventional strategies directed toward transporters and receptors within the dopamine, glutamate, GABA, serotonin, and GABA systems.
Collapse
Affiliation(s)
- Olivia C. Ahearn
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University Philadelphia, PA, USA
| | - Mia N. Watson
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University Philadelphia, PA, USA
| | - Scott M. Rawls
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University Philadelphia, PA, USA,Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| |
Collapse
|
24
|
Chen J, Wang G, Sun T, Ma C, Huo X, Kong Y. Involvement of TCF7L2 in generation of morphine-induced antinociceptive tolerance and hyperalgesia by modulating TLR4/ NF-κB/NLRP3 in microglia. Toxicol Appl Pharmacol 2021; 416:115458. [PMID: 33607128 DOI: 10.1016/j.taap.2021.115458] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 01/29/2021] [Accepted: 02/10/2021] [Indexed: 12/28/2022]
Abstract
Morphine is an opioid agonist and a nonselective mu, kappa and delta receptor agonist. It is a commonly used analgesic drug for the treatment of acute and chronic pain as well as cancer pain. Morphine is particularly important to address the problem of morphine tolerance. Tcf7l2, known as a risk gene for schizophrenia and autism, encodes a member of the LEF1/TCF transcription factor family. TCF7L2 is an important transcription factor that is upregulated in neuropathic pain models. However, the relationship between TCF7L2 and morphine tolerance has not been reported. In this study, we found that morphine tolerance led to the upregulation of TCF7L2 in the spinal cord, and also led to the upregulation of TCF7L2 expression in glial cells, which promoted inflammation related signal, and activated TLR4 / NF-κB/NLRP3 pathway. In addition, TCF7L2 regulated microglial cell activation induced by chronic morphine treatment. Mechanically, we found that TCF7L2 transcriptionally regulated TLR4 expression, and the depletion of TCF7L2 alleviated morphine tolerance induced by chronic morphine treatment, and further alleviated pain hypersensitivity induced by chronic morphine treatment. We therefore suggested that TCF7L2 regulates the activation of TLR4/ NF-κB/NLRP3 pathway in microglia, and is involved in the formation of morphine tolerance. Our results provide a new idea for the regulation mechanism of morphine tolerance.
Collapse
Affiliation(s)
- Jing Chen
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150081, China
| | - Guonian Wang
- Harbin Medical University Sino-Russian Research Center Pain Management Research Institute, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province 150001, China.
| | - Tingting Sun
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150081, China
| | - Chao Ma
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150081, China
| | - Xing Huo
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150081, China
| | - Yiran Kong
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150081, China
| |
Collapse
|
25
|
Fan Y, Ji X, Zhang L, Zhang X. The Analgesic Effects of Static Magnetic Fields. Bioelectromagnetics 2021; 42:115-127. [PMID: 33508148 DOI: 10.1002/bem.22323] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 12/17/2020] [Accepted: 01/02/2021] [Indexed: 11/09/2022]
Abstract
Pain is one of the most common reasons why people seek medical care, which is related to most disease states. Magnetic fields (MFs) can be applied locally to specific parts of human bodies with high penetration and temporal control, which have a long-debated history in folk therapy. The purpose of this review is to collect and analyze experimental data about the analgesic effects of static magnetic fields (SMFs) so that we can have a scientific understanding regarding this topic. We collected 28 studies (25 English and 3 Chinese papers) with proper sham controls that investigated the effects of SMFs on pain relief in humans or mice. We found that 64% of the human studies and all mice studies in the literature showed positive analgesic effects of SMFs, which are related to factors including SMF intensity, treatment time, and pain types. Higher intensity and/or longer treatment time, as well as some specific pain types, may have better pain relief effects. Initial mechanistic studies indicated that membrane receptors, such as capsaicin receptor VR1/TRPV1, opioid receptors, and P2X3 receptors, might be involved. By describing experimental evidence and analysis, we found that SMFs actually hold considerable promise for managing some specific types of pain if proper SMF parameters are used. More studies comprehensively evaluating the parameters of SMF and its corresponding analgesic effects on different pain types, as well as the underlying molecular mechanisms, will be necessary to further validate its therapeutic potential in pain management in the future. Bioelectromagnetics. 00:00-00, 2021. © 2021 Bioelectromagnetics Society.
Collapse
Affiliation(s)
- Yixiang Fan
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
- Science Island Branch of Graduate School, University of Science and Technology of China, Hefei, China
| | - Xinmiao Ji
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
| | - Lei Zhang
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
| | - Xin Zhang
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
- Science Island Branch of Graduate School, University of Science and Technology of China, Hefei, China
- Institutes of Physical Science and Information Technology, Anhui University, Hefei, China
- International Magnetobiology Frontier Research Center (iMFRC), Science Island, Hefei, China
| |
Collapse
|
26
|
Willner MJ, Xiao Y, Kim HS, Chen X, Xu B, Leong KW. Modeling SARS-CoV-2 infection in individuals with opioid use disorder with brain organoids. J Tissue Eng 2021; 12:2041731420985299. [PMID: 33738089 PMCID: PMC7934045 DOI: 10.1177/2041731420985299] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 12/13/2020] [Indexed: 01/10/2023] Open
Abstract
The COVID-19 pandemic has aggravated a preexisting epidemic: the opioid crisis. Much literature has shown that the circumstances imposed by COVID-19, such as social distancing regulations, medical and financial instability, and increased mental health issues, have been detrimental to those with opioid use disorder (OUD). In addition, unexpected neurological sequelae in COVID-19 patients suggest that COVID-19 compromises neuroimmunity, induces hypoxia, and causes respiratory depression, provoking similar effects as those caused by opioid exposure. Combined conditions of COVID-19 and OUD could lead to exacerbated complications. With limited human in vivo options to study these complications, we suggest that iPSC-derived brain organoid models may serve as a useful platform to investigate the physiological connection between COVID-19 and OUD. This mini-review highlights the advances of brain organoids in other neuropsychiatric and infectious diseases and suggests their potential utility for investigating OUD and COVID-19, respectively.
Collapse
Affiliation(s)
- Moshe J Willner
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Yang Xiao
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Hye Sung Kim
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan, Republic of Korea
- Department of Regenerative Dental Medicine, College of Dentistry, Dankook University, Cheonan, Republic of Korea
- Cell & Matter Institute, Dankook University, Cheonan, Republic of Korea
| | - Xuejing Chen
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Department of Physics, Tsinghua University, Beijing, China
| | - Bin Xu
- Department of Psychiatry, Columbia University Medical Center, New York, NY, USA
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
27
|
Guerrero-Moreno A, Fakih D, Parsadaniantz SM, Réaux-Le Goazigo A. How does chronic dry eye shape peripheral and central nociceptive systems? Neural Regen Res 2021; 16:306-307. [PMID: 32859787 PMCID: PMC7896235 DOI: 10.4103/1673-5374.290895] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Affiliation(s)
| | - Darine Fakih
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | | | | |
Collapse
|
28
|
Guerrero-Moreno A, Baudouin C, Melik Parsadaniantz S, Réaux-Le Goazigo A. Morphological and Functional Changes of Corneal Nerves and Their Contribution to Peripheral and Central Sensory Abnormalities. Front Cell Neurosci 2020; 14:610342. [PMID: 33362474 PMCID: PMC7758484 DOI: 10.3389/fncel.2020.610342] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 11/18/2020] [Indexed: 11/24/2022] Open
Abstract
The cornea is the most densely innervated and sensitive tissue in the body. The cornea is exclusively innervated by C- and A-delta fibers, including mechano-nociceptors that are triggered by noxious mechanical stimulation, polymodal nociceptors that are excited by mechanical, chemical, and thermal stimuli, and cold thermoreceptors that are activated by cooling. Noxious stimulations activate corneal nociceptors whose cell bodies are located in the trigeminal ganglion (TG) and project central axons to the trigeminal brainstem sensory complex. Ocular pain, in particular, that driven by corneal nerves, is considered to be a core symptom of inflammatory and traumatic disorders of the ocular surface. Ocular surface injury affecting corneal nerves and leading to inflammatory responses can occur under multiple pathological conditions, such as chemical burn, persistent dry eye, and corneal neuropathic pain as well as after some ophthalmological surgical interventions such as photorefractive surgery. This review depicts the morphological and functional changes of corneal nerve terminals following corneal damage and dry eye disease (DED), both ocular surface conditions leading to sensory abnormalities. In addition, the recent fundamental and clinical findings of the importance of peripheral and central neuroimmune interactions in the development of corneal hypersensitivity are discussed. Next, the cellular and molecular changes of corneal neurons in the TG and central structures that are driven by corneal nerve abnormalities are presented. A better understanding of the corneal nerve abnormalities as well as neuroimmune interactions may contribute to the identification of a novel therapeutic targets for alleviating corneal pain.
