1
|
Zhu MY, Dong WY, Guo JR, Huang JY, Cheng PK, Yang Y, Liu A, Yang XL, Zhu X, Zhang Z, Wang Y, Tao W. A Neural Circuit For Bergamot Essential Oil-Induced Anxiolytic Effects. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2406766. [PMID: 39487959 DOI: 10.1002/advs.202406766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 09/08/2024] [Indexed: 11/04/2024]
Abstract
Aromatic essential oils have been shown to relieve anxiety and enhance relaxation, although the neural circuits underlying these effects have remained unknown. Here, it is found that treatment with 1.0% bergamot essential oil (BEO) exerts anxiolytic-like effects through a neural circuit projecting from the anterior olfactory nucleus (AON) to the anterior cingulate cortex (ACC) in acute restraint stress model mice. Collectively, in vivo two-photon calcium imaging, viral tracing, and whole-cell patch clamp recordings show that inhalation exposure to 1.0% BEO can activate glutamatergic projections from the AON to GABAergic neurons in the ACC, which drives inhibition of local glutamatergic neurons (AONGlu→ACCGABA→Glu). Optogenetic or chemogenetic manipulation of this pathway can recapitulate or abolish the BEO-induced anxiolytic-like behavioral effects in mice with ARS. Beyond depicting a previously unrecognized pathway involved in stress response, this study provides a circuit mechanism for the effects of BEO and suggests a potential target for anxiety treatment.
Collapse
Affiliation(s)
- Meng-Yu Zhu
- College & Hospital of Stomatology, Anhui Medical University, Key Lab of Oral Diseases Research of Anhui Province, Hefei, 230032, China
- Department of Physiology, Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Wan-Ying Dong
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Jin-Rong Guo
- Department of Physiology, Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Ji-Ye Huang
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Ping-Kai Cheng
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Yumeng Yang
- College & Hospital of Stomatology, Anhui Medical University, Key Lab of Oral Diseases Research of Anhui Province, Hefei, 230032, China
- Department of Physiology, Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - An Liu
- College & Hospital of Stomatology, Anhui Medical University, Key Lab of Oral Diseases Research of Anhui Province, Hefei, 230032, China
- Department of Physiology, Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Xin-Lu Yang
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Xia Zhu
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Zhi Zhang
- Department of Physiology, Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
- Center for Advanced Interdisciplinary Science and Biomedicine, Institute of Health and Medicine, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Yuanyin Wang
- College & Hospital of Stomatology, Anhui Medical University, Key Lab of Oral Diseases Research of Anhui Province, Hefei, 230032, China
| | - Wenjuan Tao
- College & Hospital of Stomatology, Anhui Medical University, Key Lab of Oral Diseases Research of Anhui Province, Hefei, 230032, China
- Department of Physiology, Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| |
Collapse
|
2
|
Fan Y, Xu Y, Huo Z, Zhang H, Peng L, Jiang X, Thomson AW, Dai H. Role of triggering receptor expressed on myeloid cells-1 in kidney diseases: A biomarker and potential therapeutic target. Chin Med J (Engl) 2024; 137:1663-1673. [PMID: 38809056 PMCID: PMC11268828 DOI: 10.1097/cm9.0000000000003197] [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: 02/05/2024] [Indexed: 05/30/2024] Open
Abstract
ABSTRACT Triggering receptor expressed on myeloid cells-1 (TREM-1) is a member of the immunoglobulin superfamily. As an amplifier of the inflammatory response, TREM-1 is mainly involved in the production of inflammatory mediators and the regulation of cell survival. TREM-1 has been studied in infectious diseases and more recently in non-infectious disorders. More and more studies have shown that TREM-1 plays an important pathogenic role in kidney diseases. There is evidence that TREM-1 can not only be used as a biomarker for diagnosis of disease but also as a potential therapeutic target to guide the development of novel therapeutic agents for kidney disease. This review summarized molecular biology of TREM-1 and its signaling pathways as well as immune response in the progress of acute kidney injury, renal fibrosis, diabetic nephropathy, immune nephropathy, and renal cell carcinoma.
