1
|
Yao C, Xie D, Zhang Y, Shen Y, Sun P, Ma Z, Li J, Tao J, Fang M. Tryptophan metabolism and ischemic stroke: An intricate balance. Neural Regen Res 2026; 21:466-477. [PMID: 40326980 DOI: 10.4103/nrr.nrr-d-24-00777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 11/27/2024] [Indexed: 05/07/2025] Open
Abstract
Ischemic stroke, which is characterized by hypoxia and ischemia, triggers a cascade of injury responses, including neurotoxicity, inflammation, oxidative stress, disruption of the blood-brain barrier, and neuronal death. In this context, tryptophan metabolites and enzymes, which are synthesized through the kynurenine and 5-hydroxytryptamine pathways, play dual roles. The delicate balance between neurotoxic and neuroprotective substances is a crucial factor influencing the progression of ischemic stroke. Neuroprotective metabolites, such as kynurenic acid, exert their effects through various mechanisms, including competitive blockade of N-methyl-D-aspartate receptors, modulation of α7 nicotinic acetylcholine receptors, and scavenging of reactive oxygen species. In contrast, neurotoxic substances such as quinolinic acid can hinder the development of vascular glucose transporter proteins, induce neurotoxicity mediated by reactive oxygen species, and disrupt mitochondrial function. Additionally, the enzymes involved in tryptophan metabolism play major roles in these processes. Indoleamine 2,3-dioxygenase in the kynurenine pathway and tryptophan hydroxylase in the 5-hydroxytryptamine pathway influence neuroinflammation and brain homeostasis. Consequently, the metabolites generated through tryptophan metabolism have substantial effects on the development and progression of ischemic stroke. Stroke treatment aims to restore the balance of various metabolite levels; however, precise regulation of tryptophan metabolism within the central nervous system remains a major challenge for the treatment of ischemic stroke. Therefore, this review aimed to elucidate the complex interactions between tryptophan metabolites and enzymes in ischemic stroke and develop targeted therapies that can restore the delicate balance between neurotoxicity and neuroprotection.
Collapse
Affiliation(s)
- Chongjie Yao
- Rehabilitation Department, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- School of Acupuncture-Moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Dong Xie
- Rehabilitation Department, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuchen Zhang
- Rehabilitation Department, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- School of Acupuncture-Moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuanhao Shen
- School of Acupuncture-Moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Pingping Sun
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhao Ma
- Rehabilitation Department, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jin Li
- Rehabilitation Department, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiming Tao
- Rehabilitation Department, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Min Fang
- Rehabilitation Department, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Research Institute of Tuina, Shanghai Academy of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
2
|
Chen X, Zhu M, Shi Q, Huang Z, Zhu J, Sun P, Zhang H, Yang L, Chen X, Zhang Y, Feng L, Horimoto K, Li F, Han F, Chen D. Proof of concept study for developing 1-thienyl-β-carboline derivatives as IDO1 and TDO dual inhibitors to treat Parkinson's disease complicating depression. Eur J Med Chem 2025; 291:117597. [PMID: 40215562 DOI: 10.1016/j.ejmech.2025.117597] [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: 01/10/2025] [Revised: 03/31/2025] [Accepted: 04/01/2025] [Indexed: 05/03/2025]
Abstract
Depressive symptoms are the most common neuropsychiatric disorders at all stages of Parkinson's disease (PD). Imbalances of the kynurenine pathway of tryptophan metabolism have been closely linked to the pathogenesis of PD and depression. Herein, we designed and synthesized a series of 1-thienyl-β-carboline derivatives as IDO1 and TDO dual inhibitors; among them, compound CZ-17 manifested moderate inhibitory activities to IDO1 and TDO with IC50 values of 0.33 and 1.78 μM, respectively. CZ-17 inhibited the kynurenine pathway of tryptophan degradation at the cellular level, and remarkably reduced the kynurenine/tryptophan ratio. CZ-17 displayed directly neuroprotective effect in corticosterone-induced PC12 neural cell injury model. In vivo experiments demonstrated that CZ-17 significantly increased dopamine and serotonin levels, improved MPTP-induced motor disability and rescued LPS-induced depressive behavior in zebrafish model. Acute toxicity tests of CZ-17 in zebrafish embryos showed no toxicity within the effective dose range. Additionally, CZ-17 displayed the potential to cross the BBB via passive diffusion according to ADMET prediction and Caco-2 permeability assay. Thus, CZ-17 may be a promising drug candidate for PD complicating depression.
Collapse
Affiliation(s)
- Xuan Chen
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China; School of Pharmaceutical Engineering, Jiangsu Food and Pharmaceutical Science College, Huaian, 223003, China
| | - Mengxiao Zhu
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Qingyuan Shi
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Zhenquan Huang
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Junjie Zhu
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Pingping Sun
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Hongzhen Zhang
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Lili Yang
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Xun Chen
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Yu Zhang
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China; Innovation Center of Suzhou Nanjing Medical University, Suzhou, 215000, Jiangsu, China; National Center of Technology Innovation for Biopharmaceuticals, Suzhou, 215000, Jiangsu, China
| | - Lili Feng
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | | | - Fei Li
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China.
| | - Feng Han
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China; Innovation Center of Suzhou Nanjing Medical University, Suzhou, 215000, Jiangsu, China; The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Northern Jiangsu Institute of Clinical Medicine, Huaian, 223300, China; Institute of Brain Science, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Dongyin Chen
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China; Innovation Center of Suzhou Nanjing Medical University, Suzhou, 215000, Jiangsu, China; National Center of Technology Innovation for Biopharmaceuticals, Suzhou, 215000, Jiangsu, China; The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Northern Jiangsu Institute of Clinical Medicine, Huaian, 223300, China.
| |
Collapse
|
3
|
Zhao T, Cui X, Zhang X, Zhao M, Rastegar-Kashkooli Y, Wang J, Li Q, Jiang C, Li N, Xing F, Han X, Zhang J, Xing N, Wang J, Wang J. Hippocampal sclerosis: A review on current research status and its mechanisms. Ageing Res Rev 2025; 108:102716. [PMID: 40058463 DOI: 10.1016/j.arr.2025.102716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 02/27/2025] [Accepted: 03/02/2025] [Indexed: 03/27/2025]
Abstract
Hippocampal sclerosis (HS) is a pathological condition characterized by significant loss of hippocampal neurons and gliosis. This condition represents the most common neuropathological change observed in patients with temporal lobe epilepsy (TLE) and is also found in aging individuals. TLE related to HS is the most prevalent type of drug-resistant epilepsy in adults, and its underlying mechanisms are not yet fully understood. Therefore, developing improved methods for predicting and treating drug-resistant patients with TLE-HS is crucial. Patients with TLE-HS often experience cognitive impairment and psychological comorbidities, significantly affecting their quality of life. Consequently, a thorough review of the current research status of TLE-HS is essential, focusing on its prediction, diagnosis, treatment, and underlying mechanisms. The hippocampus plays a pivotal role in memory and cognition. HS of aging (HS-Aging), a condition linked to dementia in the ultra-elderly, is marked by severe CA1 (cornu ammonis) neuronal loss and frequent transactive response DNA-binding protein of 43 kDa (TDP-43) proteinopathy, often misdiagnosed as Alzheimer's disease (AD). Nonetheless, clinical characteristics and patterns of hippocampal atrophy can help differentiate between the two disorders. This review aims to provide a comprehensive overview of the pathological features of HS, the relevant mechanisms underlying TLE-HS and HS-Aging, current imaging diagnostic techniques, including machine learning, and available treatment modalities. It also explores the prognosis and comorbidities related to these conditions. Future research directions include establishing animal models to clarify the poorly understood mechanisms underlying HS, particularly those related to emotional processing. Investigating post-HS behavioral and cognitive changes in these models will lay the foundation for further advancements in this field. This review is a cornerstone for future investigations and suggests additional research endeavors.
Collapse
Affiliation(s)
- Ting Zhao
- Department of Neurology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China.
| | - Xiaoxiao Cui
- Department of Neurology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China
| | - Xinru Zhang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Mengke Zhao
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Yousef Rastegar-Kashkooli
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; School of International Education, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Junyang Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Qiang Li
- Department of Neurology, Shanghai Gongli Hospital of Pudong New Area, Shanghai 200135, China
| | - Chao Jiang
- Department of Neurology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China
| | - Nan Li
- Department of Neurology, The 2nd Affiliated Hospital of Zhengzhou University, Zhengzhou 450014, China
| | - Fei Xing
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Xiong Han
- Department of Neurology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China
| | - Jiewen Zhang
- Department of Neurology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China
| | - Na Xing
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China.
| | - Junmin Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Jian Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| |
Collapse
|
4
|
Liu HX, Yang MK, Li YC, Liu CX, Li GP, Meng XL, Pei K, Wen SY. Shenqi granules enhance recovery from cerebral ischemia-reperfusion injury by modulating tryptophan and tyrosine metabolism and activating NFE2L2/NRF2. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156623. [PMID: 40068292 DOI: 10.1016/j.phymed.2025.156623] [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: 11/08/2024] [Revised: 03/04/2025] [Accepted: 03/06/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Stroke is a multifaceted physiological event linked to imbalances in gut microbiota and disruptions in metabolic pathways. Traditional Chinese medicines, leveraging the gut-brain axis, have been shown to significantly ameliorate ischemic stroke. However, the specific role and molecular mechanism of Shenqi granules (SQF) in enhancing the recovery from ischemic stroke remain to be elucidated. PURPOSE This study aims to explore the therapeutic effects of SQF on rats with cerebral ischemia-reperfusion injury (CIRI) and its regulatory effects on the gut microbiota, providing a basis for the clinical rational use of drugs in ischemic stroke. METHODS The study conducted a comprehensive biological assessment of SQF's role in improving CIRI at the whole-animal level. Subsequently, Weighted Gene Co-expression Network Analysis (WGCNA) and network pharmacology analysis were used for component analysis and target prediction. Then, the therapeutic targets of SQF were further validated through molecular docking and molecular experiments. Finally, an integrated omics approach combining fecal untargeted metabolome and 16S rRNA sequencing was employed to state the anti-CIRI effects of SQF and its potential mechanisms. RESULTS SQF alleviates cerebral infarct volume and improves cognitive functions in MCAO rats. Network pharmacology analysis shows 20 potential active ingredients of SQF could target 13 target proteins. Further employing WGCNA, our study identified four key targets of SQF in the treatment of ischemic stroke. Based on molecular docking and molecular experiments, SQF improves CIRI by activating NFE2L2/NRF2. Serum metabolomics analysis identified six metabolites related to the tryptophan and tyrosine metabolic pathways, which interact with NFE2L2/NRF2 protein. Fecal metabolome and microbiome reveal that SQF's protective effect on CIRI is linked to the tryptophan metabolism and tyrosine metabolism and gut microbiome modulation. In particular, metabolites related to tryptophan and tyrosine metabolism, such as kynurenic acid and dopamine, may exert their protective effects by interacting with NFE2L2/NRF2. CONCLUSION This pioneering study unveils the therapeutic potential of SQF in addressing CIRI, highlighting the pivotal role of NFE2L2/NRF2 upregulation in its mechanism of action. Furthermore, SQF demonstrates its efficacy in restoring gut microbiota balance by modulating the metabolism of tryptophan and tyrosine in CIRI. By elucidating the intricate interplay among constituents, targets, metabolites, and gut microbiota, this research offers novel insights into the multifaceted mechanisms underlying SQF's therapeutic impact on CIRI.
Collapse
Affiliation(s)
- Hai-Xin Liu
- College of Traditional Chinese Medicine and Food Engineering, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Ming-Kuan Yang
- College of Traditional Chinese Medicine and Food Engineering, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Yu-Chang Li
- College of Traditional Chinese Medicine and Food Engineering, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Cai-Xia Liu
- College of Traditional Chinese Medicine and Food Engineering, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Gai-Ping Li
- College of Traditional Chinese Medicine and Food Engineering, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Xiang-Long Meng
- College of Traditional Chinese Medicine and Food Engineering, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Ke Pei
- College of Traditional Chinese Medicine and Food Engineering, Shanxi University of Chinese Medicine, Taiyuan, China.
| | - Shi-Yuan Wen
- College of Basic Medical Sciences, Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
5
|
Zhang X, Wang J, Zhang J, Jiang C, Liu X, Wang S, Zhang Z, Rastegar-Kashkooli Y, Dialameh F, Peng Q, Tao J, Ding R, Wang J, Cheng N, Wang M, Wang F, Li N, Xing N, Chen X, Fan X, Wang J, Wang J. Humanized rodent models of neurodegenerative diseases and other brain disorders. Neurosci Biobehav Rev 2025; 172:106112. [PMID: 40120962 DOI: 10.1016/j.neubiorev.2025.106112] [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: 01/13/2025] [Revised: 02/26/2025] [Accepted: 03/15/2025] [Indexed: 03/25/2025]
Abstract
Central Nervous System (CNS) diseases significantly affect human health. However, replicating the onset, progression, and pathology of these diseases in rodents is challenging. To address this issue, researchers have developed humanized animal models. These models introduce human genes or cells into rodents. As a result, rodents become more suitable for studying human CNS diseases and their therapies in vivo. This review explores the preparation protocols, pathological and behavioral characteristics, benefits, significance, and limitations of humanized rodent models in researching various CNS diseases, particularly Alzheimer's disease, Parkinson's disease, Huntington's disease, Amyotrophic lateral sclerosis, glial cells-related CNS diseases, N-methyl-D-aspartic acid receptor encephalitis, and others. Humanized rodent models have expanded the opportunities for in vivo exploration of human neurodegenerative diseases, other brain disorders, and their treatments. We can enhance translational research on CNS disorders by developing, investigating, and utilizing these models.