Collapse
Affiliation(s)
| | - Christophe Baudouin
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, IHU FOReSIGHT, Paris, France.,CHNO des Quinze-Vingts, IHU FOReSIGHT, INSERM-DGOS CIC 1423, Paris, France.,Department of Ophthalmology, Ambroise Paré Hospital, AP-HP, University of Versailles Saint-Quentin-en-Yvelines, Boulogne-Billancourt, France
| | | | | |
Collapse
|
29
|
Santoni A, Mercadante S, Arcuri E. Chronic cancer and non-cancer pain and opioid-induced hyperalgesia share common mechanisms: neuroinflammation and central sensitization. Minerva Anestesiol 2020; 87:210-222. [PMID: 33300326 DOI: 10.23736/s0375-9393.20.14822-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Neuroinflammation, a peculiar form of inflammation that occurs in response to noxious stimuli in peripheral and central nervous system (CNS), consists in altered vascular permeability followed by leukocyte recruitment and activation in the inflamed tissue, release of inflammatory mediators including cytokines and chemokines, and finally in the activation of microglia and astrocytes in the spinal cord and CNS. This phenomenon mediates and even worsen the inflammatory pain in many painful states and is responsible for central sensitization leading to pain chronicity. We describe the major neuroinflammatory mechanisms shared by cancer and non-cancer pain. Particular attention is given to two different chronic inflammatory painful diseases such as the complex regional pain syndrome and the rheumatoid arthritis as prototypes of neuroinflammatory diseases (gliopathies). In addition, we describe the complexity of tumor microenvironment, their main cellular components (tumor cells, tumor infiltrating leukocytes and sensory neurons) and their reciprocal interactions that characterize different forms and intensity of cancer pain. We also hypothesize that one type of cancer pain, the breakthrough pain, can be attributable to receptor-mediated interaction of opioids with tumor cells and intratumoral leukocytes. Surprisingly, long-term opioid treatment shares the same neuroinflammatory potential responsible for the chronicity of both cancer and non-cancer pain; thus, resulting in paradoxical worsening rather than relieving pain. This paradox has upset the world of pain therapy, with neuroinflammation now being a main target of emerging therapies.
Collapse
Affiliation(s)
- Angela Santoni
- Department of Molecular Medicine, Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, Rome, Italy.,IRCCS Neuromed, Pozzilli, Isernia, Italy
| | - Sebastiano Mercadante
- Main Regional Center of Supportive/Palliative Care, La Maddalena Cancer Center, Palermo, Italy
| | - Edoardo Arcuri
- IRCCS Regina Elena Cancer Institute, IFO, Rome, Italy - .,Ars Medica Pain Clinic, Rome, Italy
| |
Collapse
|
30
|
Ma H, Wang H, Li M, Barreto-de-Souza V, Reinecke BA, Gunta R, Zheng Y, Kang G, Nassehi N, Zhang H, An J, Selley DE, Hauser KF, Zhang Y. Bivalent Ligand Aiming Putative Mu Opioid Receptor and Chemokine Receptor CXCR4 Dimers in Opioid Enhanced HIV-1 Entry. ACS Med Chem Lett 2020; 11:2318-2324. [PMID: 33214847 PMCID: PMC7667867 DOI: 10.1021/acsmedchemlett.0c00444] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 09/10/2020] [Indexed: 12/17/2022] Open
Abstract
A bivalent compound 1a featuring both a mu opioid receptor (MOR) and a CXCR4 antagonist pharmacophore (naltrexone and IT1t) was designed and synthesized. Further binding and functional studies demonstrated 1a acting as a MOR and a CXCR4 dual antagonist with reasonable binding affinities at both receptors. Furthermore, compound 1a seemed more effective than a combination of IT1t and naltrexone in inhibiting HIV entry at the presence of morphine. Additional molecular modeling results suggested that 1a may bind with the putative MOR-CXCR4 heterodimer to induce its anti-HIV activity. Collectively, bivalent ligand 1a may serve as a promising lead to develop chemical probes targeting the putative MOR-CXCR4 heterodimer in comprehending opioid exacerbated HIV-1 invasion.
Collapse
Affiliation(s)
- Hongguang Ma
- Department
of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, 800 E Leigh Street, Richmond, Virginia 23298, United States
| | - Huiqun Wang
- Department
of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, 800 E Leigh Street, Richmond, Virginia 23298, United States
| | - Mengchu Li
- Department
of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, 800 E Leigh Street, Richmond, Virginia 23298, United States
| | - Victor Barreto-de-Souza
- Department
of Pharmacology and Toxicology, Virginia
Commonwealth University, 410 North 12th Street, Richmond, Virginia 23298, United
States
| | - Bethany A. Reinecke
- Department
of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, 800 E Leigh Street, Richmond, Virginia 23298, United States
| | - Rama Gunta
- Department
of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, 800 E Leigh Street, Richmond, Virginia 23298, United States
| | - Yi Zheng
- Department
of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, 800 E Leigh Street, Richmond, Virginia 23298, United States
| | - Guifeng Kang
- Department
of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, 800 E Leigh Street, Richmond, Virginia 23298, United States
| | - Nima Nassehi
- Department
of Pharmacology and Toxicology, Virginia
Commonwealth University, 410 North 12th Street, Richmond, Virginia 23298, United
States
| | - Huijun Zhang
- Department
of Medicine, Division of Infectious Diseases, School of Medicine, University of California San Diego, 9500 Gilman Drive, Stein Clinical
Research Building, Suite 410, La Jolla, California 92093, United States
| | - Jing An
- Department
of Medicine, Division of Infectious Diseases, School of Medicine, University of California San Diego, 9500 Gilman Drive, Stein Clinical
Research Building, Suite 410, La Jolla, California 92093, United States
| | - Dana E. Selley
- Department
of Pharmacology and Toxicology, Virginia
Commonwealth University, 410 North 12th Street, Richmond, Virginia 23298, United
States
| | - Kurt F. Hauser
- Department
of Pharmacology and Toxicology, Virginia
Commonwealth University, 410 North 12th Street, Richmond, Virginia 23298, United
States
| | - Yan Zhang
- Department
of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, 800 E Leigh Street, Richmond, Virginia 23298, United States
| |
Collapse
|
31
|
Pelechas E, Voulgari PV, Drosos AA. Recent advances in the opioid mu receptor based pharmacotherapy for rheumatoid arthritis. Expert Opin Pharmacother 2020; 21:2153-2160. [PMID: 33135514 DOI: 10.1080/14656566.2020.1796969] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Opioids are used for severe forms of acute and cancer pain. Over the last years, their potential use in patients with noncancer pain such as those with rheumatoid arthritis (RA) has been postulated. A recent population-based comparative study showed that chronic opioid use was 12% vs. 4% among RA and non-RA patients, respectively. Another study showed an increase from 7.4% to 16.9% (2002 to 2015). In general, there has been an increasing tendency to use opioids in recent years. AREAS COVERED The authors have performed an extensive literature search using PubMed for articles including noncancer pain and the use of the mu opioid receptor (MOR) agonists in patients with RA. EXPERT OPINION Data is not sufficient to support opioid use for the treatment of chronic pain in patients with RA. Data is scarce and inconclusive. Rheumatologists should think and ponder the question: Why is this patient in pain? Differential diagnosis should include a disease flare, degenerative changes of the musculoskeletal system, and fibromyalgia. And while there are new strategies for opioid administration currently being researched, unfortunately, they are far from being applied to human subjects in the everyday clinical setting, and are still being evaluated at an experimental level. CNS: Central nervous system; DORs: delta opioid receptor agonists; GI: Gastrointestinal; GPCRs: G protein-coupled receptors; IL: Interleukin; JAK: Janus kinase; KORs: kappa opioid receptor agonists; MCPs: Metacarpophalangeal joints; MORs: Mu opioid receptor agonists; MTPs: Metatarsophalangeal joints; NSAIDs: Non-steroidal anti-inflammatory drugsOA: Osteoarthritis; ORs: Opioid receptors; PD: Pharmacodynamic; PIPs: Proximal interphalangeal joints; PK: Pharmacokinetic; PNS: Peripheral nervous system; RA: Rheumatoid arthritis; RGS: Regulator of G protein signaling; SSRIs: Selective serotonin reuptake inhibitors; TNF: Tumor necrosis factor.