Collapse
Affiliation(s)
- Yuxi Fan
- Department of Immunology, School of Basic Medical Science, Central South University, Changsha, Hunan 410013, China
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Ye Xu
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
- Medical College of Guangxi University, Nanning, Guangxi 530004, China
| | - Zhi Huo
- Department of Immunology, School of Basic Medical Science, Central South University, Changsha, Hunan 410013, China
| | - Hedong Zhang
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Longkai Peng
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Xin Jiang
- Department of Organ Transplantation, The Fifth Clinical Medical College of Henan University of Chinese Medicine (Zhengzhou People’s Hospital), Zhengzhou, Henan 450000, China
| | - Angus W. Thomson
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Helong Dai
- Department of Kidney Transplantation, Center of Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| |
Collapse
|
3
|
Aydogan Baykara R, Kiran TR, Otlu Ö, Erdem M, Pihtili Taş N. Could TREM-1 be a novel marker in the diagnosis of fibromyalgia?: A cross-sectional study. Medicine (Baltimore) 2024; 103:e38806. [PMID: 38996167 PMCID: PMC11245259 DOI: 10.1097/md.0000000000038806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 06/12/2024] [Indexed: 07/14/2024] Open
Abstract
Triggering receptors expressed on myeloid cells-1 (TREM-1) are transmembrane molecules expressed in cells of the immune system. Activation of TREM-1 leads to the release of pro-inflammatory mediators, which act as amplifiers of inflammation and thereby contribute to the pathogenesis of various diseases, whether inflammatory or not. This study explored the role of TREM-1 in the etiopathogenic context of fibromyalgia syndrome (FMS) and its association with disease activity. This randomized controlled and observational study included 45 patients diagnosed with FMS according to the 2016 American College of Rheumatology criteria. Serum TREM-1 levels were assessed using ELISA, and disease activity was measured using various scales such as the fibromyalgia impact questionnaire (FIQ). Patients were divided into 2 groups according to disease severity based on the FIQ score. Compared to a control group of 46 healthy individuals, patients with FMS exhibited significantly elevated concentrations of TREM-1 (mean ± SD = 216.97 pg/mL ± 16.04), P < .05. The FIQ, Pittsburgh sleep quality index, hospital anxiety and depression scale, fatigue severity scale, and visual analog scale, which confirm symptoms such as pain, disease severity, sleep disturbance, depression, anxiety, and fatigue seen in FMS was significantly correlated with TREM-1 level (P < .001). The optimal threshold value for TREM-1 to disease activity was determined to be 182.250, showing (area under the curve) (CI (95%)): [0.940] (0.887-0.993), a sensitivity of 97% and a specificity of 89% according to the receiver operating characteristic analysis. The positive correlation of TREM-1 with various symptom severity scales and hematological inflammatory indices may be a suitable biomarker for the diagnosis of FMS and a potential therapeutic target.
Collapse
Affiliation(s)
- Rabia Aydogan Baykara
- Department of Physical Medicine and Rehabilitation, Malatya Turgut Özal University, Malatya, Turkey
| | - Tuğba Raika Kiran
- Department of Medical Biochemistry, Malatya Turgut Özal University, Malatya, Turkey
| | - Önder Otlu
- Department of Medical Biochemistry, Malatya Turgut Özal University, Malatya, Turkey
| | - Mehmet Erdem
- Department of Medical Biochemistry, Malatya Turgut Özal University, Malatya, Turkey
| | - Nevsun Pihtili Taş
- Department of Physical Medicine and Rehabilitation, Health Sciences University, Fethi Sekin City Hospital, Elaziğ, Turkey
| |
Collapse
|
4
|
Zhao T, Zhou Y, Zhang D, Han D, Ma J, Li S, Li T, Hu S, Li Z. Inhibition of TREM-1 alleviates neuroinflammation by modulating microglial polarization via SYK/p38MAPK signaling pathway after traumatic brain injury. Brain Res 2024; 1834:148907. [PMID: 38570153 DOI: 10.1016/j.brainres.2024.148907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/24/2024] [Accepted: 03/31/2024] [Indexed: 04/05/2024]
Abstract
BACKGROUND Traumatic brain injury (TBI), as a major public health problem, is characterized by high incidence rate, disability rate, and mortality rate. Neuroinflammation plays a crucial role in the pathogenesis of TBI. Triggering receptor expressed on myeloid cells-1 (TREM-1) is recognized as an amplifier of the inflammation in diseases of the central nervous system (CNS). However, the function of TREM-1 remains unclear post-TBI. This study aimed to investigate the function of TREM-1 in neuroinflammation induced by TBI. METHODS Brain water content (BWC), modified neurological severity score (mNSS), and Morris Water Maze (MWM) were measured to evaluate the effect of TREM-1 inhibition on nervous system function and outcome after TBI. TREM-1 expression in vivo was evaluated by Western blotting. The cellular localization of TREM-1 in the damaged region was observed via immunofluorescence staining. We also conducted Western blotting to examine expression of SYK, p-SYK and other downstream proteins. RESULTS We found that inhibition of TREM-1 reduced brain edema, decreased mNSS and improved neurobehavioral outcomes after TBI. It was further determined that TREM-1 was expressed on microglia and modulated subtype transition of microglia. Inhibition of TREM-1 alleviated neuroinflammation, which was associated with SYK/p38MAPK signaling pathway. CONCLUSIONS These findings suggest that TREM-1 can be a potential clinical therapeutic target for alleviating neuroinflammation after TBI.