Collapse
Affiliation(s)
- Xinru Zhang
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China; Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Jianxiang Wang
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Jiewen Zhang
- Department of Neurology, People's Hospital of Zhengzhou University & Henan Provincial People's Hospital, Zhengzhou, Henan 450000, China
| | - Chao Jiang
- Department of Neurology, People's Hospital of Zhengzhou University & Henan Provincial People's Hospital, Zhengzhou, Henan 450000, China
| | - Xuezhong Liu
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Shuaijiang Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Zhenhua Zhang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Yousef Rastegar-Kashkooli
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; School of International Education, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Fatemeh Dialameh
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; School of International Education, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Qinfeng Peng
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Jin Tao
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Ruoqi Ding
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Junyang Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Nannan Cheng
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Menglu Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Fushun Wang
- Department of Psychology, Sichuan Normal University, Chengdu, Sichuan 610060, China
| | - Nan Li
- Department of Neurology, The 2nd Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450014, China
| | - Na Xing
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Xuemei Chen
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Xiaochong Fan
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| | - Junmin Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Jian Wang
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China; Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| |
Collapse
|
6
|
Wu SN, Wang YJ, Gao ZH, Liutkevičienė R, Rovite V. Recent Advances in Ionic Mechanisms in Pituitary Cells: Implications for Electrophysiological and Electropharmacological Research. J Clin Med 2025; 14:3117. [PMID: 40364147 PMCID: PMC12072979 DOI: 10.3390/jcm14093117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/14/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025] Open
Abstract
Pituitary cells are specialized cells located within the pituitary gland, a small, pea-sized gland situated at the base of the brain. Through the use of cellular electrophysiological techniques, the electrical properties of these cells have been revealed. This review paper aims to introduce the ion currents that are known to be functionally expressed in pituitary cells. These currents include a voltage-gated Na+ current (INa), erg-mediated K+ current (IK(erg)), M-type K+ current (IK(M)), hyperpolarization-activated cation current (Ih), and large-conductance Ca2+-activated K+ (BKCa) channel. The biophysical characteristics of the respective ion current were described. Additionally, we also provide explanations for the effect of various drugs or compounds on each of these currents. GH3-cell exposure to GV-58 can increase the magnitude of INa with a concurrent rise in the inactivation time constant of the current. The presence of esaxerenone, an antagonist of the aldosterone receptor, directly suppresses the magnitude of peak and late INa. Risperidone, an atypical antipsychotic agent, is effective at suppressing the IK(erg) amplitude directly, and di(2-ethylhexyl)-phthalate suppressed IK(erg). Solifenacin and kynurenic acid can interact with the KM channel to stimulate IK(M), while carisbamate and cannabidiol inhibit the Ih amplitude activated by sustained hyperpolarization. Moreover, the presence of either rufinamide or QO-40 can enhance the activity of single BKCa channels. To summarize, alterations in ion currents within native pituitary cells or pituitary tumor cells can influence their functional activity, particularly in processes like stimulus-secretion coupling. The effects of small-molecule modulators, as demonstrated here, bear significance in clinical, therapeutic, and toxicological contexts.
Collapse
Affiliation(s)
- Sheng-Nan Wu
- Department of Research and Education, An-Nan Hospital, China Medical University, No. 66, Section 2, Changhe Road, An Nan District, Tainan 70965, Taiwan
- Institute of Basic Medical Sciences, National Cheng Kung University Medical College, Tainan 701401, Taiwan
| | - Ya-Jean Wang
- Department of Senior Services Industry Management, Minghsin University of Science and Technology, Hsinchu 300401, Taiwan
| | - Zi-Han Gao
- Institute of Basic Medical Sciences, National Cheng Kung University Medical College, Tainan 701401, Taiwan
| | - Rasa Liutkevičienė
- Neuroscience Institute, Medical Academy, Lithuanian University of Health Sciences, Eiveniu 2, 50106 Kaunas, Lithuania
| | - Vita Rovite
- Latvian Biomedical Research and Study Centre (BMC), LV-1067 Riga, Latvia
| |
Collapse
|
7
|
Liu Y, Zhao Z, Xin R, Li D, Dong X, Kushwaha A, Khalid Parvez M, Al-Dosari MS, Kumar A, Huang Y. Electrochemical sensing properties of cobalt-based coordination polymers for trace L-tryptophan in milk. Dalton Trans 2025; 54:6472-6485. [PMID: 40136109 DOI: 10.1039/d5dt00116a] [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: 03/27/2025]
Abstract
Two Co(II)-based coordination polymers (CPs), {[Co3(L)2(H2O)8]·6H2O}n (1) and [Co(L)(H2O)(4,4'-dipy)0.5]n (2) (H3L = 2-(4-carboxyphenyl)-1H-imidazole-4,5-dicarboxylic acid and 4,4'-dipy = 4,4'-bipyridine), were synthesized via a hydrothermal method and characterized using single crystal X-ray diffraction (SC-XRD), powder X-ray diffraction (PXRD), and electrochemical methods. We obtained sensors 1@GCE and 2@GCE by modifying a glassy carbon electrode (GCE), and their electrochemical sensing behavior for L-tryptophan (L-Try) was investigated. The linear range for L-Try sensing was 1-10 μmol, and the limit of detection (LOD) was 0.039 μmol and 0.4 μmol, for 1 and 2, respectively. It was verified by theoretical calculation that their superior electrochemical sensing performance for L-Try was due to the influence of the structure of the CPs. The CP with more active sites and larger void area could interact with L-Try better, and thus, 1 exhibited an excellent sensing effect. In addition, as 1 and 2 successfully detected L-Try in the analysis of milk samples, these materials are expected to serve as high-performance electrochemical sensors for the detection of L-Try.
Collapse
Affiliation(s)
- Yichen Liu
- School of Chemistry and Chemical Engineering, Lanzhou Jiaotong University, Lanzhou 730070, P. R. China.
| | - Zhongqin Zhao
- School of Chemistry and Chemical Engineering, Lanzhou Jiaotong University, Lanzhou 730070, P. R. China.
| | - Rui Xin
- School of Chemistry and Chemical Engineering, Lanzhou Jiaotong University, Lanzhou 730070, P. R. China.
| | - Duqingcuo Li
- School of Chemistry and Chemical Engineering, Lanzhou Jiaotong University, Lanzhou 730070, P. R. China.
| | - Xiuyan Dong
- School of Chemistry and Chemical Engineering, Lanzhou Jiaotong University, Lanzhou 730070, P. R. China.
| | - Aparna Kushwaha
- Department of Chemistry, Faculty of Science, University of Lucknow, Lucknow 226007, India.
| | - Mohammad Khalid Parvez
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammed S Al-Dosari
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Abhinav Kumar
- Department of Chemistry, Faculty of Science, University of Lucknow, Lucknow 226007, India.
| | - Yong Huang
- Dongguan Key Laboratory of Drug Design and Formulation Technology, Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, and School of Pharmacy, Guangdong Medical University, Guangdong Medical University Key Laboratory of Research and Development of New Medical Materials, Dongguan, 523808, China.
| |
Collapse
|
8
|
Wang Y, Zhang Y, Wang W, Zhang Y, Dong X, Liu Y. Diverse Physiological Roles of Kynurenine Pathway Metabolites: Updated Implications for Health and Disease. Metabolites 2025; 15:210. [PMID: 40137174 PMCID: PMC11943880 DOI: 10.3390/metabo15030210] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 03/07/2025] [Accepted: 03/10/2025] [Indexed: 03/27/2025] Open
Abstract
Tryptophan is an essential amino acid critical for human health. It plays a pivotal role in numerous physiological and biochemical processes through its metabolism. The kynurenine (KYN) pathway serves as the principal metabolic route for tryptophan, producing bioactive metabolites, including KYN, quinolinic acid, and 3-hydroxykynurenine. Numerous studies are actively investigating the relationship between tryptophan metabolism and physiological functions. These studies are highlighting the interactions among metabolites that may exert synergistic or antagonistic effects, such as neuroprotective or neurotoxic, and pro-oxidative or antioxidant activities. Minor disruptions in the homeostasis of these metabolites can result in immune dysregulation, contributing to a spectrum of diseases. These diseases include neurological disorders, mental illnesses, cardiovascular conditions, autoimmune diseases, and chronic kidney disease. Therefore, understanding the physiological roles of the KYN pathway metabolites is essential for elucidating the contribution of tryptophan metabolism to health regulation. The present review emphasizes the physiological roles of KYN pathway metabolites and their mechanisms in disease development, aiming to establish a theoretical basis for leveraging dietary nutrients to enhance human health.
Collapse
Affiliation(s)
| | | | | | | | | | - Yang Liu
- Shandong Food Ferment Industry & Design Institute, QiLu University of Technology (Shandong Academy of Sciences), No. 41, Jiefang Road, Jinan 250013, China
| |
Collapse
|
9
|
Rangel MVDS, Lopes KG, Qin X, Borges JP. Exercise-induced adaptations in the kynurenine pathway: implications for health and disease management. Front Sports Act Living 2025; 7:1535152. [PMID: 40115437 PMCID: PMC11922725 DOI: 10.3389/fspor.2025.1535152] [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: 11/26/2024] [Accepted: 02/25/2025] [Indexed: 03/23/2025] Open
Abstract
Background Tryptophan (TRP) metabolism through the kynurenine (KYN) pathway is influenced by inflammatory mediators, generating metabolites that regulate immune and inflammatory responses. Exercise has been proposed as a modulator of this pathway, but its role in health benefits and chronic disease management remains unclear. Objective This systematic review examines exercise-induced adaptations in the KYN pathway and their potential implications for health and disease management. Additionally, we identify key methodological considerations for future research. Methods A structured search of PubMed/Medline, Web of Science, and Scopus was conducted up to October 2024 to identify clinical trials investigating the effects of exercise training on the KYN pathway. Results Of 2,795 articles initially found, 13 clinical trials involving 592 participants met the inclusion criteria. Most studies reported exercise-induced adaptations in the KYN pathway, particularly in cancer survivors. These adaptations appeared to be influenced by exercise intensity and duration. However, several methodological limitations were noted, and no trials included patients with metabolic or cardiovascular diseases. Conclusions Here, we show that exercise training modulates the KYN pathway in both healthy and diseased populations, highlighting its potential for disease prevention and management. However, further randomized-controlled trials are needed to clarify its mechanisms and clinical applications, particularly in metabolic and cardiovascular diseases. Systematic Review Registration https://www.crd.york.ac.uk/PROSPERO/view/CRD42022351481, PROSPERO (CRD42022351481).
Collapse
Affiliation(s)
- Marcus Vinicius Dos Santos Rangel
- Laboratory of Physical Activity and Health Promotion, Institute of Physical Education and Sports, University of Rio de Janeiro State, Rio de Janeiro, RJ, Brazil
| | - Karynne Grutter Lopes
- Postgraduate Program in Clinical and Experimental Physiopathology, Faculty of Medical Sciences, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Xuebin Qin
- Department of Microbiology and Immunology, Tulane National Primate Research Center and Tulane University School of Medicine, Covington, LA, United States
| | - Juliana Pereira Borges
- Laboratory of Physical Activity and Health Promotion, Institute of Physical Education and Sports, University of Rio de Janeiro State, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
10
|
Wang M, Liu Y, Zhong L, Wu F, Wang J. Advancements in the investigation of gut microbiota-based strategies for stroke prevention and treatment. Front Immunol 2025; 16:1533343. [PMID: 40103814 PMCID: PMC11914130 DOI: 10.3389/fimmu.2025.1533343] [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: 11/23/2024] [Accepted: 02/11/2025] [Indexed: 03/20/2025] Open
Abstract
Stroke represents a predominant cause of mortality and disability on a global scale, impacting millions annually and exerting a considerable strain on healthcare systems. The incidence of stroke exhibits regional variability, with ischemic stroke accounting for the majority of occurrences. Post-stroke complications, such as cognitive impairment, motor dysfunction, and recurrent stroke, profoundly affect patients' quality of life. Recent advancements have elucidated the microbiota-gut-brain axis (MGBA), underscoring the complex interplay between gut health and brain function. Dysbiosis, characterized by an imbalance in gut microbiota, is significantly linked to an elevated risk of stroke and unfavorable outcomes. The MGBA plays a crucial role in modulating immune function, neurotransmitter levels, and metabolic byproducts, which may intensify neuroinflammation and impair cerebral health. This review elucidates the role of MGBA in stroke pathophysiology and explores potential gut-targeted therapeutic strategies to reduce stroke risk and promote recovery, including probiotics, prebiotics, pharmacological interventions, and dietary modifications. However, the current prevention and treatment strategies based on intestinal flora still face many problems, such as the large difference of individual intestinal flora, the stability of efficacy, and the long-term safety need to be considered. Further research needs to be strengthened to promote its better application in clinical practice.