Collapse
Affiliation(s)
- Eleftherios Pelechas
- Rheumatology Clinic, Department of Internal Medicine, Medical School, University of Ioannina , Ioannina, Greece
| | - Paraskevi V Voulgari
- Rheumatology Clinic, Department of Internal Medicine, Medical School, University of Ioannina , Ioannina, Greece
| | - Alexandros A Drosos
- Rheumatology Clinic, Department of Internal Medicine, Medical School, University of Ioannina , Ioannina, Greece
| |
Collapse
|
32
|
The ambiguity of opioids revealed by immunology is changing the knowledge and the therapeutic approach in cancer and non-cancer pain: A narrative review. Immunol Lett 2020; 226:12-21. [DOI: 10.1016/j.imlet.2020.06.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 06/17/2020] [Indexed: 12/18/2022]
|
33
|
Carranza-Aguilar CJ, Hernández-Mendoza A, Mejias-Aponte C, Rice KC, Morales M, González-Espinosa C, Cruz SL. Morphine and Fentanyl Repeated Administration Induces Different Levels of NLRP3-Dependent Pyroptosis in the Dorsal Raphe Nucleus of Male Rats via Cell-Specific Activation of TLR4 and Opioid Receptors. Cell Mol Neurobiol 2020; 42:677-694. [DOI: 10.1007/s10571-020-00957-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 08/30/2020] [Indexed: 02/07/2023]
|
34
|
Kan T, Yoshikawa M, Watanabe M, Miura M, Ito K, Matsuda M, Iwao K, Kobayashi H, Suzuki T, Suzuki T. Sialorphin Potentiates Effects of [Met 5]Enkephalin without Toxicity by Action other than Peptidase Inhibition. J Pharmacol Exp Ther 2020; 375:104-114. [PMID: 32759368 DOI: 10.1124/jpet.120.266080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 07/28/2020] [Indexed: 11/22/2022] Open
Abstract
This dose-response study investigated the effects of sialorphin on [Met5]enkephalin (ME)-induced inhibition of contractions in mouse vas deferens and antinociception in male rats. Differences were compared among combinations of three chemical peptidase inhibitors: amastatin, captopril, and phosphoramidon. The ratio of potencies of ME in mouse vas deferens pretreated with both sialorphin (100 µM) and a mixture of the three peptidase inhibitors (1 µM each) was higher than that with the mixture of peptidase inhibitors alone at any dose. Intrathecal administration of sialorphin (100-400 nmol) significantly and dose dependently increased ME (3 nmol)-induced antinociception with the mixture of three peptidase inhibitors (10 nmol each). The degree of antinociception with a combination of any two of the peptidase inhibitors (10 nmol each) in the absence of sialorphin was less than that in the presence of sialorphin (200 nmol). Pretreatment with both sialorphin (200 nmol) and the mixture of three peptidase inhibitors (10 nmol each) produced an approximately 100-fold augmentation in ME (10 nmol)-induced antinociception, but without signs of toxicity such as motor dysfunction in rats. Radioligand receptor binding assay revealed that sialorphin did not affect either binding affinity or maximal binding capacity of [d-Ala2,N-MePhe4,Gly-ol5]enkephalin. These results indicate that sialorphin potentiates the effects of ME without toxicity by a mechanism other than peptidase inhibition and with no effect on its affinity to µ-opioid receptors. SIGNIFICANCE STATEMENT: Sialorphin is regarded as an endogenous peptidase inhibitor that interacts with enkephalin-degrading enzymes. The results of these in vitro and in vivo studies confirm that sialorphin potentiates the effects of [Met5]enkephalin without toxicity by an action other than peptidase inhibition. This suggests that sialorphin offers the advantage of reducing or negating the side effects of opioid drugs and endogenous opioid peptides.
Collapse
Affiliation(s)
- Takugi Kan
- Departments of Anesthesiology (T.K., M.W., M.Mi., K.I., M.Ma., Ta.S., To.S.) and Clinical Pharmacology (M.Y., H.K.) and Education and Research Support Center (K.I.), Tokai University School of Medicine, Kanagawa, Japan
| | - Masanobu Yoshikawa
- Departments of Anesthesiology (T.K., M.W., M.Mi., K.I., M.Ma., Ta.S., To.S.) and Clinical Pharmacology (M.Y., H.K.) and Education and Research Support Center (K.I.), Tokai University School of Medicine, Kanagawa, Japan
| | - Mariko Watanabe
- Departments of Anesthesiology (T.K., M.W., M.Mi., K.I., M.Ma., Ta.S., To.S.) and Clinical Pharmacology (M.Y., H.K.) and Education and Research Support Center (K.I.), Tokai University School of Medicine, Kanagawa, Japan
| | - Masaaki Miura
- Departments of Anesthesiology (T.K., M.W., M.Mi., K.I., M.Ma., Ta.S., To.S.) and Clinical Pharmacology (M.Y., H.K.) and Education and Research Support Center (K.I.), Tokai University School of Medicine, Kanagawa, Japan
| | - Kenji Ito
- Departments of Anesthesiology (T.K., M.W., M.Mi., K.I., M.Ma., Ta.S., To.S.) and Clinical Pharmacology (M.Y., H.K.) and Education and Research Support Center (K.I.), Tokai University School of Medicine, Kanagawa, Japan
| | - Mitsumasa Matsuda
- Departments of Anesthesiology (T.K., M.W., M.Mi., K.I., M.Ma., Ta.S., To.S.) and Clinical Pharmacology (M.Y., H.K.) and Education and Research Support Center (K.I.), Tokai University School of Medicine, Kanagawa, Japan
| | - Kayoko Iwao
- Departments of Anesthesiology (T.K., M.W., M.Mi., K.I., M.Ma., Ta.S., To.S.) and Clinical Pharmacology (M.Y., H.K.) and Education and Research Support Center (K.I.), Tokai University School of Medicine, Kanagawa, Japan
| | - Hiroyuki Kobayashi
- Departments of Anesthesiology (T.K., M.W., M.Mi., K.I., M.Ma., Ta.S., To.S.) and Clinical Pharmacology (M.Y., H.K.) and Education and Research Support Center (K.I.), Tokai University School of Medicine, Kanagawa, Japan
| | - Takeshi Suzuki
- Departments of Anesthesiology (T.K., M.W., M.Mi., K.I., M.Ma., Ta.S., To.S.) and Clinical Pharmacology (M.Y., H.K.) and Education and Research Support Center (K.I.), Tokai University School of Medicine, Kanagawa, Japan
| | - Toshiyasu Suzuki
- Departments of Anesthesiology (T.K., M.W., M.Mi., K.I., M.Ma., Ta.S., To.S.) and Clinical Pharmacology (M.Y., H.K.) and Education and Research Support Center (K.I.), Tokai University School of Medicine, Kanagawa, Japan
| |
Collapse
|
35
|
Davoli-Ferreira M, de Lima KA, Fonseca MM, Guimarães RM, Gomes FI, Cavallini MC, Quadros AU, Kusuda R, Cunha FQ, Alves-Filho JC, Cunha TM. Regulatory T cells counteract neuropathic pain through inhibition of the Th1 response at the site of peripheral nerve injury. Pain 2020; 161:1730-1743. [PMID: 32701834 DOI: 10.1097/j.pain.0000000000001879] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The inflammatory/immune response at the site of peripheral nerve injury participates in the pathophysiology of neuropathic pain. Nevertheless, little is known about the local regulatory mechanisms underlying peripheral nerve injury that counteracts the development of pain. Here, we investigated the contribution of regulatory T (Treg) cells to the development of neuropathic pain by using a partial sciatic nerve ligation model in mice. We showed that Treg cells infiltrate and proliferate in the site of peripheral nerve injury. Local Treg cells suppressed the development of neuropathic pain mainly through the inhibition of the CD4 Th1 response. Treg cells also indirectly reduced neuronal damage and neuroinflammation at the level of the sensory ganglia. Finally, we identified IL-10 signaling as an intrinsic mechanism by which Treg cells counteract neuropathic pain development. These results revealed Treg cells as important inhibitory modulators of the immune response at the site of peripheral nerve injury that restrains the development of neuropathic pain. In conclusion, the boosting of Treg cell function/activity might be explored as a possible interventional approach to reduce neuropathic pain development after peripheral nerve damage.