Collapse
Affiliation(s)
- Tianqi Zhao
- Department of Forensic Science, Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Medical Engineering Research Center of Gene Detection, Xuzhou, Jiangsu, China
| | - Yuxin Zhou
- Department of Forensic Science, Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Medical Engineering Research Center of Gene Detection, Xuzhou, Jiangsu, China
| | - Dabing Zhang
- Department of Forensic Science, Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Medical Engineering Research Center of Gene Detection, Xuzhou, Jiangsu, China
| | - Dong Han
- Laboratory of Emergency Medicine, Second Clinical Medical College of Xuzhou Medical University, Xuzhou, Jiangsu, China; Xuzhou Key Laboratory of Emergency Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jingyuan Ma
- Department of Forensic Science, Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Medical Engineering Research Center of Gene Detection, Xuzhou, Jiangsu, China
| | - Shanshan Li
- Department of Forensic Science, Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Medical Engineering Research Center of Gene Detection, Xuzhou, Jiangsu, China
| | - Ting Li
- Jiangsu Medical Engineering Research Center of Gene Detection, Xuzhou, Jiangsu, China; School of Life Sciences, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Shuqun Hu
- Laboratory of Emergency Medicine, Second Clinical Medical College of Xuzhou Medical University, Xuzhou, Jiangsu, China; Xuzhou Key Laboratory of Emergency Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Zhouru Li
- Department of Forensic Science, Xuzhou Medical University, Xuzhou, Jiangsu, China; Jiangsu Medical Engineering Research Center of Gene Detection, Xuzhou, Jiangsu, China.
| |
Collapse
|
5
|
Zhao L, Tao X, Wang K, Song Y, Zhang B, Yang L, Wang Z. Astaxanthin alleviates fibromyalgia pain and depression via NLRP3 inflammasome inhibition. Biomed Pharmacother 2024; 176:116856. [PMID: 38852510 DOI: 10.1016/j.biopha.2024.116856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/27/2024] [Accepted: 06/03/2024] [Indexed: 06/11/2024] Open
Abstract
Fibromyalgia is characterised by widespread chronic pain and is often accompanied by comorbidities such as sleep disorders, anxiety, and depression. Because it is often accompanied by many adverse symptoms and lack of effective treatment, it is important to search for the pathogenesis and treatment of fibromyalgia. Astaxanthin, a carotenoid pigment known for its anti-inflammatory and antioxidant properties, has demonstrated effective analgesic effects in neuropathic pain. However, its impact on fibromyalgia remains unclear. Therefore, in this study, we constructed a mouse model of fibromyalgia and investigated the effect of astaxanthin on chronic pain and associated symptoms through multiple intragastrical injections. We conducted behavioural assessments to detect pain and depression-like states in mice, recorded electroencephalograms to monitor sleep stages, examined c-Fos activation in the anterior cingulate cortex, measured activation of spinal glial cells, and assessed levels of inflammatory factors in the brain and spinal cord, including interleukin (IL)-1β, IL-6, and tumour necrosis factor- α(TNF-α).Additionally, we analysed the expression levels of IL-6, IL-10, NOD-like receptor thermal protein domain associated protein 3 (NLRP3), Apoptosis-associated speck-like protein containing CARD, and Caspase-1 proteins. The findings revealed that astaxanthin significantly ameliorated mechanical and thermal pain in mice with fibromyalgia and mitigated sleep disorders and depressive-like symptoms induced by pain. A potential mechanism underlying these effects is the anti-inflammatory action of astaxanthin, likely mediated through the inhibition of the NLRP3 inflammasome, which could be one of the pathways through which astaxanthin alleviates fibromyalgia. In conclusion, our study suggests that astaxanthin holds promise as a potential analgesic medication for managing fibromyalgia and its associated symptoms.
Collapse
Affiliation(s)
- Lin Zhao
- Department of Pain, The First Hospital of China Medical University, Shenyang 110001, People's Republic of China
| | - Xueshu Tao
- Department of Pain, The First Hospital of China Medical University, Shenyang 110001, People's Republic of China
| | - Kunpeng Wang
- Department of Pain, The First Hospital of China Medical University, Shenyang 110001, People's Republic of China
| | - Yuqing Song
- Department of Pain, The First Hospital of China Medical University, Shenyang 110001, People's Republic of China
| | - Bohan Zhang
- Department of Pain, The First Hospital of China Medical University, Shenyang 110001, People's Republic of China
| | - Li Yang
- Department of Anesthesiology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, People's Republic of China.
| | - Zhilin Wang
- Department of Pain, The First Hospital of China Medical University, Shenyang 110001, People's Republic of China.