Collapse
Affiliation(s)
| | | | | | | | - Jinjin Wang
- Department of Gastroenterology, The First People’s Hospital of Xiaoshan District, Hangzhou, Zhejiang, China
| |
Collapse
|
11
|
Li S, Zhou L, Ren J, Zhang Q, Xiao X. Maternal exercise programs placental miR-495-5p-mediated Snx7 expression and kynurenic acid metabolic pathway induced by prenatal high-fat diet: Based on miRNA-seq, transcriptomics, and metabolomics. J Nutr Biochem 2025; 137:109830. [PMID: 39647668 DOI: 10.1016/j.jnutbio.2024.109830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 11/21/2024] [Accepted: 12/03/2024] [Indexed: 12/10/2024]
Abstract
Poor intrauterine environments increase the prevalence of chronic metabolic diseases in offspring, whereas maternal exercise is an effective measure to break this vicious intergenerational cycle. Placenta is increasingly being studied to explore its role in maternal-fetal metabolic cross-talk. The association between placental miRNA and offspring development trajectories has been established, yet the specific role and mechanism thereof in maternal exercise-induced metabolic protection remain elusive. Here, C57BL/6 female mice were subjected to either a normal control or a high-fat diet (HFD), half of the HFD-fed dams were housed with voluntary wheel running for 3 weeks before and during gestation. At embryonic day 18.5, we sacrificed parturient mice and then conducted miRNA-seq, transcriptomic, and metabolomic profiling of the placenta. Our data revealed that maternal HFD resulted in significant alterations in both miRNA and gene expressions, as well as metabolic pathways of the placenta, whereas prenatal exercise negated these perturbations. The common differentially expressed transcripts among three groups were enriched in multiple critical pathways involving energy expenditure, signal transduction, and fetal development. Through integrated analysis of multiomics data, we speculated that maternal exercise reversed the suppression of miR-495-5p induced by HFD, thereby inhibiting miR-495-5p-targeted Snx7 and modulating kynurenic acid production. These datasets provided novel mechanistic insight into how maternal exercise positively affects the metabolic homeostasis of offspring. The discovered important miRNAs, mRNAs, and metabolites could be promising predictive and therapeutic targets for protecting offspring metabolic health.
Collapse
Affiliation(s)
- Shunhua Li
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Liyuan Zhou
- Department of Endocrinology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Jing Ren
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Qian Zhang
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Xinhua Xiao
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| |
Collapse
|
12
|
Zhang Y, Chen F, Yu M, Li Y, Chen S, Choudhary MI, Liu X, Jiang N. Sex differences in reward-based operant conditioning performance and neurotransmitter changes following chronic sleep restriction stress in rats. BEHAVIORAL AND BRAIN FUNCTIONS : BBF 2025; 21:3. [PMID: 40022197 PMCID: PMC11871611 DOI: 10.1186/s12993-025-00268-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 02/15/2025] [Indexed: 03/03/2025]
Abstract
BACKGROUND Sleep deprivation significantly impairs cognitive function, which disrupts daily life. However, sex differences in these impairments are not well understood, as most preclinical studies primarily use male animals, neglecting potential differences between sexes. This study aims to investigate sex-specific differences in cognitive function under sleep deprivation using reward-based operant conditioning tasks. RESULTS Sprague-Dawley rats were pre-trained on a lever-press task and subsequently divided into control and chronic sleep restriction (CSR) groups. The CSR group underwent 14 days of sleep restriction. After CSR modeling, rats were assessed using the open field test, retraining on the lever-pressing task, signal discrimination task, and extinction task to evaluate motor abilities, memory formation, learning, and cognitive flexibility. CSR significantly impaired task performance in both sexes, with rats requiring more time and exhibiting lower accuracy. In the signal discrimination task, male rats showed longer feeding latency and lower accuracy compared to females. CSR also specifically increased the frequency of operant responses in male rats. In the extinction task, CSR enhanced exploration time and frequency in both sexes, with females exhibiting significantly higher exploration frequencies than males. Biochemically, CSR induced sex-specific alterations, including elevated serum MDA and MAO levels in males and increased serotonin, dopamine, and epinephrine in both sexes. Although activation was observed in metabolites of the tryptophan-kynurenine pathway, sex differences were evident in the kynurenic acid metabolism levels in the prefrontal cortex. CONCLUSIONS CSR impairs cognitive function in both male and female rats, with significant sex differences observed. Male CSR rats exhibited impaired signal discrimination, while CSR impaired extinction learning in female rats. These impairments are accompanied by CSR-induced oxidative stress, neurotransmitter dysregulation, and disturbances in the tryptophan metabolic pathway. These findings underscore the importance of considering sex differences in understanding the effects of sleep deprivation on cognitive function and developing targeted intervention strategies.
Collapse
Affiliation(s)
- Yiwen Zhang
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fang Chen
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mubiao Yu
- Testing Center, SHINVA Medical Instrument Co., Ltd, Shandong, China
| | - Yinghui Li
- Institute of Drug Discovery Technology, Ningbo University, Ningbo, China
| | - Shanguang Chen
- National Laboratory of Human Factors Engineering, State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Muhammad Iqbal Choudhary
- International Center for Chemical and Biological Sciences, H. E. J. Research Institute of Chemistry, University of Karachi, Karachi, Pakistan
| | - Xinmin Liu
- Institute of Drug Discovery Technology, Ningbo University, Ningbo, China.
| | - Ning Jiang
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
13
|
Smadja DM, Abreu MM. Hyperthermia and targeting heat shock proteins: innovative approaches for neurodegenerative disorders and Long COVID. Front Neurosci 2025; 19:1475376. [PMID: 39967803 PMCID: PMC11832498 DOI: 10.3389/fnins.2025.1475376] [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: 08/03/2024] [Accepted: 01/03/2025] [Indexed: 02/20/2025] Open
Abstract
Neurodegenerative diseases (NDs) and Long COVID represent critical and growing global health challenges, characterized by complex pathophysiological mechanisms including neuronal deterioration, protein misfolding, and persistent neuroinflammation. The emergence of innovative therapeutic approaches, such as whole-body hyperthermia (WBH), offers promising potential to modulate underlying pathophysiological mechanisms in NDs and related conditions like Long COVID. WBH, particularly in fever-range, enhances mitochondrial function, induces heat shock proteins (HSPs), and modulates neuroinflammation-benefits that pharmacological treatments often struggle to replicate. HSPs such as HSP70 and HSP90 play pivotal roles in protein folding, aggregation prevention, and cellular protection, directly targeting pathological processes seen in NDs like Alzheimer's, Parkinson's, and Huntington's disease. Preliminary findings also suggest WBH's potential to alleviate neurological symptoms in Long COVID, where persistent neuroinflammation and serotonin dysregulation are prominent. Despite the absence of robust clinical trials, the therapeutic implications of WBH extend to immune modulation and the restoration of disrupted physiological pathways. However, the dual nature of hyperthermia's effects-balancing pro-inflammatory and anti-inflammatory responses-emphasizes the need for dose-controlled applications and stringent patient monitoring to minimize risks in vulnerable populations. While WBH shows potential interest, significant challenges remain. These include individual variability in response, limited accessibility to advanced hyperthermia technologies, and the need for standardized clinical protocols. Future research must focus on targeted clinical trials, biomarker identification, and personalized treatment strategies to optimize WBH's efficacy in NDs and Long COVID. The integration of WBH into therapeutic paradigms could mark a transformative step in addressing these complex conditions.
Collapse
Affiliation(s)
- David M. Smadja
- Paris Cité University, INSERM, Paris Cardiovascular Research Centre, Team Endotheliopathy and Hemostasis Disorders, Paris, France
- Hematology Department, Hôpital Européen Georges Pompidou, Assistance Publique Hôpitaux de Paris-Centre Université Paris Cité (APHP-CUP), Paris, France
| | - M. Marc Abreu
- BTT Medical Institute, Aventura, FL, United States
- BTT Engineering Department, Aventura, FL, United States
| |
Collapse
|
14
|
Guo Z, Zheng Y, Balmer L. Myasthenia Gravis and Depression: A Multifaceted Exploration through Omics and Beyond. ALPHA PSYCHIATRY 2025; 26:38754. [PMID: 40110379 PMCID: PMC11916054 DOI: 10.31083/ap38754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 08/26/2024] [Accepted: 08/28/2024] [Indexed: 03/22/2025]
Affiliation(s)
- Zheng Guo
- Center for Precision Health, Edith Cowan University, 6027 Joondalup, Australia
| | - Yulu Zheng
- Center for Precision Health, Edith Cowan University, 6027 Joondalup, Australia
| | - Lois Balmer
- Center for Precision Health, Edith Cowan University, 6027 Joondalup, Australia
| |
Collapse
|
15
|
Liu L, Jia P, Liu T, Liang J, Dang Y, Rastegar-Kashkooli Y, Li Q, Liu J, Man J, Zhao T, Xing N, Wang F, Chen X, Zhang J, Jiang C, Zille M, Zhang Z, Fan X, Wang J, Wang J. Metabolic dysfunction contributes to mood disorders after traumatic brain injury. Ageing Res Rev 2025; 104:102652. [PMID: 39746403 DOI: 10.1016/j.arr.2024.102652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 12/15/2024] [Accepted: 12/28/2024] [Indexed: 01/04/2025]
Abstract
Traumatic brain injury (TBI) presents significant risks concerning mortality and morbidity. Individuals who suffer from TBI may exhibit mood disorders, including anxiety and depression. Both preclinical and clinical research have established correlations between TBI and disturbances in the metabolism of amino acids, lipids, iron, zinc, and copper, which are implicated in the emergence of mood disorders post-TBI. The purpose of this review is to elucidate the impact of metabolic dysfunction on mood disorders following TBI and to explore potential strategies for mitigating anxiety and depression symptoms. We researched the PubMed and Web of Science databases to delineate the mechanisms by which metabolic dysfunction contributes to mood disorders in the context of TBI. Particular emphasis was placed on the roles of glutamate, kynurenine, lipids, iron, zinc, and copper metabolism. Metabolic dysfunction is linked to mood disorders post-TBI through multiple pathways, encompassing the glutamatergic system, the kynurenine pathway, endocannabinoids, iron deposition, iron-related ferroptosis, zinc deficiency, and copper dysregulation. Furthermore, this review addresses the influence of metabolic dysfunction on mood disorders in the elderly demographic following TBI. Targeting metabolic dysfunction for therapeutic intervention appears promising in alleviating symptoms of anxiety and depression that arise after TBI. While further investigation is warranted to delineate the underlying pathophysiologic mechanisms of mood disorders post-TBI, current evidence underscores the potential contribution of metabolic dysfunction to these conditions. Therefore, rectifying metabolic dysfunction represents a viable and strategic approach to addressing mood disorders following TBI.
Collapse
Affiliation(s)
- Lang Liu
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| | - Peijun Jia
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Tongzhou Liu
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Jiaxin Liang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Yijia Dang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Yousef Rastegar-Kashkooli
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; School of International Education, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Qiang Li
- Department of Neurology, Shanghai Gongli Hospital of Pudong New Area, Shanghai 200135, China.
| | - Jingqi Liu
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Jiang Man
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| | - Ting Zhao
- Department of Neurology, The People's Hospital of Zhengzhou University & Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China.
| | - Na Xing
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| | - Fushun Wang
- Department of Psychology, Sichuan Normal University, Chengdu, Sichuan 610060, China.
| | - Xuemei Chen
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Jiewen Zhang
- Department of Neurology, The People's Hospital of Zhengzhou University & Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China.
| | - Chao Jiang
- Department of Neurology, The People's Hospital of Zhengzhou University & Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China.
| | - Marietta Zille
- Department of Pharmaceutical Sciences, Division of Pharmacology and Toxicology, University of Vienna, Vienna 1090, Austria.
| | - Zhenhua Zhang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Xiaochong Fan
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| | - Junmin Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Jian Wang
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China; Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| |
Collapse
|
16
|
Zhu Q, Yang J, Shi L, Zhang J, Zhang P, Li J, Song X. Exploring the role of ubiquitination modifications in migraine headaches. Front Immunol 2025; 16:1534389. [PMID: 39958329 PMCID: PMC11825825 DOI: 10.3389/fimmu.2025.1534389] [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: 11/25/2024] [Accepted: 01/09/2025] [Indexed: 02/18/2025] Open
Abstract
Migraine is a complex neurovascular disorder whose pathogenesis involves activation of the trigeminal vascular system, central and peripheral sensitization, and neuroinflammation. Calcitonin gene-related peptide (CGRP) plays a dominant role and activation of MAPK and NF-κB signaling pathways regulates neuropeptide release, glial cell activation, and amplification of nociceptive signals. Aberrant activation of these pathways drives migraine onset and chronicity. The ubiquitin-proteasome system (UPS) is involved in neurological and inflammatory disorders. ubiquitination in the UPS is achieved through a cascade of enzymes, including Ub-activating enzyme (E1), Ub-coupling enzyme (E2), and Ub-ligase (E3). The aim of this review is to systematically explore the role of ubiquitination in the regulation of MAPK and NF-κB signaling pathways, with a focus on the mechanisms of ubiquitinating enzymes in neuroinflammation and pain signal amplification, and to explore their potential as diagnostics, biomarkers, predictors of response to therapy, and monitoring of chronicity in migraine disease.