Collapse
Affiliation(s)
- Marcela Davoli-Ferreira
- Department of Pharmacology, Center for Research on Inflammatory Diseases (CRID), Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
- Graduate Program in Basic and Applied Immunology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil. Dr. de Lima is now with the Department of Neuroscience, Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA, United States. Dr. Fonseca is now with the Department of Anesthesiology, Pain Mechanisms Laboratory, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Kalil A de Lima
- Department of Pharmacology, Center for Research on Inflammatory Diseases (CRID), Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
- Graduate Program in Basic and Applied Immunology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil. Dr. de Lima is now with the Department of Neuroscience, Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA, United States. Dr. Fonseca is now with the Department of Anesthesiology, Pain Mechanisms Laboratory, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Miriam M Fonseca
- Department of Pharmacology, Center for Research on Inflammatory Diseases (CRID), Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Rafaela M Guimarães
- Department of Pharmacology, Center for Research on Inflammatory Diseases (CRID), Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Francisco I Gomes
- Department of Pharmacology, Center for Research on Inflammatory Diseases (CRID), Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Maria C Cavallini
- Department of Pharmacology, Center for Research on Inflammatory Diseases (CRID), Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
- Graduate Program in Basic and Applied Immunology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil. Dr. de Lima is now with the Department of Neuroscience, Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA, United States. Dr. Fonseca is now with the Department of Anesthesiology, Pain Mechanisms Laboratory, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Andreza U Quadros
- Department of Pharmacology, Center for Research on Inflammatory Diseases (CRID), Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Ricardo Kusuda
- Department of Pharmacology, Center for Research on Inflammatory Diseases (CRID), Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Fernando Q Cunha
- Department of Pharmacology, Center for Research on Inflammatory Diseases (CRID), Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Jose C Alves-Filho
- Department of Pharmacology, Center for Research on Inflammatory Diseases (CRID), Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Thiago M Cunha
- Department of Pharmacology, Center for Research on Inflammatory Diseases (CRID), Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| |
Collapse
|
36
|
Mischel RA, Muchhala KH, Dewey WL, Akbarali HI. The "Culture" of Pain Control: A Review of Opioid-Induced Dysbiosis (OID) in Antinociceptive Tolerance. THE JOURNAL OF PAIN 2020; 21:751-762. [PMID: 31841668 PMCID: PMC7286790 DOI: 10.1016/j.jpain.2019.11.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 11/08/2019] [Accepted: 11/11/2019] [Indexed: 12/15/2022]
Abstract
It is increasingly recognized that chronic opioid use leads to maladaptive changes in the composition and localization of gut bacteria. Recently, this "opioid-induced dysbiosis" (OID) has been linked to antinociceptive tolerance development in preclinical models and may therefore identify promising targets for new opioid-sparing strategies. Such developments are critical to curb dose escalations in the clinical setting and combat the ongoing opioid epidemic. In this article, we review the existing literature that pertains to OID, including the current evidence regarding its qualitative nature, influence on antinociceptive tolerance, and future prospects. PERSPECTIVE: This article reviews the current literature on OID of gut bacteria, including its qualitative nature, influence on antinociceptive tolerance, and future prospects. This work may help identify targets for new opioid-sparing strategies.
Collapse
Affiliation(s)
- Ryan A Mischel
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia
| | - Karan H Muchhala
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia
| | - William L Dewey
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia
| | - Hamid I Akbarali
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia.
| |
Collapse
|
37
|
Liu Q, Su LY, Sun C, Jiao L, Miao Y, Xu M, Luo R, Zuo X, Zhou R, Zheng P, Xiong W, Xue T, Yao YG. Melatonin alleviates morphine analgesic tolerance in mice by decreasing NLRP3 inflammasome activation. Redox Biol 2020; 34:101560. [PMID: 32413745 PMCID: PMC7225735 DOI: 10.1016/j.redox.2020.101560] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 04/27/2020] [Accepted: 04/27/2020] [Indexed: 01/03/2023] Open
Abstract
Morphine is frequently used for pain relief, but long-term morphine therapy in patients with chronic pain results in analgesic tolerance and hyperalgesia. There are no effective therapeutic treatments that limit these detrimental side effects. We found pretreatment with melatonin could decrease morphine-induced analgesic tolerance. There was a significant activation of the NLRP3 inflammasome in the prefrontal cortex and the peripheral blood of morphine-treated mice compared to control animals, which could be blocked by melatonin. The inflammasome activation induced by morphine was mediated by the microglia. SiRNA knockdown or pharmacological inhibition of the NLRP3 abolished the morphine-induced inflammasome activation. Co-administration of melatonin and low-dose morphine had better analgesia effects in the murine models of pain and led to a lower NLRP3 inflammasome activity in brain tissues. Mice deficient for Nlrp3 had a higher nociceptive threshold and were less sensitive to develop morphine-induced analgesic tolerance and acetic acid-induced pain relative to wild-type animals. Concordantly, we observed a significantly elevated level of serum IL-1β, which indicates an increase of NLRP3 inflammasome activity associated with the reduced level of serum melatonin, in heroin-addicted patients relative to healthy individuals. Our results provide a solid basis for conducting a clinical trial with the co-administration of melatonin and morphine for the relief of severe pain.
Collapse
Affiliation(s)
- Qianjin Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China
| | - Ling-Yan Su
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, 650223, China.
| | - Chunli Sun
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China; State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
| | - Lijin Jiao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China
| | - Ying Miao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, 650223, China; Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Min Xu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China
| | - Rongcan Luo
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, 650223, China
| | - Xin Zuo
- Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Rongbin Zhou
- Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Ping Zheng
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China; State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China; KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
| | - Wei Xiong
- Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230026, China; CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Tian Xue
- Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230026, China; CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yong-Gang Yao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China; KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China; CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
38
|
Meyrath M, Szpakowska M, Zeiner J, Massotte L, Merz MP, Benkel T, Simon K, Ohnmacht J, Turner JD, Krüger R, Seutin V, Ollert M, Kostenis E, Chevigné A. The atypical chemokine receptor ACKR3/CXCR7 is a broad-spectrum scavenger for opioid peptides. Nat Commun 2020; 11:3033. [PMID: 32561830 PMCID: PMC7305236 DOI: 10.1038/s41467-020-16664-0] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 05/15/2020] [Indexed: 12/29/2022] Open
Abstract
Endogenous opioid peptides and prescription opioid drugs modulate pain, anxiety and stress by activating opioid receptors, currently classified into four subtypes. Here we demonstrate that ACKR3/CXCR7, hitherto known as an atypical scavenger receptor for chemokines, is a broad-spectrum scavenger of opioid peptides. Phylogenetically, ACKR3 is intermediate between chemokine and opioid receptors and is present in various brain regions together with classical opioid receptors. Functionally, ACKR3 is a scavenger receptor for a wide variety of opioid peptides, especially enkephalins and dynorphins, reducing their availability for the classical opioid receptors. ACKR3 is not modulated by prescription opioids, but we show that an ACKR3-selective subnanomolar competitor peptide, LIH383, can restrain ACKR3’s negative regulatory function on opioid peptides in rat brain and potentiate their activity towards classical receptors, which may open alternative therapeutic avenues for opioid-related disorders. Altogether, our results reveal that ACKR3 is an atypical opioid receptor with cross-family ligand selectivity. Opioids modulate pain, anxiety and stress by activating four subtypes of opioid receptors. The authors show that atypical chemokine receptor 3 (ACKR3) is a scavenger for various endogenous opioid peptides regulating their availability without activating downstream signaling.