| |
Collapse
|
6
|
Cao B, Xu Q, Shi Y, Zhao R, Li H, Zheng J, Liu F, Wan Y, Wei B. Pathology of pain and its implications for therapeutic interventions. Signal Transduct Target Ther 2024; 9:155. [PMID: 38851750 PMCID: PMC11162504 DOI: 10.1038/s41392-024-01845-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 04/08/2024] [Accepted: 04/25/2024] [Indexed: 06/10/2024] Open
Abstract
Pain is estimated to affect more than 20% of the global population, imposing incalculable health and economic burdens. Effective pain management is crucial for individuals suffering from pain. However, the current methods for pain assessment and treatment fall short of clinical needs. Benefiting from advances in neuroscience and biotechnology, the neuronal circuits and molecular mechanisms critically involved in pain modulation have been elucidated. These research achievements have incited progress in identifying new diagnostic and therapeutic targets. In this review, we first introduce fundamental knowledge about pain, setting the stage for the subsequent contents. The review next delves into the molecular mechanisms underlying pain disorders, including gene mutation, epigenetic modification, posttranslational modification, inflammasome, signaling pathways and microbiota. To better present a comprehensive view of pain research, two prominent issues, sexual dimorphism and pain comorbidities, are discussed in detail based on current findings. The status quo of pain evaluation and manipulation is summarized. A series of improved and innovative pain management strategies, such as gene therapy, monoclonal antibody, brain-computer interface and microbial intervention, are making strides towards clinical application. We highlight existing limitations and future directions for enhancing the quality of preclinical and clinical research. Efforts to decipher the complexities of pain pathology will be instrumental in translating scientific discoveries into clinical practice, thereby improving pain management from bench to bedside.
Collapse
Affiliation(s)
- Bo Cao
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Qixuan Xu
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
- Medical School of Chinese PLA, Beijing, 100853, China
| | - Yajiao Shi
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing, 100191, China
| | - Ruiyang Zhao
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
- Medical School of Chinese PLA, Beijing, 100853, China
| | - Hanghang Li
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
- Medical School of Chinese PLA, Beijing, 100853, China
| | - Jie Zheng
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing, 100191, China
| | - Fengyu Liu
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing, 100191, China.
| | - You Wan
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing, 100191, China.
| | - Bo Wei
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
7
|
Chen YH, Wang ZB, Liu XP, Xu JP, Mao ZQ. Sex differences in the relationship between depression and Alzheimer's disease-mechanisms, genetics, and therapeutic opportunities. Front Aging Neurosci 2024; 16:1301854. [PMID: 38903903 PMCID: PMC11188317 DOI: 10.3389/fnagi.2024.1301854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 04/25/2024] [Indexed: 06/22/2024] Open
Abstract
Depression and Alzheimer's disease (AD) are prevalent neuropsychiatric disorders with intriguing epidemiological overlaps. Their interrelation has recently garnered widespread attention. Empirical evidence indicates that depressive disorders significantly contribute to AD risk, and approximately a quarter of AD patients have comorbid major depressive disorder, which underscores the bidirectional link between AD and depression. A growing body of evidence substantiates pervasive sex differences in both AD and depression: both conditions exhibit a higher incidence among women than among men. However, the available literature on this topic is somewhat fragmented, with no comprehensive review that delineates sex disparities in the depression-AD correlation. In this review, we bridge these gaps by summarizing recent progress in understanding sex-based differences in mechanisms, genetics, and therapeutic prospects for depression and AD. Additionally, we outline key challenges in the field, holding potential for improving treatment precision and efficacy tailored to male and female patients' distinct needs.
Collapse
Affiliation(s)
- Yu-Han Chen
- The First Clinical Medical School, Hebei North University, Zhangjiakou, China
| | - Zhi-Bo Wang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Center for Neurological Disorders, Beijing, China
| | - Xi-Peng Liu
- Department of Neurosurgery, The First Affiliated Hospital of Hebei North, Zhangjiakou, China
| | - Jun-Peng Xu
- Department of Neurosurgery, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Zhi-Qi Mao
- Department of Neurosurgery, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
8
|
Deng Q, Parker E, Wu C, Zhu L, Liu TCY, Duan R, Yang L. Repurposing Ketamine in the Therapy of Depression and Depression-Related Disorders: Recent Advances and Future Potential. Aging Dis 2024:AD.2024.0239. [PMID: 38916735 DOI: 10.14336/ad.2024.0239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 04/29/2024] [Indexed: 06/26/2024] Open
Abstract
Depression represents a prevalent and enduring mental disorder of significant concern within the clinical domain. Extensive research indicates that depression is very complex, with many interconnected pathways involved. Most research related to depression focuses on monoamines, neurotrophic factors, the hypothalamic-pituitary-adrenal axis, tryptophan metabolism, energy metabolism, mitochondrial function, the gut-brain axis, glial cell-mediated inflammation, myelination, homeostasis, and brain neural networks. However, recently, Ketamine, an ionotropic N-methyl-D-aspartate (NMDA) receptor antagonist, has been discovered to have rapid antidepressant effects in patients, leading to novel and successful treatment approaches for mood disorders. This review aims to summarize the latest findings and insights into various signaling pathways and systems observed in depression patients and animal models, providing a more comprehensive view of the neurobiology of anxious-depressive-like behavior. Specifically, it highlights the key mechanisms of ketamine as a rapid-acting antidepressant, aiming to enhance the treatment of neuropsychiatric disorders. Moreover, we discuss the potential of ketamine as a prophylactic or therapeutic intervention for stress-related psychiatric disorders.