Collapse
Affiliation(s)
- Qian Zhu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Jin Yang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Lei Shi
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Jieying Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Peng Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Junlong Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Xiaoli Song
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| |
Collapse
|
17
|
Szabó Á, Galla Z, Spekker E, Szűcs M, Martos D, Takeda K, Ozaki K, Inoue H, Yamamoto S, Toldi J, Ono E, Vécsei L, Tanaka M. Oxidative and Excitatory Neurotoxic Stresses in CRISPR/Cas9-Induced Kynurenine Aminotransferase Knockout Mice: A Novel Model for Despair-Based Depression and Post-Traumatic Stress Disorder. FRONT BIOSCI-LANDMRK 2025; 30:25706. [PMID: 39862084 DOI: 10.31083/fbl25706] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 10/24/2024] [Accepted: 11/18/2024] [Indexed: 01/27/2025]
Abstract
BACKGROUNDS Memory and emotion are especially vulnerable to psychiatric disorders such as post-traumatic stress disorder (PTSD), which is linked to disruptions in serotonin (5-HT) metabolism. Over 90% of the 5-HT precursor tryptophan (Trp) is metabolized via the Trp-kynurenine (KYN) metabolic pathway, which generates a variety of bioactive molecules. Dysregulation of KYN metabolism, particularly low levels of kynurenic acid (KYNA), appears to be linked to neuropsychiatric disorders. The majority of KYNA is produced by the aadat (kat2) gene-encoded mitochondrial kynurenine aminotransferase (KAT) isotype 2. Little is known about the consequences of deleting the KYN enzyme gene. METHODS In CRISPR/Cas9-induced aadat knockout (kat2-/-) mice, we examined the effects on emotion, memory, motor function, Trp and its metabolite levels, enzyme activities in the plasma and urine of 8-week-old males compared to wild-type mice. RESULTS Transgenic mice showed more depressive-like behaviors in the forced swim test, but not in the tail suspension, anxiety, or memory tests. They also had fewer center field and corner entries, shorter walking distances, and fewer jumping counts in the open field test. Plasma metabolite levels are generally consistent with those of urine: antioxidant KYNs, 5-hydroxyindoleacetic acid, and indole-3-acetic acid levels were lower; enzyme activities in KATs, kynureninase, and monoamine oxidase/aldehyde dehydrogenase were lower, but kynurenine 3-monooxygenase was higher; and oxidative stress and excitotoxicity indices were higher. Transgenic mice displayed depression-like behavior in a learned helplessness model, emotional indifference, and motor deficits, coupled with a decrease in KYNA, a shift of Trp metabolism toward the KYN-3-hydroxykynurenine pathway, and a partial decrease in the gut microbial Trp-indole pathway metabolite. CONCLUSIONS This is the first evidence that deleting the aadat gene induces depression-like behaviors uniquely linked to experiences of despair, which appear to be associated with excitatory neurotoxic and oxidative stresses. This may lead to the development of a double-hit preclinical model in despair-based depression, a better understanding of these complex conditions, and more effective therapeutic strategies by elucidating the relationship between Trp metabolism and PTSD pathogenesis.
Collapse
Affiliation(s)
- Ágnes Szabó
- Department of Neurology, Albert Szent-Györgyi Medical School, University of Szeged, H-6725 Szeged, Hungary
- Doctoral School of Clinical Medicine, University of Szeged, H-6720 Szeged, Hungary
| | - Zsolt Galla
- Department of Pediatrics, Albert Szent-Györgyi Faculty of Medicine, University of Szeged, H-6725 Szeged, Hungary
| | - Eleonóra Spekker
- HUN-REN-SZTE Neuroscience Research Group, Hungarian Research Network, University of Szeged (HUN-REN-SZTE), Danube Neuroscience Research Laboratory, H-6725 Szeged, Hungary
| | - Mónika Szűcs
- Department of Medical Physics and Informatics, Albert Szent-Györgyi Medical School, Faculty of Science and Informatics, University of Szeged, H-6720 Szeged, Hungary
| | - Diána Martos
- HUN-REN-SZTE Neuroscience Research Group, Hungarian Research Network, University of Szeged (HUN-REN-SZTE), Danube Neuroscience Research Laboratory, H-6725 Szeged, Hungary
| | - Keiko Takeda
- Department of Biomedicine, Graduate School of Medical Sciences, Kyushu University, 812-8582 Fukuoka, Japan
| | - Kinuyo Ozaki
- Center of Biomedical Research, Research Center for Human Disease Modeling, Graduate School of Medical Sciences, Kyushu University, 812-8582 Fukuoka, Japan
| | - Hiromi Inoue
- Center of Biomedical Research, Research Center for Human Disease Modeling, Graduate School of Medical Sciences, Kyushu University, 812-8582 Fukuoka, Japan
| | - Sayo Yamamoto
- Center of Biomedical Research, Research Center for Human Disease Modeling, Graduate School of Medical Sciences, Kyushu University, 812-8582 Fukuoka, Japan
| | - József Toldi
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, H-6726 Szeged, Hungary
| | - Etsuro Ono
- Department of Biomedicine, Graduate School of Medical Sciences, Kyushu University, 812-8582 Fukuoka, Japan
- Center of Biomedical Research, Research Center for Human Disease Modeling, Graduate School of Medical Sciences, Kyushu University, 812-8582 Fukuoka, Japan
| | - László Vécsei
- Department of Neurology, Albert Szent-Györgyi Medical School, University of Szeged, H-6725 Szeged, Hungary
- HUN-REN-SZTE Neuroscience Research Group, Hungarian Research Network, University of Szeged (HUN-REN-SZTE), Danube Neuroscience Research Laboratory, H-6725 Szeged, Hungary
| | - Masaru Tanaka
- HUN-REN-SZTE Neuroscience Research Group, Hungarian Research Network, University of Szeged (HUN-REN-SZTE), Danube Neuroscience Research Laboratory, H-6725 Szeged, Hungary
| |
Collapse
|
18
|
Li M, Wu Y, Xu Y, Huang X, Gao K, Hu N, Zhu S, Wang C, Liang S. Peripheral tryptophan-kynurenine pathway dysfunction in first-episode schizophrenia. Sci Rep 2025; 15:2432. [PMID: 39827210 PMCID: PMC11742721 DOI: 10.1038/s41598-025-86390-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 01/10/2025] [Indexed: 01/22/2025] Open
Abstract
The tryptophan (TRP)-kynurenine (KYN) pathway is involved in the pathogenesis of schizophrenia. This study aimed to investigate the levels of TRP-KYN metabolites in serum and urine of patients with first-episode schizophrenia (FES) and their association with clinical manifestations. This study included 38 drug-naive patients with FES and 43 healthy controls (HCs). Clinical symptoms were evaluated using the Positive and Negative Syndrome Scale (PANSS). Levels of TRP-KYN metabolites in serum and urine were quantified. Patients with FES showed significantly higher serum quinolinic acid/kynurenic acid (QUIN/KYNA) ratio and urine KYN/TRP ratio compared to HCs, while neuroprotective metabolites, including serum KYNA, xanthurenic acid (XA), and urine picolinic acid (PIC) levels, were significantly reduced, along with a decreased urine PIC/QUIN ratio (p < 0.05). The urine KYNA/KYN ratio was negatively correlated with PANSS general psychopathology scores (r = -0.35, p = 0.04) and with PANSS total scores (r = -0.35, p = 0.046). Patients with FES exhibited dysregulation of the peripheral TRP-KYN pathway, characterized by an increased neurotoxic-to-neuroprotective QUIN/KYNA ratio and reduced levels of neuroprotective metabolites. This shift towards increased neurotoxic product generation suggests that the dysregulation of the TRP-KYN pathway could play a role in the pathophysiology of schizophrenia.
Collapse
Affiliation(s)
- Mian Li
- Affiliated Mental Health Centre & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310013, China
- The Fourth Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yue Wu
- Affiliated Mental Health Centre & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310013, China
| | - Yan Xu
- Affiliated Mental Health Centre & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310013, China
| | - Xin Huang
- Affiliated Mental Health Centre & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310013, China
| | - Kerun Gao
- Affiliated Mental Health Centre & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310013, China
| | - Nannan Hu
- Affiliated Mental Health Centre & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310013, China
| | - Shuangyue Zhu
- Affiliated Mental Health Centre & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310013, China
| | - Chengpeng Wang
- Affiliated Mental Health Centre & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310013, China
| | - Sugai Liang
- Affiliated Mental Health Centre & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310013, China.
| |
Collapse
|
19
|
Wang R, Gan C, Mao R, Chen Y, Yan R, Li G, Xiong T, Guo J. Rat models of postintracerebral hemorrhage pneumonia induced by nasal inoculation with Klebsiella pneumoniae or intratracheal inoculation with LPS. Front Immunol 2025; 15:1477902. [PMID: 39845950 PMCID: PMC11750689 DOI: 10.3389/fimmu.2024.1477902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 12/16/2024] [Indexed: 01/24/2025] Open
Abstract
Background A stable and reproducible experimental bacterial pneumonia model postintracerebral hemorrhage (ICH) is necessary to help investigating the pathogenesis and novel treatments of Stroke-associated pneumonia (SAP). Aim To establish a Gram-negative bacterial pneumonia-complicating ICH rat model and an acute lung injury (ALI)-complicating ICH rat model. Methods We established two standardized models of post-ICH pneumonia by nasal inoculation with Klebsiella pneumoniae (Kp) or intratracheal inoculation with lipopolysaccharide (LPS). Survival and neurological scores were monitored. Magnetic resonance imaging was performed to evaluate hematoma volume. Abdominal aortic blood was collected for leukocyte counting, serum was isolated to determine concentrations of S100β and proinflammatory cytokines using ELISAs. Histopathological changes of brain, lung and gut were assessed using hematoxylin-eosin staining. Lung was isolated for immunofluorescence staining for myeloperoxidase (MPO). Bronchoalveolar lavage fluid was collected for leukocyte counting, and supernatant was prepared to measure MPO activity. Ileum was isolated for immunofluorescence staining for tight junction proteins ZO-1 and γδ TCRs/IL-17A and for Alcian blue-nuclear fast red staining of acidic mucins. Feces were collected, 16S rRNA sequencing, untargeted metabolomics and Spearman's correlation analyses were performed to explore changes of gut microbiota, metabolites and their interactions. Results In Kp-induced bacterial pneumonia-complicating ICH rats, we demonstrated that Kp challenge caused more severe neurological deficits, brain damage, neuroinflammation, and aggravated pneumonia and lung injury. Disruptions of the intestinal structure and gut barrier and the reductions of the protective intestinal IL-17A-producing γδT cells were also observed. Kp challenge exacerbated the gut microbiota dysbiosis and fecal metabolic profile disorders, which were characterized by abnormal sphingolipid metabolism especially elevated ceramide levels; increased levels of neurotoxic quinolinic acid and an upregulation of tryptophan (Trp)-serotonin-melatonin pathway. Spearman's correlation analyses further revealed that the reduction or depletion of some beneficial bacteria, such as Allobaculum and Faecalitalea, and the blooming of some opportunistic pathogens, such as Turicibacter, Dietzia, Corynebacterium and Clostridium_sensu_stricto_1 in Kp-induced SAP rats were associated with the disordered sphingolipid and Trp metabolism. Using an LPS-induced ALI complicating ICH model, we also characterized SAP-induced brain, lung and gut histopathology injuries; peripheral immune disorders and intense pulmonary inflammatory responses. Conclusions These two models may be highly useful for investigating the pathogenesis and screening and optimizing potential treatments for SAP. Moreover, the differential genera and sphingolipid or Trp metabolites identified above seem to be promising therapeutic targets.
Collapse
Affiliation(s)
- Ruihua Wang
- Research Team of Prevention and Treatment of Cerebral Hemorrhage Applying Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Changlian Gan
- School of Traditional Dai Medicine, West Yunnan University of Applied Science, Xishuangbanna, China
| | - Rui Mao
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yang Chen
- Department of Bioinformatics, State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ru Yan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, Macao SAR, China
| | - Geng Li
- Laboratory Animal Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Tianqin Xiong
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jianwen Guo
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Department of Neurology, Guangdong Provincial Academy of Chinese Medical Sciences, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
20
|
Li XY, Yu WK, Wu JH, He WJ, Cheng YN, Gao K, Wei YH, Li YS. Tryptophan metabolism-related gene CYP1B1 serves as a shared biomarker for both Parkinson's disease and insomnia. Sci Rep 2025; 15:1362. [PMID: 39779759 PMCID: PMC11711247 DOI: 10.1038/s41598-024-84362-8] [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: 07/03/2024] [Accepted: 12/23/2024] [Indexed: 01/11/2025] Open
Abstract
Parkinson's disease (PD) and insomnia are prevalent neurological disorders, with emerging evidence implicating tryptophan (TRP) metabolism in their pathogenesis. However, the precise mechanisms by which TRP metabolism contributes to these conditions remain insufficiently elucidated. This study explores shared tryptophan metabolism-related genes (TMRGs) and molecular mechanisms underlying PD and insomnia, aiming to provide insights into their shared pathogenesis. We analyzed datasets for PD (GSE100054) and insomnia (GSE208668) obtained from the Gene Expression Omnibus (GEO) database. TMRGs were obtained from the Molecular Signatures Database (MSigDB) and the Genecards database. Tryptophan metabolism-related differentially expressed genes (TM-DEGs) were identified by intersecting TMRGs with shared differentially expressed genes (DEGs) from these datasets. Through Protein-Protein Interaction (PPI) network analysis, Support Vector Machine-Recursive Feature Elimination (SVM-RFE) , and Extreme Gradient Boosting (XGBoost) machine learning, we identified Cytochrome P4501B1 (CYP1B1) and Electron Transfer Flavoprotein Alpha (ETFA) as key hub genes. Subsequently, we employed CIBERSORT and single-sample gene set enrichment analysis (ssGSEA) to further investigate the association between hub genes and peripheral immune activation and inflammatory response. Additionally, gene interaction, Drug-mRNA, Transcription Factor (TF)-mRNA, and competing endogenous RNA (ceRNA) networks centered on these hub genes were constructed to explore regulatory mechanisms and potential drug interactions. Finally, validation through bioinformatics and animal experiments identified CYP1B1 as a promising biomarker associated with both PD and insomnia.