Collapse
Affiliation(s)
- Max Meyrath
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), rue Henri Koch 29, L-4354, Esch-sur-Alzette, Luxembourg
| | - Martyna Szpakowska
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), rue Henri Koch 29, L-4354, Esch-sur-Alzette, Luxembourg
| | - Julian Zeiner
- Molecular, Cellular and Pharmacobiology Section, Institute of Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115, Bonn, Germany
| | - Laurent Massotte
- Neurophysiology Unit, GIGA Neurosciences, University of Liège, avenue de l'hopital, B-4000, Liège, Belgium
| | - Myriam P Merz
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), rue Henri Koch 29, L-4354, Esch-sur-Alzette, Luxembourg
| | - Tobias Benkel
- Molecular, Cellular and Pharmacobiology Section, Institute of Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115, Bonn, Germany.,Research Training Group 1873, University of Bonn, Bonn, Germany
| | - Katharina Simon
- Molecular, Cellular and Pharmacobiology Section, Institute of Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115, Bonn, Germany
| | - Jochen Ohnmacht
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, avenue du Swing 6, L-4367, Belvaux, Luxembourg.,Department of Life Sciences and Medicine, University of Luxembourg, avenue du Swing 6, L-4367, Belvaux, Luxembourg
| | - Jonathan D Turner
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), rue Henri Koch 29, L-4354, Esch-sur-Alzette, Luxembourg
| | - Rejko Krüger
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, avenue du Swing 6, L-4367, Belvaux, Luxembourg.,Transversal Translational Medicine, Luxembourg Institute of Health (LIH), rue Thomas Edison 1A-B, L-1445, Strassen, Luxembourg
| | - Vincent Seutin
- Neurophysiology Unit, GIGA Neurosciences, University of Liège, avenue de l'hopital, B-4000, Liège, Belgium
| | - Markus Ollert
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), rue Henri Koch 29, L-4354, Esch-sur-Alzette, Luxembourg.,Department of Dermatology and Allergy Center, Odense Research Center for Anaphylaxis, University of Southern Denmark, 5000, Odense, Denmark
| | - Evi Kostenis
- Molecular, Cellular and Pharmacobiology Section, Institute of Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115, Bonn, Germany
| | - Andy Chevigné
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), rue Henri Koch 29, L-4354, Esch-sur-Alzette, Luxembourg.
| |
Collapse
|
39
|
Xu X, Yang K, Zhang F, Liu W, Wang Y, Yu C, Wang J, Zhang K, Zhang C, Nenadic G, Tao D, Zhou X, Shang H, Chen J. Identification of herbal categories active in pain disorder subtypes by machine learning help reveal novel molecular mechanisms of algesia. Pharmacol Res 2020; 156:104797. [PMID: 32278044 DOI: 10.1016/j.phrs.2020.104797] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 03/26/2020] [Accepted: 03/29/2020] [Indexed: 02/06/2023]
Abstract
Chronic pain is highly prevalent and poorly controlled, of which the accurate underlying mechanisms need be further elucidated. Herbal drugs have been widely used for controlling various pain disorders. The systematic integration of pain herbal data resources might be promising to help investigate the molecular mechanisms of pain phenotypes. Here, we integrated large-scale bibliographic literatures and well-established data sources to obtain high-quality pain relevant herbal data (i.e. 426 pain related herbs with their targets). We used machine learning method to identify three distinct herb categories with their specific indications of symptoms, targets and enriched pathways, which were characterized by the efficacy of treatment to the chronic cough related neuropathic pain, the reproduction and autoimmune related pain, and the cancer pain, respectively. We further detected the novel pathophysiological mechanisms of the pain subtypes by network medicine approach to evaluate the interactions between herb targets and the pain disease modules. This work increased the understanding of the underlying molecular mechanisms of pain subtypes that herbal drugs are participating and with the ultimate aim of developing novel personalized drugs for pain disorders.
Collapse
Affiliation(s)
- Xue Xu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China; Marcus Institute for Aging Research, Hebrew SeniorLife and Harvard Medical School, Boston, MA, 02131, USA
| | - Kuo Yang
- School of Computer and Information Technology, Beijing Jiaotong University, Beijing, 100044, China; MOE Key Laboratory of Bioinformatics, TCM-X Centre/Bioinformatics Division, BNRIST/Department of Automation, Tsinghua University, Beijing, 10084, China
| | - Feilong Zhang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China; Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Wenwen Liu
- School of Computer and Information Technology, Beijing Jiaotong University, Beijing, 100044, China
| | - Yinyan Wang
- School of Computer and Information Technology, Beijing Jiaotong University, Beijing, 100044, China
| | - Changying Yu
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Junyao Wang
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Keke Zhang
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Chao Zhang
- School of Mathematical Sciences, Dalian University of Technology, DaLian, Liaoning, 116024, China
| | - Goran Nenadic
- Computer Science, Faculty of Engineering and Physical Sciences, University of Manchester, Manchester, UK
| | - Dacheng Tao
- School of Information Technologies, The University of Sydney, Darlington, NSW, 2008, Australia
| | - Xuezhong Zhou
- School of Computer and Information Technology, Beijing Jiaotong University, Beijing, 100044, China.
| | - Hongcai Shang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China.
| | - Jianxin Chen
- Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
40
|
Reiss D, Maduna T, Maurin H, Audouard E, Gaveriaux-Ruff C. Mu opioid receptor in microglia contributes to morphine analgesic tolerance, hyperalgesia, and withdrawal in mice. J Neurosci Res 2020; 100:203-219. [PMID: 32253777 DOI: 10.1002/jnr.24626] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 02/23/2020] [Accepted: 03/18/2020] [Indexed: 12/17/2022]
Abstract
A major challenge in medicine is developing potent pain therapies without the adverse effects of opiates. Neuroinflammation and in particular microglial activation have been shown to contribute to these effects. However, the implication of the microglial mu opioid receptor (MOR) is not known. We developed a novel conditional knockout (cKO) mouse line, wherein MOR is deleted in microglia. Morphine analgesic tolerance was delayed in both sexes in cKO mice in the hot plate assay. Opioid-induced hyperalgesia (OIH) as measured in the tail immersion assay was abolished in male cKO mice, and physical dependence to morphine as assessed by naloxone-induced withdrawal was attenuated in female cKO mice. Our results show a sex-dependent contribution of microglial MOR in morphine analgesic tolerance, OIH, and physical dependence. In conclusion, our data suggest that blockade of microglial MOR could represent a therapeutic target for opiate analgesia without the opiate adverse effects.
Collapse
Affiliation(s)
- David Reiss
- Translational Medicine and Neurogenetics Department, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,IGBMC, Université de Strasbourg, Illkirch, France.,UMR7104, Centre National de la Recherche Scientifique, Illkirch, France.,U1258, Institut National de la Santé et de la Recherche Médicale, Illkirch, France
| | - Tando Maduna
- Translational Medicine and Neurogenetics Department, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,IGBMC, Université de Strasbourg, Illkirch, France.,UMR7104, Centre National de la Recherche Scientifique, Illkirch, France.,U1258, Institut National de la Santé et de la Recherche Médicale, Illkirch, France.,Neurology Research Group, Department of Physiology, Stellenbosch University, Stellenbosch, South Africa
| | - Hervé Maurin
- Translational Medicine and Neurogenetics Department, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,IGBMC, Université de Strasbourg, Illkirch, France.,UMR7104, Centre National de la Recherche Scientifique, Illkirch, France.,U1258, Institut National de la Santé et de la Recherche Médicale, Illkirch, France
| | - Emilie Audouard
- Translational Medicine and Neurogenetics Department, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,IGBMC, Université de Strasbourg, Illkirch, France.,UMR7104, Centre National de la Recherche Scientifique, Illkirch, France.,U1258, Institut National de la Santé et de la Recherche Médicale, Illkirch, France
| | - Claire Gaveriaux-Ruff
- Translational Medicine and Neurogenetics Department, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,IGBMC, Université de Strasbourg, Illkirch, France.,UMR7104, Centre National de la Recherche Scientifique, Illkirch, France.,U1258, Institut National de la Santé et de la Recherche Médicale, Illkirch, France.,Ecole Supérieure de Biotechnologie de Strasbourg, Illkirch, France
| |
Collapse
|
41
|
Pawlik K, Piotrowska A, Kwiatkowski K, Ciapała K, Popiolek‐Barczyk K, Makuch W, Mika J. The blockade of CC chemokine receptor type 1 influences the level of nociceptive factors and enhances opioid analgesic potency in a rat model of neuropathic pain. Immunology 2020; 159:413-428. [PMID: 31919846 PMCID: PMC7078003 DOI: 10.1111/imm.13172] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/19/2019] [Accepted: 01/07/2020] [Indexed: 12/15/2022] Open
Abstract
A growing body of evidence has indicated that the release of nociceptive factors, such as interleukins and chemokines, by activated immune and glial cells has crucial significance for neuropathic pain generation and maintenance. Moreover, changes in the production of nociceptive immune factors are associated with low opioid efficacy in the treatment of neuropathy. Recently, it has been suggested that CC chemokine receptor type 1 (CCR1) signaling is important for nociception. Our study provides evidence that the development of hypersensitivity in rats following chronic constriction injury (CCI) of the sciatic nerve is associated with significant up-regulation of endogenous CCR1 ligands, namely, CCL2, CCL3, CCL4, CCL6, CCL7 and CCL9 in the spinal cord and CCL2, CCL6, CCL7 and CCL9 in dorsal root ganglia (DRG). We showed that single and repeated intrathecal administration of J113863 (an antagonist of CCR1) attenuated mechanical and thermal hypersensitivity. Moreover, repeated administration of a CCR1 antagonist enhanced the analgesic properties of morphine and buprenorphine after CCI. Simultaneously, repeated administration of J113863 reduced the protein levels of IBA-1 in the spinal cord and MPO and CD4 in the DRG and, as a consequence, the level of pronociceptive factors, such as interleukin-1β (IL-1β), IL-6 and IL-18. The data obtained provide evidence that CCR1 blockade reduces hypersensitivity and increases opioid-induced analgesia through the modulation of neuroimmune interactions.