Collapse
Affiliation(s)
- Qianting Deng
- College of Physical Education and Sport Science, South China Normal University, Guangzhou, China
| | - Emily Parker
- Medical College of Georgia at Augusta University, Augusta, GA 30912, USA
| | - Chongyun Wu
- College of Physical Education and Sport Science, South China Normal University, Guangzhou, China
| | - Ling Zhu
- College of Physical Education and Sport Science, South China Normal University, Guangzhou, China
| | - Timon Cheng-Yi Liu
- College of Physical Education and Sport Science, South China Normal University, Guangzhou, China
| | - Rui Duan
- College of Physical Education and Sport Science, South China Normal University, Guangzhou, China
| | - Luodan Yang
- College of Physical Education and Sport Science, South China Normal University, Guangzhou, China
| |
Collapse
|
9
|
Jain A, Hakim S, Woolf CJ. Immune drivers of physiological and pathological pain. J Exp Med 2024; 221:e20221687. [PMID: 38607420 PMCID: PMC11010323 DOI: 10.1084/jem.20221687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 09/25/2023] [Accepted: 04/02/2024] [Indexed: 04/13/2024] Open
Abstract
Physiological pain serves as a warning of exposure to danger and prompts us to withdraw from noxious stimuli to prevent tissue damage. Pain can also alert us of an infection or organ dysfunction and aids in locating such malfunction. However, there are instances where pain is purely pathological, such as unresolved pain following an inflammation or injury to the nervous system, and this can be debilitating and persistent. We now appreciate that immune cells are integral to both physiological and pathological pain, and that pain, in consequence, is not strictly a neuronal phenomenon. Here, we discuss recent findings on how immune cells in the skin, nerve, dorsal root ganglia, and spinal cord interact with somatosensory neurons to mediate pain. We also discuss how both innate and adaptive immune cells, by releasing various ligands and mediators, contribute to the initiation, modulation, persistence, or resolution of various modalities of pain. Finally, we propose that the neuroimmune axis is an attractive target for pain treatment, but the challenges in objectively quantifying pain preclinically, variable sex differences in pain presentation, as well as adverse outcomes associated with immune system modulation, all need to be considered in the development of immunotherapies against pain.
Collapse
Affiliation(s)
- Aakanksha Jain
- F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA, USA
| | - Sara Hakim
- F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Clifford J. Woolf
- F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
10
|
Chang X, Zhang H, Chen S. Neural circuits regulating visceral pain. Commun Biol 2024; 7:457. [PMID: 38615103 PMCID: PMC11016080 DOI: 10.1038/s42003-024-06148-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 04/05/2024] [Indexed: 04/15/2024] Open
Abstract
Visceral hypersensitivity, a common clinical manifestation of irritable bowel syndrome, may contribute to the development of chronic visceral pain, which is a major challenge for both patients and health providers. Neural circuits in the brain encode, store, and transfer pain information across brain regions. In this review, we focus on the anterior cingulate cortex and paraventricular nucleus of the hypothalamus to highlight the progress in identifying the neural circuits involved in visceral pain. We also discuss several neural circuit mechanisms and emphasize the importance of cross-species, multiangle approaches and the identification of specific neurons in determining the neural circuits that control visceral pain.
Collapse
Affiliation(s)
- Xiaoli Chang
- College of Acupuncture and Massage, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
- Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| | - Haiyan Zhang
- Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Shaozong Chen
- Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| |
Collapse
|
11
|
Teng S, Yang Y, Zhang W, Li X, Li W, Cui Z, Min L, Wu J. Antidepressant fluoxetine alleviates colitis by reshaping intestinal microenvironment. Cell Commun Signal 2024; 22:176. [PMID: 38475799 PMCID: PMC10935910 DOI: 10.1186/s12964-024-01538-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 02/21/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND The impact of antidepressants on Inflammatory bowel diseases (IBD) has been extensively studied. However, the biological effects and molecular mechanisms of antidepressants in alleviating colitis remain unclear. METHODS We systematically assessed how antidepressants (fluoxetine, fluvoxamine and venlafaxine) affected IBD and chose fluoxetine, the most effective one, for mechanism studies. We treated the C56BL/6 mice of the IBD model with fluoxetine and their controls. We initially assessed the severity of intestinal inflammation in mice by body weight loss, disease Activity Index scores and the length of the colon. The H&E staining and immunohistochemical staining of MUC2 of colon sections were performed to observe the pathological changes. RT-qPCR and western blot were conducted to assess the expression level of the barrier and inflammation-associated genes. Then, single-cell RNA sequencing was performed on mouse intestinal mucosa. Seurat was used to visualize the data. Uniform Manifold Approximation and Projection (UMAP) was used to perform the dimensionality reduction. Cell Chat package was used to perform cell-cell communication analysis. Monocle was used to conduct developmental pseudotime analysis. Last, RT-qPCR, western blot and immunofluorescence staining were conducted to test the phenomenon discovered by single-cell RNA sequencing in vitro. RESULTS We found that fluoxetine treatment significantly alleviated colon inflammation. Notably, single-cell RNA sequencing analysis revealed that fluoxetine affected the distribution of different cell clusters, cell-cell communication and KEGG pathway enrichment. Under the treatment of fluoxetine, enterocytes, Goblet cells and stem cells became the dominating cells. The pseudotime analysis showed that there was a trend for M1 macrophages to differentiate into M2 macrophages. Lastly, we tested this phenomenon in vitro, which exhibited anti-inflammatory effects on enterocytes. CONCLUSIONS Fluoxetine exhibited anti-inflammatory effects on intestinal mucosa via remodeling of the intestinal cells and macrophages, which reveals that fluoxetine is a promising therapeutic drug for the treatment of IBD and psychiatric comorbidities.