Collapse
Affiliation(s)
- Xin-Yu Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe Dong Road, Zhengzhou, Henan, China
- Henan Engineering Research Center of Neural Function Detection and Regulation, Zhengzhou, Henan, China
- Henan Key Laboratory of Cerebrovascular Diseases (Zhengzhou University), Zhengzhou, China
| | - Wen-Kai Yu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe Dong Road, Zhengzhou, Henan, China
- Henan Engineering Research Center of Neural Function Detection and Regulation, Zhengzhou, Henan, China
- Henan Key Laboratory of Cerebrovascular Diseases (Zhengzhou University), Zhengzhou, China
| | - Jing-Hao Wu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe Dong Road, Zhengzhou, Henan, China
- Henan Engineering Research Center of Neural Function Detection and Regulation, Zhengzhou, Henan, China
- Henan Key Laboratory of Cerebrovascular Diseases (Zhengzhou University), Zhengzhou, China
| | - Wen-Jun He
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe Dong Road, Zhengzhou, Henan, China
- Henan Engineering Research Center of Neural Function Detection and Regulation, Zhengzhou, Henan, China
- Henan Key Laboratory of Cerebrovascular Diseases (Zhengzhou University), Zhengzhou, China
| | - Yu-Nan Cheng
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe Dong Road, Zhengzhou, Henan, China
- Henan Engineering Research Center of Neural Function Detection and Regulation, Zhengzhou, Henan, China
- Henan Key Laboratory of Cerebrovascular Diseases (Zhengzhou University), Zhengzhou, China
| | - Kai Gao
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe Dong Road, Zhengzhou, Henan, China
- Henan Engineering Research Center of Neural Function Detection and Regulation, Zhengzhou, Henan, China
- Henan Key Laboratory of Cerebrovascular Diseases (Zhengzhou University), Zhengzhou, China
| | - Yi-Han Wei
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe Dong Road, Zhengzhou, Henan, China
- Henan Engineering Research Center of Neural Function Detection and Regulation, Zhengzhou, Henan, China
- Henan Key Laboratory of Cerebrovascular Diseases (Zhengzhou University), Zhengzhou, China
| | - Yu-Sheng Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe Dong Road, Zhengzhou, Henan, China.
- Henan Engineering Research Center of Neural Function Detection and Regulation, Zhengzhou, Henan, China.
- Henan Key Laboratory of Cerebrovascular Diseases (Zhengzhou University), Zhengzhou, China.
| |
Collapse
|
21
|
Shao X, Li M, Shi S, Wu T, Zhang Y, Guo S, Lin H, Qi X. Neuroprotective Effects of Paeonia lactiflora Through the Regulation of Gut Dubosiella in an MPTP-Induced Parkinson's Disease Mouse Model. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2025; 53:833-862. [PMID: 40374370 DOI: 10.1142/s0192415x25500314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2025]
Abstract
Emerging evidence suggests that changes in the composition of the gut microbiota may play an important role in the pathogenesis of Parkinson's disease (PD). Paeonia lactiflora Pall., a traditional Chinese medicinal herb belonging to the genus Paeonia, is commonly used in Chinese medicinal practice for the treatment of PD. However, the specific mechanisms of its action remain poorly understood. This study aimed to further determine the neuroprotective properties of Paeonia lactiflora Pall. water extract (PWE) in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD model, and to investigate its potential implications for the pathogenesis of PD. A PD mouse model was established via the intraperitoneal administration of MPTP, followed by the assessment of motor function using behavioral tests. Western blotting and histopathological analysis were used to measure the levels of dopaminergic (DAergic) neurodegeneration-related factors in the midbrain (containing the substantia nigra (SN)) and striatum. 16S rRNA gene sequencing and metabolomic analysis were applied to identify differences in the gut microbiota and metabolites, respectively. Our results indicate that PWE effectively protects against MPTP-induced motor deficits, loss of DAergic neurons, blood-brain barrier (BBB) damage, and neuroinflammation in PD. The protective effects of PWE against PD are mediated through modulation of the gut microbiota, specifically by an increase in the abundance of the genus Dubosiella. In this study, we selected D. newyorkensis as a representative strain of the genus, and determined its therapeutic effects in an MPTP-induced PD mouse model. Our preliminary findings suggest that the neuroprotective effects of D. newyorkensis may be related to the production of serum indoleacetic acid.
Collapse
Affiliation(s)
- Xian Shao
- Department of Medical Research Center, Shaoxing People's Hospital, Zhejiang University Shaoxing Hospital, Shaoxing, Zhejiang 312000, China
| | - Mengyun Li
- Department of Medical Research Center, Shaoxing People's Hospital, Zhejiang University Shaoxing Hospital, Shaoxing, Zhejiang 312000, China
| | - Shuai Shi
- Department of Neurosurgery, Shaoxing People's Hospital, Zhejiang University Shaoxing Hospital, Shaoxing, Zhejiang 312000, China
| | - Tao Wu
- Department of Neurosurgery, Shaoxing People's Hospital, Zhejiang University Shaoxing Hospital, Shaoxing, Zhejiang 312000, China
| | - Yanxing Zhang
- Department of Neurology, Shaoxing People's Hospital, Zhejiang University Shaoxing Hospital, Shaoxing, Zhejiang 312000, China
| | - Shitian Guo
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Hui Lin
- The Affiliated Lihuili Hospital of Ningbo University, Healthy Science Center, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Xuchen Qi
- Department of Neurosurgery, Shaoxing People's Hospital, Zhejiang University Shaoxing Hospital, Shaoxing, Zhejiang 312000, China
- Department of Neurosurgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310020, China
| |
Collapse
|
22
|
Wang X, Wu L, Liu J, Ma C, Liu J, Zhang Q. The neuroimmune mechanism of pain induced depression in psoriatic arthritis and future directions. Biomed Pharmacother 2025; 182:117802. [PMID: 39742638 DOI: 10.1016/j.biopha.2024.117802] [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: 10/31/2024] [Revised: 12/16/2024] [Accepted: 12/27/2024] [Indexed: 01/03/2025] Open
Abstract
Patients suffering from psoriatic arthritis (PsA) often experience depression due to chronic joint pain, which significantly hinders their recovery process. However, the relationship between these two conditions is not well understood. Through a review of existing studies, we revealed that certain neuroendocrine hormones and neurotransmitters are involved in the neuroimmune interactions related to both PsA and depression. These include adrenocorticotropin-releasing hormone (CRH), adrenocorticotropin (ACTH), cortisol, monoamine neurotransmitters, and brain-derived neurotrophic factor (BDNF). Notably, the signalling pathway involving CRH, MCs, and Th17 cells plays a crucial role in linking PsA with depression; thus, this pathway may help clarify their connection. In this review, we outline the inflammatory immune changes associated with PsA and depression. Additionally, we explore how neuroendocrine hormones and neurotransmitters influence inflammatory responses in these two conditions. Finally, our focus will be on potential treatment strategies for patients with PsA and depression through the targeting of the CRH-MC-Th17 pathway. This review aims to provide a theoretical framework as well as new therapeutic targets for managing PsA alongside depression.
Collapse
Affiliation(s)
- Xiaoxu Wang
- Rheumatology Department, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China.
| | - Lingjun Wu
- Shunyi Hospital of Beijing Traditional Chinese Medicine Hospital, Beijing 101300, China
| | - Jing Liu
- Department of Oncology and Hematology, Dongzhimen Hospital, Beijing University of Chinese Medicine (BUCM), Beijing 100010, China
| | - Cong Ma
- Rheumatology Department, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
| | - Juan Liu
- Rheumatology Department, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
| | - Qin Zhang
- Rheumatology Department, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China.
| |
Collapse
|
23
|
Wang C, Tang Y, Yang T, Wang Y, Niu Z, Zhang K, Lin N, Li Q. Causal Relationship Between Intestinal Microbiota, Inflammatory Cytokines, Peripheral Immune Cells, Plasma Metabolome and Parkinson's Disease: A Mediation Mendelian Randomization Study. Eur J Neurosci 2025; 61:e16665. [PMID: 39831637 DOI: 10.1111/ejn.16665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 12/19/2024] [Accepted: 12/29/2024] [Indexed: 01/22/2025]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease involving multiple factors. We explored the connection between intestinal microbiome levels and PD by examining inflammatory cytokines, peripheral immune cell counts and plasma metabolomics as potential factors. By obtaining the Genome-Wide Association Study (GWAS) data needed for this study from GWAS Catalog, including summary data for 473 intestinal microbiota traits (N = 5959), 91 inflammatory cytokine traits (N = 14,824), 118 peripheral immune cell count traits (N = 3757), 1400 plasma metabolite traits (N = 8299) and PD traits (N = 482,730). We used two-step Mendelian randomization (MR) mediated analysis to investigate possible pathways from intestinal microbiota to PD mediated by inflammatory cytokines, peripheral immune cells and plasma metabolites. MR has revealed the causal effects of 19 intestinal microbiota, 1 inflammatory cytokine and 12 plasma metabolites on PD, whereas there is no significant causal relationship between immune cell count characteristics and the occurrence of PD. Mediation analysis showed that the associations between the genus Demequina and PD were mediated by tryptophan with mediated proportions of 17.51% (p = 0.0393). Our study demonstrates that genus Demequina may promote the occurrence of PD by reducing the levels of tryptophan.
Collapse
Affiliation(s)
- Chengcheng Wang
- Department of Neurosurgery, Affiliated Chuzhou Hospital of Anhui Medical University, First People's Hospital of Chuzhou, Chuzhou, China
| | - Yuhang Tang
- Department of Neurosurgery, Affiliated Chuzhou Hospital of Anhui Medical University, First People's Hospital of Chuzhou, Chuzhou, China
| | - Tao Yang
- Department of Neurosurgery, Affiliated Chuzhou Hospital of Anhui Medical University, First People's Hospital of Chuzhou, Chuzhou, China
| | - Yuhao Wang
- Department of Neurosurgery, Affiliated Chuzhou Hospital of Anhui Medical University, First People's Hospital of Chuzhou, Chuzhou, China
| | - Zihui Niu
- Department of Neurosurgery, Affiliated Chuzhou Hospital of Anhui Medical University, First People's Hospital of Chuzhou, Chuzhou, China
| | - Kang Zhang
- Department of Neurosurgery, Affiliated Chuzhou Hospital of Anhui Medical University, First People's Hospital of Chuzhou, Chuzhou, China
| | - Ning Lin
- Department of Neurosurgery, Affiliated Chuzhou Hospital of Anhui Medical University, First People's Hospital of Chuzhou, Chuzhou, China
| | - Qun Li
- Health Examination Center, Affiliated Chuzhou Hospital of Anhui Medical University, First People's Hospital of Chuzhou, Chuzhou, China
| |
Collapse
|
24
|
Li J, Hao G, Yan Y, Li M, Li G, Lu Z, Sun Z, Chen Y, Liu H, Zhao Y, Wu M, Bao X, Wang Y, Li Y. Hydrogen restores central tryptophan and metabolite levels and maintains mitochondrial homeostasis to protect rats from chronic mild unpredictable stress damage. Neurochem Int 2025; 182:105914. [PMID: 39653185 DOI: 10.1016/j.neuint.2024.105914] [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: 10/12/2024] [Revised: 11/25/2024] [Accepted: 11/28/2024] [Indexed: 12/12/2024]
Abstract
BACKGROUND AND PURPOSE The field of hydrogen medicine has garnered extensive attention since Professor Ohsawa established that low concentrations of hydrogen (2%-4%) exert antioxidant effects. The present study aimed to evaluate the therapeutic effect of molecular hydrogen in a CUMS rat model. METHODS A total of 40 SD rats were randomly divided into a control group, a model group, a hydrogen group, and a positive drug group. Four weeks post-modeling, hydrogen inhalation and other treatments were administered. Behavioral, biochemical, and immunohistochemical evaluations were performed after treatment. RESULTS Hydrogen inhalation alleviated depressive behavior and hippocampal neuronal damage in CUMS rats, as well as restored the levels of neurotransmitters, inflammatory factors, and oxidative stress. Moreover, it maintained mitochondrial homeostasis and up-regulated the expression of PGC-1α, PINK1, and Parkin. CONCLUSIONS The results collectively indicated that hydrogen significantly attenuated CUMS-induced depressive-like behavior and monoamine neurotransmitter deficiency, as well as protected the brain from oxidative stress and inflammatory damage and effectively preserved mitochondrial homeostasis.