Collapse
Affiliation(s)
- Katarzyna Pawlik
- Department of Pain PharmacologyMaj Institute of PharmacologyPolish Academy of SciencesKrakowPoland
| | - Anna Piotrowska
- Department of Pain PharmacologyMaj Institute of PharmacologyPolish Academy of SciencesKrakowPoland
| | - Klaudia Kwiatkowski
- Department of Pain PharmacologyMaj Institute of PharmacologyPolish Academy of SciencesKrakowPoland
| | - Katarzyna Ciapała
- Department of Pain PharmacologyMaj Institute of PharmacologyPolish Academy of SciencesKrakowPoland
| | | | - Wioletta Makuch
- Department of Pain PharmacologyMaj Institute of PharmacologyPolish Academy of SciencesKrakowPoland
| | - Joanna Mika
- Department of Pain PharmacologyMaj Institute of PharmacologyPolish Academy of SciencesKrakowPoland
| |
Collapse
|
42
|
Rogers TJ. Bidirectional Regulation of Opioid and Chemokine Function. Front Immunol 2020; 11:94. [PMID: 32076421 PMCID: PMC7006827 DOI: 10.3389/fimmu.2020.00094] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 01/14/2020] [Indexed: 12/15/2022] Open
Abstract
The opioid family of GPCRs consists of the classical opioid receptors, designated μ-, κ-, and δ-opioid receptors, and the orphanin-FQ receptor, and these proteins are expressed on both neuronal and hematopoietic cells. A number of laboratories have reported that an important degree of cross-talk can occur between the opioid receptors and the chemokine and chemokine receptor families. As a part of this, the opioid receptors are known to regulate the expression of certain chemokines and chemokine receptors, including those that possess strong pro-inflammatory activity. At the level of receptor function, it is clear that certain members of the chemokine family can mediate cross-desensitization of the opioid receptors. Conversely, the opioid receptors are all able to induce heterologous desensitization of some of the chemokine receptors. Consequently, activation of one or more of the opioid receptors can selectively cross-desensitize chemokine receptors and regulate chemokine function. These cross-talk processes have significant implications for the inflammatory response, since the regulation of both the recruitment of inflammatory cells, as well as the sensation of pain, can be controlled in this way.
Collapse
Affiliation(s)
- Thomas J Rogers
- Center for Inflammation, Translational and Clinical Lung Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| |
Collapse
|
43
|
Shao S, Xia H, Hu M, Chen C, Fu J, Shi G, Guo Q, Zhou Y, Wang W, Shi J, Zhang T. Isotalatizidine, a C 19-diterpenoid alkaloid, attenuates chronic neuropathic pain through stimulating ERK/CREB signaling pathway-mediated microglial dynorphin A expression. J Neuroinflammation 2020; 17:13. [PMID: 31924228 PMCID: PMC6953278 DOI: 10.1186/s12974-019-1696-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 12/30/2019] [Indexed: 02/08/2023] Open
Abstract
Background Isotalatizidine is a representative C19-diterpenoid alkaloid extracted from the lateral roots of Aconitum carmichaelii, which has been widely used to treat various diseases on account of its analgesic, anti-inflammatory, anti-rheumatic, and immunosuppressive properties. The aim of this study was to evaluate the analgesic effect of isotalatizidine and its underlying mechanisms against neuropathic pain. Methods A chronic constrictive injury (CCI)-induced model of neuropathic pain was established in mice, and the limb withdrawal was evaluated by the Von Frey filament test following isotalatizidine or placebo administration. The signaling pathways in primary or immortalized microglia cells treated with isotalatizidine were analyzed by Western blotting and immunofluorescence. Results Intrathecal injection of isotalatizidine attenuated the CCI-induced mechanical allodynia in a dose-dependent manner. At the molecular level, isotalatizidine selectively increased the phosphorylation of p38 and ERK1/2, in addition to activating the transcription factor CREB and increasing dynorphin A production in cultured primary microglia. However, the downstream effects of isotalatizidine were abrogated by the selective ERK1/2 inhibitor U0126-EtOH or CREB inhibitor of KG-501, but not by the p38 inhibitor SB203580. The results also were confirmed in in vivo experiments. Conclusion Taken together, isotalatizidine specifically activates the ERK1/2 pathway and subsequently CREB, which triggers dynorphin A release in the microglia, eventually leading to its anti-nociceptive action.
Collapse
Affiliation(s)
- Shuai Shao
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Huan Xia
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Min Hu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Chengjuan Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Junmin Fu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Gaona Shi
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Qinglan Guo
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yu Zhou
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Wenjie Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jiangong Shi
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Tiantai Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| |
Collapse
|
44
|
Reinecke BA, Kang G, Zheng Y, Obeng S, Zhang H, Selley DE, An J, Zhang Y. Design and synthesis of a bivalent probe targeting the putative mu opioid receptor and chemokine receptor CXCR4 heterodimer. RSC Med Chem 2020; 11:125-131. [PMID: 33479612 PMCID: PMC7451026 DOI: 10.1039/c9md00433e] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 10/18/2019] [Indexed: 12/12/2022] Open
Abstract
Opioid abuse and HIV/AIDS have been defined as synergistic epidemics. Opioids can accelerate HIV replication in the immune system by up-regulating the expression of HIV co-receptor CXCR4. Several hypotheses have been suggested as the mechanism of CXCR4 modulation by opioids through their activation on the mu opioid receptor (MOR). One hypothesis is the putative heterodimerization of the MOR and CXCR4 as a mechanism of cross-talk and subsequent exacerbation of HIV replication. Bivalent chemical probes can be powerful molecular tools to characterize protein-protein interactions, and modulate the function related to such interactions. Herein we report the design and synthesis of a novel bivalent probe to explore the putative MOR-CXCR4 dimerization and its potential pharmacological role in enhancing HIV progression. The developed bivalent probe was designed with two distinct pharmacophores linked through a spacer. One pharmacophore (naltrexone) will interact with the MOR and the other (IT1t) with the CXCR4. The overall synthetic routes to prepare the bivalent probe and its corresponding monovalent controls were comprised of 18-22 steps with acceptable yields. Preliminary biological evaluation showed that the bivalent probe preserved binding affinity and functional activity at both respective receptors, supporting the initial molecular design.