Collapse
Affiliation(s)
- Shuo Teng
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, 100050, People's Republic of China
- Peking University Ninth School of Clinical Medicine, Beijing, 100038, China
| | - Yi Yang
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, 100050, People's Republic of China
| | - Wanru Zhang
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, 100050, People's Republic of China
| | - Xiangji Li
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, 100050, People's Republic of China
| | - Wenkun Li
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, 100050, People's Republic of China
| | - Zilu Cui
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, 100050, People's Republic of China
| | - Li Min
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, 100050, People's Republic of China.
| | - Jing Wu
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, 100050, People's Republic of China.
| |
Collapse
|
12
|
Li X, Jia Y, Xiong M, Gao Y, Xu X, Ke C. MHC-I in the hippocampus promotes comorbid depressive symptoms in bone cancer pain via the upregulation of microglial TREM2/DAP12 signaling. Behav Brain Res 2024; 461:114843. [PMID: 38176616 DOI: 10.1016/j.bbr.2023.114843] [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: 10/27/2023] [Revised: 12/17/2023] [Accepted: 12/28/2023] [Indexed: 01/06/2024]
Abstract
Pain and depression comorbidity affects patients' physical and mental health, as well as quality of life. Comorbid depressive symptoms in cancer pain have a severe impact on the recognition and treatment of pain. Similarly, cancer pain patients with depression are inclined towards more despair and greater impairment. The mechanisms responsible for the comorbid depressive symptoms in bone cancer pain (BCP) have not been fully delineated. Here, it was reported that the implantation of carcinoma cells into the femoral cavity of mice led to the upregulation of major histocompatibility complex class I (MHC-I) in the hippocampus. This was associated with the activation of microglial signaling pathway mediated by the triggering receptor expressed on myeloid cells 2 protein (TREM2) and DNAX-activating protein of 12 kDa (DAP12). Pain and depression-like behaviors were reversed by the knockdown of hippocampal MHC-I via a lentiviral vector harboring ribonucleic acid interference (RNAi) sequence. Moreover, MHC-I knockdown exhibited a marked reduction in the expression of TREM2 and DAP12. These results suggested that hippocampal MHC-I was involved in BCP and depression comorbidity via upregulating the signals mediated by TREM2/DAP12 in microglia. The suppression of MHC-I could be a potential therapeutic target for BCP.
Collapse
Affiliation(s)
- Xiaohui Li
- Institute of Anesthesiology & Pain (IAP), Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei Province 442000, PR China
| | - Yifu Jia
- Institute of Anesthesiology & Pain (IAP), Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei Province 442000, PR China
| | - Mengyuan Xiong
- Institute of Anesthesiology & Pain (IAP), Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei Province 442000, PR China
| | - Yan Gao
- Institute of Anesthesiology & Pain (IAP), Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei Province 442000, PR China
| | - Xueqin Xu
- Institute of Anesthesiology & Pain (IAP), Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei Province 442000, PR China
| | - Changbin Ke
- Institute of Anesthesiology & Pain (IAP), Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei Province 442000, PR China.