Collapse
Affiliation(s)
- Jiaxin Li
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Gaimei Hao
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yupeng Yan
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Ming Li
- Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Gaifen Li
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Zhengmin Lu
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Zhibo Sun
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yanjing Chen
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Haixia Liu
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yukun Zhao
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Meng Wu
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Xiangxin Bao
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yong Wang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Yubo Li
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| |
Collapse
|
25
|
Xing C, Chen H, Bi W, Lei T, Hang Z, Du H. Targeting 5-HT Is a Potential Therapeutic Strategy for Neurodegenerative Diseases. Int J Mol Sci 2024; 25:13446. [PMID: 39769209 PMCID: PMC11679250 DOI: 10.3390/ijms252413446] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/07/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
There is increasing interest in the potential therapeutic role of 5-HT (serotonin) in the treatment of neurodegenerative diseases, which are characterized by the progressive degeneration and death of nerve cells. 5-HT is a vital neurotransmitter that plays a central role in regulating mood, cognition, and various physiological processes in the body. Disruptions in the 5-HT system have been linked to several neurological and psychiatric disorders, making it an attractive target for therapeutic intervention. Although the exact causes of neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS) are not fully understood, researchers believe that regulating the 5-HT system could help alleviate symptoms and potentially slow the progression of these diseases. Here, we delve into the potential of harnessing 5-HT as a therapeutic target for the treatment of neurodegenerative diseases. It is important to note that the current clinical drugs targeting 5-HT are still limited in the treatment of these complex diseases. Therefore, further research and clinical trials are needed to evaluate the feasibility and effectiveness of its clinical application.
Collapse
Affiliation(s)
- Cencan Xing
- Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083, China; (C.X.); (H.C.); (W.B.); (Z.H.)
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China;
| | - Hongyu Chen
- Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083, China; (C.X.); (H.C.); (W.B.); (Z.H.)
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China;
| | - Wangyu Bi
- Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083, China; (C.X.); (H.C.); (W.B.); (Z.H.)
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China;
| | - Tong Lei
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China;
| | - Zhongci Hang
- Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083, China; (C.X.); (H.C.); (W.B.); (Z.H.)
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China;
| | - Hongwu Du
- Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083, China; (C.X.); (H.C.); (W.B.); (Z.H.)
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China;
| |
Collapse
|
26
|
Zeng J, Wang C, Guo J, Zhao T, Wang H, Zhang R, Pu L, Yang H, Liang J, Han L, Li L. Multiomics Profiling of Plasma Reveals Molecular Alterations Prior to a Diagnosis with Stroke Among Chinese Hypertension Patients. J Proteome Res 2024; 23:5421-5437. [PMID: 39466185 DOI: 10.1021/acs.jproteome.4c00559] [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] [Indexed: 10/29/2024]
Abstract
We aimed to investigate the correlation between plasma proteins and metabolites and the occurrence of future strokes using mass spectrometry and bioinformatics as well as to identify other biomarkers that could predict stroke risk in hypertensive patients. In a nested case-control study, baseline plasma samples were collected from 50 hypertensive subjects who developed stroke and 50 gender-, age- and body mass index-matched controls. Plasma untargeted metabolomics and data independent acquisition-based proteomics analysis were performed in hypertensive patients, and 19 metabolites and 111 proteins were found to be differentially expressed. Integrative analyses revealed that molecular changes in plasma indicated dysregulation of protein digestion and absorption, salivary secretion, and regulation of actin cytoskeleton, along with significant metabolic suppression. C4BPA, Caprolactam, Col15A1, and HBB were identified as predictors of stroke occurrence, and the Support Vector Machines (SVM) model was determined to be the optimal predictive model by integrating six machine-learning classification models. The SVM model showed strong performance in both the internal validation set (area under the curve [AUC]: 0.977, 95% confidence interval [CI]: 0.941-1.000) and the external independent validation set (AUC: 0.973, 95% CI: 0.921-0.999).
Collapse
Affiliation(s)
- Jingjing Zeng
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Key Laboratory of Panvascular Diseases of Wenzhou, Wenzhou 325000, China
- Center for Cardiovascular and Cerebrovascular Epidemiology and Translational Medicine, Guoke Ningbo Life Science and Health Industry Research Institute, University of Chinese Academy of Sciences, Ningbo 315000, China
| | - Changyi Wang
- Department of Non-Communicable Disease Prevention and Control, Shenzhen Nanshan Center for Chronic Disease Control, Shenzhen 518000, China
| | - Jiamin Guo
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Key Laboratory of Panvascular Diseases of Wenzhou, Wenzhou 325000, China
| | - Tian Zhao
- Center for Cardiovascular and Cerebrovascular Epidemiology and Translational Medicine, Guoke Ningbo Life Science and Health Industry Research Institute, University of Chinese Academy of Sciences, Ningbo 315000, China
- Department of Clinical Epidemiology, Ningbo 2 Hospital, Ningbo 315000, China
| | - Han Wang
- Center for Cardiovascular and Cerebrovascular Epidemiology and Translational Medicine, Guoke Ningbo Life Science and Health Industry Research Institute, University of Chinese Academy of Sciences, Ningbo 315000, China
- Department of Clinical Epidemiology, Ningbo 2 Hospital, Ningbo 315000, China
| | - Ruijie Zhang
- Center for Cardiovascular and Cerebrovascular Epidemiology and Translational Medicine, Guoke Ningbo Life Science and Health Industry Research Institute, University of Chinese Academy of Sciences, Ningbo 315000, China
- Department of Clinical Epidemiology, Ningbo 2 Hospital, Ningbo 315000, China
| | - Liyuan Pu
- Center for Cardiovascular and Cerebrovascular Epidemiology and Translational Medicine, Guoke Ningbo Life Science and Health Industry Research Institute, University of Chinese Academy of Sciences, Ningbo 315000, China
- Department of Clinical Epidemiology, Ningbo 2 Hospital, Ningbo 315000, China
| | - Huiqun Yang
- Center for Cardiovascular and Cerebrovascular Epidemiology and Translational Medicine, Guoke Ningbo Life Science and Health Industry Research Institute, University of Chinese Academy of Sciences, Ningbo 315000, China
- Department of Clinical Epidemiology, Ningbo 2 Hospital, Ningbo 315000, China
| | - Jie Liang
- Center for Cardiovascular and Cerebrovascular Epidemiology and Translational Medicine, Guoke Ningbo Life Science and Health Industry Research Institute, University of Chinese Academy of Sciences, Ningbo 315000, China
- Department of Clinical Epidemiology, Ningbo 2 Hospital, Ningbo 315000, China
| | - Liyuan Han
- Center for Cardiovascular and Cerebrovascular Epidemiology and Translational Medicine, Guoke Ningbo Life Science and Health Industry Research Institute, University of Chinese Academy of Sciences, Ningbo 315000, China
- Department of Clinical Epidemiology, Ningbo 2 Hospital, Ningbo 315000, China
| | - Lei Li
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Key Laboratory of Panvascular Diseases of Wenzhou, Wenzhou 325000, China
| |
Collapse
|
27
|
Tanaka M, Vécsei L. Revolutionizing our understanding of Parkinson's disease: Dr. Heinz Reichmann's pioneering research and future research direction. J Neural Transm (Vienna) 2024; 131:1367-1387. [PMID: 39110245 PMCID: PMC11608389 DOI: 10.1007/s00702-024-02812-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 07/22/2024] [Indexed: 11/17/2024]
Abstract
Millions of individuals around the world are afflicted with Parkinson's disease (PD), a prevalent and incapacitating neurodegenerative disorder. Dr. Reichmann, a distinguished professor and neurologist, has made substantial advancements in the domain of PD research, encompassing both fundamental scientific investigations and practical applications. His research has illuminated the etiology and treatment of PD, as well as the function of energy metabolism and premotor symptoms. As a precursor to a number of neurotransmitters and neuromodulators that are implicated in the pathophysiology of PD, he has also investigated the application of tryptophan (Trp) derivatives in the disease. His principal findings and insights are summarized and synthesized in this narrative review article, which also emphasizes the challenges and implications for future PD research. This narrative review aims to identify and analyze the key contributions of Reichmann to the field of PD research, with the ultimate goal of informing future research directions in the domain. By examining Reichmann's work, the study seeks to provide a comprehensive understanding of his major contributions and how they can be applied to advance the diagnosis and treatment of PD. This paper also explores the potential intersection of Reichmann's findings with emerging avenues, such as the investigation of Trp and its metabolites, particularly kynurenines, which could lead to new insights and potential therapeutic strategies for managing neurodegenerative disorders like PD.
Collapse
Affiliation(s)
- Masaru Tanaka
- HUN-REN-SZTE Neuroscience Research Group, Hungarian Research Network, University of Szeged (HUN-REN-SZTE), Danube Neuroscience Research Laboratory, Tisza Lajos krt. 113, Szeged, H-6725, Hungary.
| | - László Vécsei
- HUN-REN-SZTE Neuroscience Research Group, Hungarian Research Network, University of Szeged (HUN-REN-SZTE), Danube Neuroscience Research Laboratory, Tisza Lajos krt. 113, Szeged, H-6725, Hungary
- Department of Neurology, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6, Szeged, H-6725, Hungary
| |
Collapse
|
28
|
Christensen J, Vlassopoulos E, Barlow CK, Schittenhelm RB, Li CN, Sgro M, Warren S, Semple BD, Yamakawa GR, Shultz SR, Mychasiuk R. The beneficial effects of modafinil administration on repeat mild traumatic brain injury (RmTBI) pathology in adolescent male rats are not dependent upon the orexinergic system. Exp Neurol 2024; 382:114969. [PMID: 39332798 DOI: 10.1016/j.expneurol.2024.114969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 08/22/2024] [Accepted: 09/21/2024] [Indexed: 09/29/2024]
Abstract
The sleep-wake cycle plays an influential role in the development and progression of repeat mild traumatic brain injury (RmTBI)-related pathology. Therefore, we first aimed to manipulate the sleep-wake cycle post-RmTBI using modafinil, a wake-promoting substance used for the treatment of narcolepsy. We hypothesized that modafinil would exacerbate RmTBI-induced deficits. Chronic behavioural analyses were completed along with a 27-plex serum cytokine array, metabolomic and proteomic analyses of cerebrospinal fluid (CSF), as well as immunohistochemical staining in structures important for sleep/wake cycles, to examine orexin, melanin-concentrating hormone, tyrosine hydroxylase, and choline acetyltransferase, in the lateral hypothalamus, locus coeruleus, and basal forebrain, respectively. Contrary to expectation, modafinil administration attenuated behavioural deficits, metabolomic changes, and neuropathological modifications. Therefore, the second aim was to determine if the beneficial effects of modafinil treatment were driven by the orexinergic system. The same experimental protocol was used; however, RmTBI rats received chronic orexin-A administration instead of modafinil. Orexin-A administration produced drastically different outcomes, exacerbating anxiety-related and motor deficits, while also significantly disrupting their metabolomic and neuropathological profiles. These results suggest that the beneficial effects of modafinil administration post-RmTBI, work independently of its wake-promoting properties, as activation of the orexinergic wake-promoting system with orexin-A was detrimental. Overall, these findings highlight the complexity of sleep-wake changes in the injured brain and showcase the potential of the arousal and sleep systems in its treatment.
Collapse
Affiliation(s)
- Jennaya Christensen
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Elaina Vlassopoulos
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Christopher K Barlow
- Monash Proteomics and Metabolomics Platform, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| | - Ralf B Schittenhelm
- Monash Proteomics and Metabolomics Platform, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| | - Crystal N Li
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Marissa Sgro
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Samantha Warren
- Monash Micro Imaging, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Bridgette D Semple
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Glenn R Yamakawa
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Sandy R Shultz
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia; Centre for Trauma and Mental Health Research, Vancouver Island University, Nanaimo, B.C., Canada
| | - Richelle Mychasiuk
- Department of Neuroscience, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia.
| |
Collapse
|
29
|
Guo L, Wu F, Yang J. Do patients with bipolar disorders have an increased risk of stroke? A systematic review and meta-analysis. Pak J Med Sci 2024; 40:2739-3745. [PMID: 39634868 PMCID: PMC11613353 DOI: 10.12669/pjms.40.11.10732] [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: 08/13/2024] [Revised: 08/21/2024] [Accepted: 10/15/2024] [Indexed: 12/07/2024] Open
Abstract
Objective The objective of the current review aimed to assess if patients with bipolar disorder (BD) have an increased risk of stroke. Methods A literature search was conducted on PubMed, Embase, ScienceDirect, and Web of Science for studies published from inception to 20th June 2023 and reporting the association between BD and stroke. Results Eight studies with 9187894 participants were eligible. Meta-analysis of all eight studies demonstrated that BD patients have an increased risk of stroke as compared to non-BD individuals (HR: 1.60 95% CI: 1.24, 2.05 I2=94%). Results did not change in significance on sensitivity analysis. However, results varied on subgroup analysis based on study location, study type, sample size, and follow-up duration. The effect magnitude was unchanged based on the adjustment of diabetes and hypertension and study quality. Conclusion Patients with bipolar disorder (BD) may have an increased risk of stroke in comparison to the general population. The results are limited by the retrospective nature of the data and high inter-study heterogeneity which demand caution in the interpretation of the outcomes. Protocol Registration: PROSPERO (CRD42023434365), https://www.crd.york.ac.uk/prospero/.