Collapse
Affiliation(s)
- Bethany A Reinecke
- Department of Medicinal Chemistry , Virginia Commonwealth University , 800 East Leigh Street , Richmond , VA 23298 , USA . ; ; Tel: +1 804 828 0021
| | - Guifeng Kang
- Department of Medicinal Chemistry , Virginia Commonwealth University , 800 East Leigh Street , Richmond , VA 23298 , USA . ; ; Tel: +1 804 828 0021
| | - Yi Zheng
- Department of Medicinal Chemistry , Virginia Commonwealth University , 800 East Leigh Street , Richmond , VA 23298 , USA . ; ; Tel: +1 804 828 0021
| | - Samuel Obeng
- Department of Medicinal Chemistry , Virginia Commonwealth University , 800 East Leigh Street , Richmond , VA 23298 , USA . ; ; Tel: +1 804 828 0021
| | - Huijun Zhang
- Department of Medicine , Division of Infectious Diseases , School of Medicine , University of California San Diego , 9500 Gilman Drive, Stein Clinical Research Building, Suite 410 , La Jolla , CA 92093 , USA
| | - Dana E Selley
- Department of Pharmacology and Toxicology , Virginia Commonwealth University , 410 North 12th Street , VA 23298 , USA
| | - Jing An
- Department of Medicine , Division of Infectious Diseases , School of Medicine , University of California San Diego , 9500 Gilman Drive, Stein Clinical Research Building, Suite 410 , La Jolla , CA 92093 , USA
| | - Yan Zhang
- Department of Medicinal Chemistry , Virginia Commonwealth University , 800 East Leigh Street , Richmond , VA 23298 , USA . ; ; Tel: +1 804 828 0021
| |
Collapse
|
45
|
Abstract
Research on the effects of opioids on immune responses was stimulated in the 1980s by the intersection of use of intravenous heroin and HIV infection, to determine if opioids were enhancing HIV progression. The majority of experiments administering opioid alkaloids (morphine and heroin) in vivo, or adding these drugs to cell cultures in vitro, showed that they were immunosuppressive. Immunosuppression was reported as down-regulation: of Natural Killer cell activity; of responses of T and B cells to mitogens; of antibody formation in vivo and in vitro; of depression of phagocytic and microbicidal activity of neutrophils and macrophages; of cytokine and chemokine production by macrophages, microglia, and astrocytes; by sensitization to various infections using animal models; and by enhanced replication of HIV in vitro. The specificity of the receptor involved in the immunosuppression was shown to be the mu opioid receptor (MOR) by using pharmacological antagonists and mice genetically deficient in MOR. Beginning with a paper published in 2005, evidence was presented that morphine is immune-stimulating via binding to MD2, a molecule associated with Toll-like Receptor 4 (TLR4), the receptor for bacterial lipopolysaccharide (LPS). This concept was pursued to implicate inflammation as a mechanism for the psychoactive effects of the opioid. This review considers the validity of this hypothesis and concludes that it is hard to sustain. The experiments demonstrating immunosuppression were carried out in vivo in rodent strains with normal levels of TLR4, or involved use of cells taken from animals that were wild-type for expression of TLR4. Since engagement of TLR4 is universally accepted to result in immune activation by up-regulation of NF-κB, if morphine were binding to TLR4, it would be predicted that opioids would have been found to be pro-inflammatory, which they were not. Further, morphine is immunosuppressive in mice with a defective TLR4 receptor. Morphine and morphine withdrawal have been shown to permit leakage of Gram-negative bacteria and LPS from the intestinal lumen. LPS is the major ligand for TLR4. It is proposed that an occult variable in experiments where morphine is being proposed to activate TLR4 is actually underlying sepsis induced by the opioid.
Collapse
Affiliation(s)
- Toby K. Eisenstein
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| |
Collapse
|
46
|
Chronic dry eye induced corneal hypersensitivity, neuroinflammatory responses, and synaptic plasticity in the mouse trigeminal brainstem. J Neuroinflammation 2019; 16:268. [PMID: 31847868 PMCID: PMC6918709 DOI: 10.1186/s12974-019-1656-4] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 11/25/2019] [Indexed: 12/17/2022] Open
Abstract
Background Dry eye disease (DED) is a multifactorial disease associated with ocular surface inflammation, pain, and nerve abnormalities. We studied the peripheral and central neuroinflammatory responses that occur during persistent DED using molecular, cellular, behavioral, and electrophysiological approaches. Methods A mouse model of DED was obtained by unilateral excision of the extraorbital lachrymal gland (ELG) and Harderian gland (HG) of adult female C57BL/6 mice. In vivo tests were conducted at 7, 14, and 21 days (d) after surgery. Tear production was measured by a phenol red test and corneal alterations and inflammation were assessed by fluorescein staining and in vivo confocal microscopy. Corneal nerve morphology was evaluated by nerve staining. Mechanical corneal sensitivity was monitored using von Frey filaments. Multi-unit extracellular recording of ciliary nerve fiber activity was used to monitor spontaneous corneal nerve activity. RT-qPCR and immunostaining were used to determine RNA and protein levels at d21. Results We observed a marked reduction of tear production and the development of corneal inflammation at d7, d14, and d21 post-surgery in DED animals. Chronic DE induced a reduction of intraepithelial corneal nerve terminals. Behavioral and electrophysiological studies showed that the DED animals developed time-dependent mechanical corneal hypersensitivity accompanied by increased spontaneous ciliary nerve fiber electrical activity. Consistent with these findings, DED mice exhibited central presynaptic plasticity, demonstrated by a higher Piccolo immunoreactivity in the ipsilateral trigeminal brainstem sensory complex (TBSC). At d21 post-surgery, mRNA levels of pro-inflammatory (IL-6 and IL-1β), astrocyte (GFAP), and oxidative (iNOS2 and NOX4) markers increased significantly in the ipsilateral trigeminal ganglion (TG). This correlated with an increase in Iba1, GFAP, and ATF3 immunostaining in the ipsilateral TG of DED animals. Furthermore, pro-inflammatory cytokines (IL-6, TNFα, IL-1β, and CCL2), iNOS2, neuronal (ATF3 and FOS), and microglial (CD68 and Itgam) markers were also upregulated in the TBSC of DED animals at d21, along with increased immunoreactivity against GFAP and Iba1. Conclusions Overall, these data highlight peripheral sensitization and neuroinflammatory responses that participate in the development and maintenance of dry eye-related pain. This model may be useful to identify new analgesic molecules to alleviate ocular pain.
Collapse
|
47
|
Zortea M, Ramalho L, Alves RL, Alves CFDS, Braulio G, Torres ILDS, Fregni F, Caumo W. Transcranial Direct Current Stimulation to Improve the Dysfunction of Descending Pain Modulatory System Related to Opioids in Chronic Non-cancer Pain: An Integrative Review of Neurobiology and Meta-Analysis. Front Neurosci 2019; 13:1218. [PMID: 31803005 PMCID: PMC6876542 DOI: 10.3389/fnins.2019.01218] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 10/29/2019] [Indexed: 12/20/2022] Open
Abstract
Background: Opioid long-term therapy can produce tolerance, opioid-induced hyperalgesia (OIH), and it induces dysfunction in pain descending pain inhibitory system (DPIS). Objectives: This integrative review with meta-analysis aimed: (i) To discuss the potential mechanisms involved in analgesic tolerance and opioid-induced hyperalgesia (OIH). (ii) To examine how the opioid can affect the function of DPIS. (ii) To show evidence about the tDCS as an approach to treat acute and chronic pain. (iii) To discuss the effect of tDCS on DPIS and how it can counter-regulate the OIH. (iv) To draw perspectives for the future about the tDCS effects as an approach to improve the dysfunction in the DPIS in chronic non-cancer pain. Methods: Relevant published randomized clinical trials (RCT) comparing active (irrespective of the stimulation protocol) to sham tDCS for treating chronic non-cancer pain were identified, and risk of bias was assessed. We searched trials in PubMed, EMBASE and Cochrane trials databases. tDCS protocols accepted were application in areas of the primary motor cortex (M1), dorsolateral prefrontal cortex (DLPFC), or occipital area. Results: Fifty-nine studies were fully reviewed, and 24 with moderate to the high-quality methodology were included. tDCS improved chronic pain with a moderate effect size [pooled standardized mean difference; -0.66; 95% confidence interval (CI) -0.91 to -0.41]. On average, active protocols led to 27.26% less pain at the end of treatment compared to sham [95% CI; 15.89-32.90%]. Protocol varied in terms of anodal or cathodal stimulation, areas of stimulation (M1 and DLPFC the most common), number of sessions (from 5 to 20) and current intensity (from 1 to 2 mA). The time of application was 20 min in 92% of protocols. Conclusion: In comparison with sham stimulation, tDCS demonstrated a superior effect in reducing chronic pain conditions. They give perspectives that the top-down neuromodulator effects of tDCS are a promising approach to improve management in refractory chronic not-cancer related pain and to enhance dysfunctional neuronal circuitries involved in the DPIS and other pain dimensions and improve pain control with a therapeutic opioid-free. However, further studies are needed to determine individualized protocols according to a biopsychosocial perspective.