| |
Collapse
|
13
|
Liu Y, Wang L, Zhou C, Yuan Y, Fang B, Lu K, Xu F, Chen L, Huang L. MiR-31-5p regulates the neuroinflammatory response via TRAF6 in neuropathic pain. Biol Direct 2024; 19:10. [PMID: 38267979 PMCID: PMC10807213 DOI: 10.1186/s13062-023-00434-1] [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: 07/27/2023] [Accepted: 11/02/2023] [Indexed: 01/26/2024] Open
Abstract
BACKGROUND Neuropathic pain is chronic pain and has few effective control strategies. Studies have demonstrated that microRNAs have functions in neuropathic pain. However, no study has been conducted to demonstrate the role and mechanism of microRNA (miR)-31-5p in neuropathic pain. Accordingly, this study sought to determine the pathological role of miR-31-5p in chronic constriction injury (CCI) -induced neuropathic pain mouse models. METHODS We used CCI surgery to establish mouse neuropathic pain model. Behavioral tests were performed to evaluate pain sensitivity of mice. Expressions of miR-31-5p and inflammatory cytokines in dorsal root ganglion (DRG) were examined by polymerase chain reaction. Animals or cells were received with/without miR-31-5p mimic or inhibitor to investigate its role in neuropathic pain. The mechanism of miR-31-5p was assayed using western blotting, immunofluorescence staining and dual-luciferase reporter assay. RESULTS We found that CCI led to a significant decrease in miR-31-5p levels. Knockout of miR-31-5p and administration of miPEP31 exacerbated pain in C57BL/6 mice. Meanwhile, miR-31-5p overexpression increased the paw withdrawal threshold and latency. TRAF6 is one of the target gene of miR-31-5p, which can trigger a complex inflammatory response. TRAF6 was associated with pain and that reducing the DRG expression of TRAF6 could alleviate pain. In addition, miR-31-5p overexpression inhibited the TRAF6 expression and reduced the neuroinflammatory response. CONCLUSIONS All the results reveal that miR-31-5p could potentially alleviate pain in CCI mouse models by inhibiting the TRAF6 mediated neuroinflammatory response. MiR-31-5p upregulation is highlighted here as new target for CCI treatment.
Collapse
Affiliation(s)
- Yuqi Liu
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 20080, China
| | - Lijuan Wang
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 20080, China
| | - Chengcheng Zhou
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 20080, China
| | - Yuan Yuan
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 20080, China
| | - Bin Fang
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 20080, China
| | - Kaimei Lu
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 20080, China
| | - Fangxia Xu
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 20080, China.
| | - Lianhua Chen
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 20080, China.
| | - Lina Huang
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 20080, China.
| |
Collapse
|
14
|
Fakhfouri G, Mijailović NR, Rahimian R. Psychiatric Comorbidities of Inflammatory Bowel Disease: It Is a Matter of Microglia's Gut Feeling. Cells 2024; 13:177. [PMID: 38247868 PMCID: PMC10814793 DOI: 10.3390/cells13020177] [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: 12/07/2023] [Revised: 01/13/2024] [Accepted: 01/15/2024] [Indexed: 01/23/2024] Open
Abstract
Inflammatory bowel disease (IBD), a common term for Crohn's disease and ulcerative colitis, is a chronic, relapse-remitting condition of the gastrointestinal tract that is increasing worldwide. Psychiatric comorbidities, including depression and anxiety, are more prevalent in IBD patients than in healthy individuals. Evidence suggests that varying levels of neuroinflammation might underlie these states in IBD patients. Within this context, microglia are the crucial non-neural cells in the brain responsible for innate immune responses following inflammatory insults. Alterations in microglia's functions, such as secretory profile, phagocytic activity, and synaptic pruning, might play significant roles in mediating psychiatric manifestations of IBD. In this review, we discuss the role played by microglia in IBD-associated comorbidities.
Collapse
Affiliation(s)
- Gohar Fakhfouri
- Department of Psychiatry, Douglas Hospital, McGill University, Montreal, QC H4H 1R3, Canada;
| | - Nataša R. Mijailović
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Reza Rahimian
- McGill Group for Suicide Studies, Douglas Mental Health Institute, McGill University, 6875 Boulevard LaSalle, Montreal, QC H4H 1R3, Canada
| |
Collapse
|
15
|
Dou Z, Su N, Zhou Z, Mi A, Xu L, Zhou J, Sun S, Liu Y, Hao M, Li Z. Modulation of visceral pain by brain nuclei and brain circuits and the role of acupuncture: a narrative review. Front Neurosci 2023; 17:1243232. [PMID: 38027491 PMCID: PMC10646320 DOI: 10.3389/fnins.2023.1243232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 10/17/2023] [Indexed: 12/01/2023] Open
Abstract
Visceral pain is a complex and heterogeneous pain condition that is often associated with pain-related negative emotional states, including anxiety and depression, and can exert serious effects on a patient's physical and mental health. According to modeling stimulation protocols, the current animal models of visceral pain mainly include the mechanical dilatation model, the ischemic model, and the inflammatory model. Acupuncture can exert analgesic effects by integrating and interacting input signals from acupuncture points and the sites of pain in the central nervous system. The brain nuclei involved in regulating visceral pain mainly include the nucleus of the solitary tract, parabrachial nucleus (PBN), locus coeruleus (LC), rostral ventromedial medulla (RVM), anterior cingulate cortex (ACC), paraventricular nucleus (PVN), and the amygdala. The neural circuits involved are PBN-amygdala, LC-RVM, amygdala-insula, ACC-amygdala, claustrum-ACC, bed nucleus of the stria terminalis-PVN and the PVN-ventral lateral septum circuit. Signals generated by acupuncture can modulate the central structures and interconnected neural circuits of multiple brain regions, including the medulla oblongata, cerebral cortex, thalamus, and hypothalamus. This analgesic process also involves the participation of various neurotransmitters and/or receptors, such as 5-hydroxytryptamine, glutamate, and enkephalin. In addition, acupuncture can regulate visceral pain by influencing functional connections between different brain regions and regulating glucose metabolism. However, there are still some limitations in the research efforts focusing on the specific brain mechanisms associated with the effects of acupuncture on the alleviation of visceral pain. Further animal experiments and clinical studies are now needed to improve our understanding of this area.