Collapse
Affiliation(s)
- Lingli Guo
- Lingli Guo, Department of Psychosomatic Disorders, Huzhou Third Municipal Hospital, The Affiliated Hospital of Huzhou University, Huzhou, Zhejiang Province 313000, P.R. China
| | - Fanghua Wu
- Fanghua Wu, Department of Psychosomatic Disorders, Huzhou Third Municipal Hospital, The Affiliated Hospital of Huzhou University, Huzhou, Zhejiang Province 313000, P.R. China
| | - Jiaojiao Yang
- Jiaojiao Yang, Department of Psychosomatic Disorders, Huzhou Third Municipal Hospital, The Affiliated Hospital of Huzhou University, Huzhou, Zhejiang Province 313000, P.R. China
| |
Collapse
|
30
|
Tanaka M, Szabó Á, Vécsei L. Redefining Roles: A Paradigm Shift in Tryptophan-Kynurenine Metabolism for Innovative Clinical Applications. Int J Mol Sci 2024; 25:12767. [PMID: 39684480 DOI: 10.3390/ijms252312767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 11/16/2024] [Accepted: 11/23/2024] [Indexed: 12/18/2024] Open
Abstract
The tryptophan-kynurenine (KYN) pathway has long been recognized for its essential role in generating metabolites that influence various physiological processes. Traditionally, these metabolites have been categorized into distinct, often opposing groups, such as pro-oxidant versus antioxidant, excitotoxic/neurotoxic versus neuroprotective. This dichotomous framework has shaped much of the research on conditions like neurodegenerative and neuropsychiatric disorders, as well as cancer, where metabolic imbalances are a key feature. The effects are significantly influenced by various factors, including the concentration of metabolites and the particular cellular milieu in which they are generated. A molecule that acts as neuroprotective at low concentrations may exhibit neurotoxic effects at elevated levels. The oxidative equilibrium of the surrounding environment can alter the function of KYN from an antioxidant to a pro-oxidant. This narrative review offers a comprehensive examination and analysis of the contemporary understanding of KYN metabolites, emphasizing their multifaceted biological functions and their relevance in numerous physiological and pathological processes. This underscores the pressing necessity for a paradigm shift in the comprehension of KYN metabolism. Understanding the context-dependent roles of KYN metabolites is vital for novel therapies in conditions like Alzheimer's disease, multiple sclerosis, and cancer. Comprehensive pathway modulation, including balancing inflammatory signals and enzyme regulation, offers promising avenues for targeted, effective treatments.
Collapse
Affiliation(s)
- Masaru Tanaka
- HUN-REN-SZTE Neuroscience Research Group, Hungarian Research Network, University of Szeged (HUN-REN-SZTE), Danube Neuroscience Research Laboratory, Tisza Lajos krt. 113, H-6725 Szeged, Hungary
| | - Ágnes Szabó
- Department of Neurology, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary
- Doctoral School of Clinical Medicine, University of Szeged, Korányi fasor 6, H-6720 Szeged, Hungary
| | - László Vécsei
- HUN-REN-SZTE Neuroscience Research Group, Hungarian Research Network, University of Szeged (HUN-REN-SZTE), Danube Neuroscience Research Laboratory, Tisza Lajos krt. 113, H-6725 Szeged, Hungary
- Department of Neurology, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary
| |
Collapse
|
31
|
Das A, Suar M, Reddy K. Hormones in malaria infection: influence on disease severity, host physiology, and therapeutic opportunities. Biosci Rep 2024; 44:BSR20240482. [PMID: 39492784 PMCID: PMC11581842 DOI: 10.1042/bsr20240482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 11/01/2024] [Accepted: 11/01/2024] [Indexed: 11/05/2024] Open
Abstract
Human malaria, caused by Plasmodium parasites, is a fatal disease that disrupts the host's physiological balance and affects the neuroendocrine system. This review explores how malaria influences and is influenced by hormones. Malaria activates the Hypothalamus-Pituitary-Adrenal axis, leading to increased cortisol, aldosterone, and epinephrine. Cortisol, while reducing inflammation, aids parasite survival, whereas epinephrine helps manage hypoglycemia. The Hypothalamus-Pituitary-Gonad and Hypothalamus-Pituitary-Thyroid axes are also impacted, resulting in lower sex and thyroid hormone levels. Malaria disrupts the renin-angiotensin-aldosterone system (RAAS), causing higher angiotensin-II and aldosterone levels, contributing to edema, hyponatremia and hypertension. Malaria-induced anemia is exacerbated by increased hepcidin, which impairs iron absorption, reducing both iron availability for the parasite and red blood cell formation, despite elevated erythropoietin. Hypoglycemia is common due to decreased glucose production and hyperinsulinemia, although some cases show hyperglycemia due to stress hormones and inflammation. Hypocalcemia, and hypophosphatemia are associated with low Vitamin D3 and parathyroid hormone but high calcitonin. Hormones such as DHEA, melatonin, PTH, Vitamin D3, hepcidin, progesterone, and erythropoietin protects against malaria. Furthermore, synthetic analogs, receptor agonists and antagonists or mimics of hormones like DHEA, melatonin, serotonin, PTH, vitamin D3, estrogen, progesterone, angiotensin, and somatostatin are being explored as potential antimalarial treatments or adjunct therapies. Additionally, hormones like leptin and PCT are being studied as probable markers of malaria infection.
Collapse
Affiliation(s)
- Aleena Das
- School of Biotechnology, Kalinga Institute of Industrial Technology (Deemed University), Bhubaneswar, 751024, India
| | - Mrutyunjay Suar
- School of Biotechnology, Kalinga Institute of Industrial Technology (Deemed University), Bhubaneswar, 751024, India
- Technology Business Incubator, Kalinga Institute of Industrial Technology (Deemed University), Bhubaneswar, 751024, India
| | - K Sony Reddy
- School of Biotechnology, Kalinga Institute of Industrial Technology (Deemed University), Bhubaneswar, 751024, India
| |
Collapse
|
32
|
Frost ED, Shi SX, Byroju VV, Pitton Rissardo J, Donlon J, Vigilante N, Murray BP, Walker IM, McGarry A, Ferraro TN, Hanafy KA, Echeverria V, Mitrev L, Kling MA, Krishnaiah B, Lovejoy DB, Rahman S, Stone TW, Koola MM. Galantamine-Memantine Combination in the Treatment of Parkinson's Disease Dementia. Brain Sci 2024; 14:1163. [PMID: 39766362 PMCID: PMC11674513 DOI: 10.3390/brainsci14121163] [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: 10/24/2024] [Revised: 11/16/2024] [Accepted: 11/20/2024] [Indexed: 01/11/2025] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder that affects over 1% of population over age 60. It is defined by motor and nonmotor symptoms including a spectrum of cognitive impairments known as Parkinson's disease dementia (PDD). Currently, the only US Food and Drug Administration-approved treatment for PDD is rivastigmine, which inhibits acetylcholinesterase and butyrylcholinesterase increasing the level of acetylcholine in the brain. Due to its limited efficacy and side effect profile, rivastigmine is often not prescribed, leaving patients with no treatment options. PD has several derangements in neurotransmitter pathways (dopaminergic neurons in the nigrostriatal pathway, kynurenine pathway (KP), acetylcholine, α7 nicotinic receptor, and N-methyl-D-aspartate (NMDA) receptors) and rivastigmine is only partially effective as it only targets one pathway. Kynurenic acid (KYNA), a metabolite of tryptophan metabolism, affects the pathophysiology of PDD in multiple ways. Both galantamine (α7 nicotinic receptor) and memantine (antagonist of the NMDA subtype of the glutamate receptor) are KYNA modulators. When used in combination, they target multiple pathways. While randomized controlled trials (RCTs) with each drug alone for PD have failed, the combination of galantamine and memantine has demonstrated a synergistic effect on cognitive enhancement in animal models. It has therapeutic potential that has not been adequately assessed, warranting future randomized controlled trials. In this review, we summarize the KYNA-centric model for PD pathophysiology and discuss how this treatment combination is promising in improving cognitive function in patients with PDD through its action on KYNA.
Collapse
Affiliation(s)
- Emma D. Frost
- Cooper Neurological Institute, Cooper University Health Care, Camden, NJ 08103, USA
| | - Swanny X. Shi
- Department of Neurology, Montefiore Medical Center, Bronx, NY 10467, USA
| | - Vishnu V. Byroju
- Cooper Neurological Institute, Cooper University Health Care, Camden, NJ 08103, USA
| | | | - Jack Donlon
- Cooper Medical School, Rowan University, Camden, NJ 08103, USA
| | | | | | - Ian M. Walker
- Cooper Neurological Institute, Cooper University Health Care, Camden, NJ 08103, USA
- Cooper Medical School, Rowan University, Camden, NJ 08103, USA
| | - Andrew McGarry
- Cooper Neurological Institute, Cooper University Health Care, Camden, NJ 08103, USA
- Cooper Medical School, Rowan University, Camden, NJ 08103, USA
| | - Thomas N. Ferraro
- Department of Biomedical Sciences, Cooper Medical School, Rowan University, Camden, NJ 08103, USA
| | - Khalid A. Hanafy
- Cooper Neurological Institute, Cooper University Health Care, Camden, NJ 08103, USA
- Cooper Medical School, Rowan University, Camden, NJ 08103, USA
| | - Valentina Echeverria
- Research and Development Department, Bay Pines VAHCS, Bay Pines, FL 33744, USA
- Medicine Department, Universidad San Sebastián, Concepción 4081339, Bío Bío, Chile
| | - Ludmil Mitrev
- Cooper Medical School, Rowan University, Camden, NJ 08103, USA
| | - Mitchel A. Kling
- Department of Geriatrics and Gerontology, New Jersey Institute for Successful Aging, Rowan-Virtua School of Osteopathic Medicine, Stratford, NJ 08084, USA
| | - Balaji Krishnaiah
- Department of Neurology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - David B. Lovejoy
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2113, Australia
| | - Shafiqur Rahman
- Department of Pharmaceutical Sciences, College of Pharmacy, South Dakota State University, Brookings, SD 57007, USA
| | - Trevor W. Stone
- Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford OX3 7LD, UK
| | - Maju Mathew Koola
- Department of Public Safety and Correctional Services, Baltimore, MD 21215, USA
| |
Collapse
|
33
|
Kis-György R, Körtési T, Anicka A, Nagy-Grócz G. The Connection Between the Oral Microbiota and the Kynurenine Pathway: Insights into Oral and Certain Systemic Disorders. Curr Issues Mol Biol 2024; 46:12641-12657. [PMID: 39590344 PMCID: PMC11593024 DOI: 10.3390/cimb46110750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 11/02/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
The oral microbiome, comprising bacteria, fungi, viruses, and protozoa, is essential for maintaining both oral and systemic health. This complex ecosystem includes over 700 bacterial species, such as Streptococcus mutans, which contributes to dental caries through acid production that demineralizes tooth enamel. Fungi like Candida and pathogens such as Porphyromonas gingivalis are also significant, as they can lead to periodontal diseases through inflammation and destruction of tooth-supporting structures. Dysbiosis, or microbial imbalance, is a key factor in the development of these oral diseases. Understanding the composition and functions of the oral microbiome is vital for creating targeted therapies for these conditions. Additionally, the kynurenine pathway, which processes the amino acid tryptophan, plays a crucial role in immune regulation, neuroprotection, and inflammation. Oral bacteria can metabolize tryptophan, influencing the production of kynurenine, kynurenic acid, and quinolinic acid, thereby affecting the kynurenine system. The balance of microbial species in the oral cavity can impact tryptophan levels and its metabolites. This narrative review aims to explore the relationship between the oral microbiome, oral diseases, and the kynurenine system in relation to certain systemic diseases.
Collapse
Affiliation(s)
- Rita Kis-György
- Section of Health Behaviour and Health Promotion, Faculty of Health Sciences and Social Studies, University of Szeged, Temesvári krt. 31., H-6726 Szeged, Hungary;
- Doctoral School of Interdisciplinary Medicine, University of Szeged, Szőkefalvi–Nagy Béla u. 4/B, H-6720 Szeged, Hungary
| | - Tamás Körtési
- Department of Theoretical Health Sciences and Health Management, Faculty of Health Sciences and Social Studies, University of Szeged, Temesvári krt. 31., H-6726 Szeged, Hungary;
- Preventive Health Sciences Research Group, Incubation Competence Centre of the Centre of Excellence for Interdisciplinary Research, Development and Innovation of the University of Szeged, H-6720 Szeged, Hungary
- HUN-REN-SZTE Neuroscience Research Group, Hungarian Research Network, Danube Neuroscience Research Laboratory, University of Szeged (HUN-REN-SZTE), Tisza Lajos krt. 113, H-6725 Szeged, Hungary
| | - Alexandra Anicka
- Department of Obstetrics and Gynecology, Semmelweis University, Üllői Út 78/A, H-1182 Budapest, Hungary;
| | - Gábor Nagy-Grócz
- Department of Theoretical Health Sciences and Health Management, Faculty of Health Sciences and Social Studies, University of Szeged, Temesvári krt. 31., H-6726 Szeged, Hungary;
- Preventive Health Sciences Research Group, Incubation Competence Centre of the Centre of Excellence for Interdisciplinary Research, Development and Innovation of the University of Szeged, H-6720 Szeged, Hungary
| |
Collapse
|
34
|
Shen H, Zhang Y, Shao Y, Chen S, Yin P, Liu X, Wang L, Zhang L, Jin Y, Wang Y, Xing R, Cho K, Jiang B. Synergism of salvianolic acid B and ginsenoside Rg1 magnifies the therapeutic potency against ischemic stroke. Neuroreport 2024; 35:1041-1051. [PMID: 39292959 PMCID: PMC11424057 DOI: 10.1097/wnr.0000000000002099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 09/02/2024] [Indexed: 09/20/2024]
Abstract
Even though considerable progress has been made to reduce insult, ischemic stroke is still a significant cause of mortality and morbidity in the world, and new therapeutic strategies are urgently needed. In the present study, the magnesium salt of salvianolic acid B (SalB) and ginsenoside Rg1 (Rg1) combination as a multicomponent strategy against stroke was evaluated. The synergistic effect of Sa1B and Rg1 was evaluated by Bliss independence analysis on the middle cerebral artery occlusion model. The infarct volume, neuroethology, cerebral structure, and neurocyte number were evaluated by 3,5-triphenyltetrazolium chloride staining, Longa score, Garcia score, hematoxylin-eosin staining, and Nissl staining, respectively. Metabolomics was used to search for potential biomarkers and explore the mechanism of Sa1B/Rg1. First, the superior effects of SalB/Rg1 than SalB or Rg1 at the same dose were evaluated. Compared with SalB ( P < 0.001) or Rg1 ( P < 0.01), SalB/Rg1 significantly decreased infarct volume through 3,5-triphenyltetrazolium chloride staining and protected the structural integrity of cortex and striatum. The superior effect of SalB/Rg1 on neurological behavior was also detected compared with SalB or Rg1 significantly. Accompanying behavioral improvement, a considerable increase of SalB/Rg1 on neurons detected by Nissl staining was found on the cortex compared with SalB ( P < 0.05) or Rg1 ( P < 0.01). Second, the synergistic effect between SalB and Rg1 was strictly verified by Bliss independence analysis ( P < 0.01) based on infarct volume. Finally, alleviation of cerebral metabolic disorders may be the possible mechanism of SalB/Rg1. Our study provided a multicomponent strategy against ischemic stroke, with not only dose reduction but also improved efficacy relative to single agents.