Collapse
Affiliation(s)
- Maxciel Zortea
- Post-graduation Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, Brazil.,Laboratory of Pain & Neuromodulation, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| | - Leticia Ramalho
- Post-graduation Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, Brazil.,Laboratory of Pain & Neuromodulation, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| | - Rael Lopes Alves
- Post-graduation Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, Brazil.,Laboratory of Pain & Neuromodulation, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| | - Camila Fernanda da Silveira Alves
- Post-graduation Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, Brazil.,Laboratory of Pain & Neuromodulation, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| | - Gilberto Braulio
- Laboratory of Pain & Neuromodulation, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil.,Service of Anesthesia and Perioperative Medicine, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| | - Iraci Lucena da Silva Torres
- Department of Pharmacology, Institute of Health Sciences (ICBS), Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, Brazil.,Pharmacology of Pain and Neuromodulation: Pre-clinical Investigations Research Group, Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, Brazil
| | - Felipe Fregni
- Neuromodulation Center, Spaulding Rehabilitation Hospital, Harvard Medical School, Boston, MA, United States
| | - Wolnei Caumo
- Post-graduation Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, Brazil.,Laboratory of Pain & Neuromodulation, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil.,Pain Treatment and Palliative Medicine Service, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| |
Collapse
|
48
|
Vitoux MA, Kessal K, Melik Parsadaniantz S, Claret M, Guerin C, Baudouin C, Brignole-Baudouin F, Réaux-Le Goazigo A. Benzalkonium chloride-induced direct and indirect toxicity on corneal epithelial and trigeminal neuronal cells: proinflammatory and apoptotic responses in vitro. Toxicol Lett 2019; 319:74-84. [PMID: 31707104 DOI: 10.1016/j.toxlet.2019.10.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 10/03/2019] [Accepted: 10/18/2019] [Indexed: 11/17/2022]
Abstract
Benzalkonium chloride (BAK), a quaternary ammonium compound widely used as disinfecting agent as well as preservative in eye drops is known to induce toxic effects on the ocular surface with inflammation and corneal nerve damage leading to dry eye disease (DED) in the medium-to-long term. The aim of this study was to evaluate in vitro the toxicity of a conditioned medium produced by corneal epithelial cells previously exposed to BAK (BAK-CM) on trigeminal neuronal cells. A human corneal epithelial (HCE) cell line was exposed to 5.10-3% BAK (i.e. 0.005% BAK) for 15 min and let recover for 5 h to prepare a BAK-CM. This BAK concentration is the lowest one found in eye drops. After this recovery period, BAK effect on HCE cells displayed cytotoxicity, morphological alteration, apoptosis, oxidative stress, ATP release, CCL2 and IL6 gene induction, as well as an increase in CCL2, IL-6 and MIF release. Next, a mouse trigeminal ganglion primary culture was exposed to the BAK-CM for 2 h, 4 h or 24 h. Whereas BAK-CM did not alter neuronal cell morphology, or induced neuronal cytotoxicity or oxidative stress, BAK-CM induced gene expression of Fos (neuronal activation marker), Atf3 (neuronal injury marker), Ccl2 and Il6 (inflammatory markers). Two and 4 h BAK-CM exposure promoted a neuronal damage (ATF-3, phospho-p38 increases; phospho-Stat3 decreases) while 24 h-BAK-CM exposure initiated a prosurvival pathway activation (phospho-p44/42, phospho-Akt increases; ATF-3, GADD153, active Caspase-3 decreases). In conclusion, this in vitro model, simulating paracrine mechanisms, represents an interesting tool to highlight the indirect toxic effects of BAK or any other xenobiotic on corneal trigeminal neurons and may help to better understand the cellular mechanisms that occur during DED pathophysiology.
Collapse
Affiliation(s)
- Michael-Adrien Vitoux
- Sorbonne Université, INSERM, CNRS, IHU Foresight, Institut de la Vision, F-75012 Paris, France; HORUS PHARMA, F-06700 Saint-Laurent-du-Var, France
| | - Karima Kessal
- Sorbonne Université, INSERM, CNRS, IHU Foresight, Institut de la Vision, F-75012 Paris, France; Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, INSERM-DGOS CIC 1423, IHU Foresight, F-75012 Paris, France
| | | | | | | | - Christophe Baudouin
- Sorbonne Université, INSERM, CNRS, IHU Foresight, Institut de la Vision, F-75012 Paris, France; Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, INSERM-DGOS CIC 1423, IHU Foresight, F-75012 Paris, France; Université Versailles-Saint-Quentin-en-Yvelines, Hôpital Ambroise Paré, APHP, F-92100 Boulogne-Billancourt, France
| | - Françoise Brignole-Baudouin
- Sorbonne Université, INSERM, CNRS, IHU Foresight, Institut de la Vision, F-75012 Paris, France; Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, INSERM-DGOS CIC 1423, IHU Foresight, F-75012 Paris, France; Université de Paris, Faculté de Pharmacie de Paris, F-75006 Paris, France
| | | |
Collapse
|
49
|
Abstract
OBJECTIVE Trigeminal neuralgia (TN) is a common cranial nerve disease. Inflammation is suggested in many recent studies to be involved in neuropathic pain, but its role in TN remains unclear so far. Therefore, the current study aimed to explore the relationship of inflammation with TN. METHODS The levels of inflammatory markers, such as white blood cell (WBC), neutrophil (NE), lymphocyte (LY), monocyte (MO), platelet (PLT), and albumin (ALB), as well as the neutrophil/lymphocyte ratio (NLR), derived NLR (dNLR), platelet/lymphocyte ratio (PLR), monocyte/lymphocyte ratio (MLR), and prognostic nutritional index (PNI) had been compared between TN patients and healthy controls using nonparametric tests. Moreover, multiple logistic regression models had been employed to assess the associations of inflammatory markers with TN. Besides, the receiver operating characteristic (ROC) curve was plotted to analyze the values of these inflammatory makers, as well as their matched combinations in diagnosing TN. RESULTS The levels of WBC, NE, MO, NLR, dNLR, and MLR in TN patients were evidently increased combined with those in normal subjects. In addition, multivariate logistic regression models illustrated that inflammation had close correlation with TN. Meanwhile, the area under the curve (AUC) values for NE, NLR, and dNLR, as well as those for the matched combinations of NLR+PLR, NLR+PNI, dNLR+NLR, and dNLR+PLR in TN were >0.7, which might have predictive value for TN compared with those for normal subjects. CONCLUSIONS Findings of this study reveal that inflammation could have played a close and important role in the progression and etiology of TN.
Collapse
|
50
|
Sahley TL, Anderson DJ, Hammonds MD, Chandu K, Musiek FE. Evidence for a dynorphin-mediated inner ear immune/inflammatory response and glutamate-induced neural excitotoxicity: an updated analysis. J Neurophysiol 2019; 122:1421-1460. [DOI: 10.1152/jn.00595.2018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Acoustic overstimulation (AOS) is defined as the stressful overexposure to high-intensity sounds. AOS is a precipitating factor that leads to a glutamate (GLU)-induced Type I auditory neural excitotoxicity and an activation of an immune/inflammatory/oxidative stress response within the inner ear, often resulting in cochlear hearing loss. The dendrites of the Type I auditory neural neurons that innervate the inner hair cells (IHCs), and respond to the IHC release of the excitatory neurotransmitter GLU, are themselves directly innervated by the dynorphin (DYN)-bearing axon terminals of the descending brain stem lateral olivocochlear (LOC) system. DYNs are known to increase GLU availability, potentiate GLU excitotoxicity, and induce superoxide production. DYNs also increase the production of proinflammatory cytokines by modulating immune/inflammatory signal transduction pathways. Evidence is provided supporting the possibility that the GLU-mediated Type I auditory neural dendritic swelling, inflammation, excitotoxicity, and cochlear hearing loss that follow AOS may be part of a brain stem-activated, DYN-mediated cascade of inflammatory events subsequent to a LOC release of DYNs into the cochlea. In support of a DYN-mediated cascade of events are established investigations linking DYNs to the immune/inflammatory/excitotoxic response in other neural systems.
Collapse
Affiliation(s)
- Tony L. Sahley
- Department of Biological, Geological, and Environmental Sciences, Cleveland State University, Cleveland, Ohio
- School of Health Sciences, Cleveland State University, Cleveland, Ohio
| | - David J. Anderson
- Department of Chemistry, Cleveland State University, Cleveland, Ohio
| | | | - Karthik Chandu
- Department of Chemistry, Cleveland State University, Cleveland, Ohio
| | - Frank E. Musiek
- Department of Speech, Language, and Hearing Sciences, University of Arizona, Tucson, Arizona
| |
Collapse
|