Collapse
Affiliation(s)
- Zhiqiang Dou
- College of Acupuncture and Moxibustion and Tuina, Shandong University of Traditional Chinese Medicine, Ji’nan, China
| | - Na Su
- First Clinical Medicine College, Shandong University of Traditional Chinese Medicine, Ji’nan, China
| | - Ziyang Zhou
- College of Acupuncture and Moxibustion and Tuina, Shandong University of Traditional Chinese Medicine, Ji’nan, China
| | - Aoyue Mi
- College of Acupuncture and Moxibustion and Tuina, Shandong University of Traditional Chinese Medicine, Ji’nan, China
| | - Luyao Xu
- College of Acupuncture and Moxibustion and Tuina, Shandong University of Traditional Chinese Medicine, Ji’nan, China
| | - Jiazheng Zhou
- College of Acupuncture and Moxibustion and Tuina, Shandong University of Traditional Chinese Medicine, Ji’nan, China
| | - Sizhe Sun
- College of Acupuncture and Moxibustion and Tuina, Shandong University of Traditional Chinese Medicine, Ji’nan, China
| | - Yanyi Liu
- College of Acupuncture and Moxibustion and Tuina, Shandong University of Traditional Chinese Medicine, Ji’nan, China
| | - Mingyao Hao
- External Treatment Center of Traditional Chinese Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Ji’nan, China
| | - Zhaofeng Li
- College of Acupuncture and Moxibustion and Tuina, Shandong University of Traditional Chinese Medicine, Ji’nan, China
- International Office, Shandong University of Traditional Chinese Medicine, Ji’nan, China
| |
Collapse
|
16
|
Thomasi B, Valdetaro L, Ricciardi MC, Gonçalves de Carvalho M, Fialho Tavares I, Tavares-Gomes AL. Enteric glia as a player of gut-brain interactions during Parkinson's disease. Front Neurosci 2023; 17:1281710. [PMID: 38027511 PMCID: PMC10644407 DOI: 10.3389/fnins.2023.1281710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 10/10/2023] [Indexed: 12/01/2023] Open
Abstract
The enteric glia has been shown as a potential component of neuroimmune interactions that signal in the gut-brain axis during Parkinson's disease (PD). Enteric glia are a peripheral glial type found in the enteric nervous system (ENS) that, associated with enteric neurons, command various gastrointestinal (GI) functions. They are a unique cell type, with distinct phenotypes and distribution in the gut layers, which establish relevant neuroimmune modulation and regulate neuronal function. Comprehension of enteric glial roles during prodromal and symptomatic phases of PD should be a priority in neurogastroenterology research, as the reactive enteric glial profile, gastrointestinal dysfunction, and colonic inflammation have been verified during the prodromal phase of PD-a moment that may be interesting for interventions. In this review, we explore the mechanisms that should govern enteric glial signaling through the gut-brain axis to understand pathological events and verify the possible windows and pathways for therapeutic intervention. Enteric glia directly modulate several functional aspects of the intestine, such as motility, visceral sensory signaling, and immune polarization, key GI processes found deregulated in patients with PD. The search for glial biomarkers, the investigation of temporal-spatial events involving glial reactivity/signaling, and the proposal of enteric glia-based therapies are clearly demanded for innovative and intestine-related management of PD.
Collapse
Affiliation(s)
- Beatriz Thomasi
- Department of Physiology, Michigan State University, East Lansing, MI, United States
| | - Luisa Valdetaro
- Department of Molecular Pathobiology, NYU College of Dentistry, New York, NY, United States
| | - Maria Carolina Ricciardi
- Neuroglial Interaction Lab, Neuroscience Program, Universidade Federal Fluminense, Niterói, Brazil
| | | | - Isabela Fialho Tavares
- Neuroglial Interaction Lab, Neurobiology Department, Universidade Federal Fluminense, Niterói, Brazil
| | - Ana Lucia Tavares-Gomes
- Neuroglial Interaction Lab, Neuroscience Program, Universidade Federal Fluminense, Niterói, Brazil
- Neuroglial Interaction Lab, Neurobiology Department, Universidade Federal Fluminense, Niterói, Brazil
| |
Collapse
|