Collapse
Affiliation(s)
- Haishang Shen
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai
| | - Yuhan Zhang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai
| | - Yanan Shao
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai
- University of Chinese Academy of Sciences, Beijing
| | - Siqi Chen
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai
| | - Ping Yin
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan
| | - Xin Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan
| | - Linlin Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai
| | - Lingxiao Zhang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai
| | - Yi Jin
- Shenzhen Institute for Drug Control (Shenzhen Testing Center of Medical Devices), Shenzhen, China
| | - Yiyu Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai
| | - Rongrong Xing
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai
| | - Kenka Cho
- Takarazuka University of Medical and Health Care, Takarazuka, Japan
| | - Baohong Jiang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai
- University of Chinese Academy of Sciences, Beijing
| |
Collapse
|
35
|
Wang S, Qin M, Fan X, Jiang C, Hou Q, Ye Z, Zhang X, Yang Y, Xiao J, Wallace K, Rastegar-Kashkooli Y, Peng Q, Jin D, Wang J, Wang M, Ding R, Tao J, Kim YT, Bhawal UK, Wang J, Chen X, Wang J. The role of metal ions in stroke: Current evidence and future perspectives. Ageing Res Rev 2024; 101:102498. [PMID: 39243890 DOI: 10.1016/j.arr.2024.102498] [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: 03/18/2024] [Revised: 07/24/2024] [Accepted: 09/04/2024] [Indexed: 09/09/2024]
Abstract
Metal ions play a pivotal role in maintaining optimal brain function within the human body. Nevertheless, the accumulation of these ions can result in irregularities that lead to brain damage and dysfunction. Disruptions of metal ion homeostasis can result in various pathologies, including inflammation, redox dysregulation, and blood-brain barrier disruption. While research on metal ions has chiefly focused on neurodegenerative diseases, little attention has been given to their involvement in the onset and progression of stroke. Recent studies have identified cuproptosis and confirmed ferroptosis as significant factors in stroke pathology, underscoring the importance of metal ions in stroke pathology, including abnormal ion transport, neurotoxicity, blood-brain barrier damage, and cell death. Additionally, it provides an overview of contemporary metal ion chelators and detection techniques, which may offer novel approaches to stroke treatment.
Collapse
Affiliation(s)
- Shaoshuai Wang
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China; Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; Non-commissioned Officer School of Army Medical University, Shijiazhuang, Hebei 050000, China
| | - Mengzhe Qin
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Xiaochong Fan
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Chao Jiang
- Department of Neurology, People's Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China
| | - Qingchuan Hou
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Ziyi Ye
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Xinru Zhang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Yunfan Yang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Jingyu Xiao
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Kevin Wallace
- College of Mathematical and Natural Sciences, University of Maryland, College Park, MD 20742, USA
| | - Yousef Rastegar-Kashkooli
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China; School of International Education, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Qinfeng Peng
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Dongqi Jin
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Junyang Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Menglu Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Ruoqi Ding
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Jin Tao
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Yun Tai Kim
- Division of Functional Food Research, Korea Food Research Institute, 245, Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do 55365, Republic of Korea; Department of Food Biotechnology, Korea University of Science & Technology, 217 Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea
| | - Ujjal K Bhawal
- Center for Global Health Research, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu 600077, India; Research Institute of Oral Science, Nihon University School of Dentistry at Matsudo, Chiba 271-8587, Japan
| | - Junmin Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Xuemei Chen
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Jian Wang
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, China; Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| |
Collapse
|
36
|
Fu P, Zong Y, Dai Y, Zhu L, Chen S, Rastegar-Kashkooli Y, Wang J, Zhang J, Wang J, Jiang C. Vagal innervation limits brain injury by inhibiting gut-selective integrin-mediated intestinal immunocyte migration in intracerebral hemorrhage. Theranostics 2024; 14:7383-7404. [PMID: 39659582 PMCID: PMC11626938 DOI: 10.7150/thno.101680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 10/22/2024] [Indexed: 12/12/2024] Open
Abstract
Rationale: The vagus nerve, which connects the brain and gastrointestinal tract, helps to maintain immune balance in the intestines. Gut-specific integrins, on the other hand, help to keep immune cells in the intestines. Since immune cells from outside the intestines can significantly affect the outcome of strokes, we investigated how immune cells from the intestines affect the immune response in the brain during intracerebral hemorrhage (ICH). Methods: We aimed to examine the impact of vagal innervation on intestinal immunocyte trafficking and its influence on ICH outcomes using Kikume Green-Red (KikGR) and wildtype (WT) mice, with or without prior subdiaphragmatic vagotomy (SDV). Furthermore, we sought to elucidate the regulatory effects of vagal innervation on intestinal immunocyte trafficking by activating α7 nicotinic acetylcholine receptors (α7nAChR) in WT mice that underwent ICH after SDV. Additionally, we explored the potential intermediary role of gut-selective integrins in cholinergic transmitters-mediated intestinal immunocyte trafficking. Our methodology encompassed in vivo fluorescence imaging, flow cytometry, Western blotting, immunofluorescence staining, histopathology, and behavioral assessments to evaluate the outcomes. Results: Our findings reveal that during the acute phase of ICH, intestinal immunocytes migrated to various anatomical locations, including the circulation, hemorrhagic brain, meninges, and deep cervical lymph nodes. Pertinently, SDV resulted in diminished expression of α4β7 and αEβ7 integrins on immunocytes, leading to heightened intestinal immunocyte trafficking and exacerbated ICH outcomes. Conversely, the administration of α7nAChR agonists countered the adverse effects of vagotomy on α4β7 and αEβ7 integrin expression, thereby constraining the migration of immune cells from the intestines after ICH. The implication of α4β7 and αEβ7 integrins in this setting was supported by the ineffective influence of α7nAChR agonists on the trafficking of intestinal immunocytes enhanced by administering beta-7 integrin antagonists, such as etrolizumab. It was further supported by the exacerbated ICH outcomes by administering beta-7 integrin antagonists like etrolizumab alone. Conclusion: The identification of vagus nerve-mediated modulation of α4β7 and αEβ7 integrin expression in the trafficking of immune cells within the intestinal tract holds significant implications. This discovery presents new opportunities for developing therapeutic interventions for ICH and stimulates further investigation in this area.
Collapse
Affiliation(s)
- Peiji Fu
- Department of Neurology, The People's Hospital of Zhengzhou University & Henan Provincial People's Hospital, 450003, Zhengzhou, P. R. China
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, P. R. China
- The Laboratory of Cerebrovascular Diseases and Neuroimmunology, The Fifth Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, P. R. China
| | - Yan Zong
- Department of Neurology, The People's Hospital of Zhengzhou University & Henan Provincial People's Hospital, 450003, Zhengzhou, P. R. China
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, P. R. China
- The Laboratory of Cerebrovascular Diseases and Neuroimmunology, The Fifth Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, P. R. China
| | - Yinming Dai
- Department of Neurology, The People's Hospital of Zhengzhou University & Henan Provincial People's Hospital, 450003, Zhengzhou, P. R. China
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, P. R. China
| | - Li Zhu
- Department of Neurology, The People's Hospital of Zhengzhou University & Henan Provincial People's Hospital, 450003, Zhengzhou, P. R. China
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, P. R. China
| | - Shuai Chen
- Department of Neurology, The People's Hospital of Zhengzhou University & Henan Provincial People's Hospital, 450003, Zhengzhou, P. R. China
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, P. R. China
- The Laboratory of Cerebrovascular Diseases and Neuroimmunology, The Fifth Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, P. R. China
| | - Yousef Rastegar-Kashkooli
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, 450001, Zhengzhou, P. R. China
| | - Junmin Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, 450001, Zhengzhou, P. R. China
| | - Jiewen Zhang
- Department of Neurology, The People's Hospital of Zhengzhou University & Henan Provincial People's Hospital, 450003, Zhengzhou, P. R. China
| | - Jian Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, 450001, Zhengzhou, P. R. China
| | - Chao Jiang
- Department of Neurology, The People's Hospital of Zhengzhou University & Henan Provincial People's Hospital, 450003, Zhengzhou, P. R. China
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, P. R. China
- The Laboratory of Cerebrovascular Diseases and Neuroimmunology, The Fifth Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, P. R. China
| |
Collapse
|
37
|
Bogielski B, Michalczyk K, Głodek P, Tempka B, Gębski W, Stygar D. Association between small intestine bacterial overgrowth and psychiatric disorders. Front Endocrinol (Lausanne) 2024; 15:1438066. [PMID: 39497810 PMCID: PMC11532184 DOI: 10.3389/fendo.2024.1438066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 10/01/2024] [Indexed: 11/07/2024] Open
Abstract
Small intestinal bacterial overgrowth (SIBO) is a gastrointestinal condition characterized by abnormal colonization of bacteria in the small intestine, leading to overgrowth and alteration, which is linked to gastrointestinal issues, potentially affecting neurological and mental health. Despite existing research, we still do not understand how SIBO affects tryptophan metabolism and psychiatric diseases. We investigated the literature for connections between SIBO, tryptophan metabolism disruptions, and psychiatric disorders like autism, schizophrenia, Alzheimer's, and Parkinson's diseases. We also explored the interaction between thyroid disorders and their influence on SIBO and psychiatric illnesses. PubMed and Google Scholar databases were searched using keywords and phrases, individual and in combinations, like "SIBO," "gut microbiota," "neurologic disorders," "mental disorders," "tryptophan," "dopamine," and "thyroid disease." We focused on original research and review papers that presented empirical studies conducted on animal models and human subjects published in English between February 1992 to February 2023. The initial 2 634 534 records were preliminary screened based on title and abstract and then subjected to full-text review to exclude publications with insufficient data on SIBO, lack of a psychiatric disorder component, or methodological limitations compromising the integrity of the findings. The analysis highlights the significance of the association between psychiatric disorders and SIBO, emphasizing the role of gut-microbial diversity in mental health. We advocate for more detailed studies, including longitudinal research, to clarify the causal relationships between SIBO, gut dysbiosis, and psychiatric disorders and for an integrated approach while treating complex psychiatric conditions.
Collapse
Affiliation(s)
| | | | | | | | | | - Dominika Stygar
- Department of Physiology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Zabrze, Poland
| |
Collapse
|
38
|
Yuan X, Hu S, Fan X, Jiang C, Xu Y, Hao R, Xu Z, Yu Y, Rastegar-Kashkooli Y, Huang L, Wang TJ, Wang Q, Su S, Wang L, Wang J, Wang M, Kim YT, Bhawal UK, Wang F, Zhao T, Wang J, Chen X, Wang J. Central post-stroke pain: advances in clinical and preclinical research. Stroke Vasc Neurol 2024:svn-2024-003418. [PMID: 39343438 DOI: 10.1136/svn-2024-003418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 09/04/2024] [Indexed: 10/01/2024] Open
Abstract
Central poststroke pain (CPSP) is a medical complication that arises poststroke and significantly impacts the quality of life and social functioning of affected individuals. Despite ongoing research, the exact pathomechanisms of CPSP remain unclear, and practical treatments are still unavailable. Our review aims to systematically analyse current clinical and preclinical studies on CPSP, which is critical for identifying gaps in knowledge and guiding the development of effective therapies. The review will clarify the clinical characteristics, evaluation scales and contemporary therapeutic approaches for CPSP based on clinical investigations. It will particularly emphasise the CPSP model initiated by stroke, shedding light on its underlying mechanisms and evaluating treatments validated in preclinical studies. Furthermore, the review will not only highlight methodological limitations in animal trials but also offer specific recommendations to researchers to improve the quality of future investigations and guide the development of effective therapies. This review is expected to provide valuable insights into the current knowledge regarding CPSP and can serve as a guide for future research and clinical practice. The review will contribute to the scientific understanding of CPSP and help develop effective clinical interventions.
Collapse
Affiliation(s)
- Xiqian Yuan
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Siyuan Hu
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiaochong Fan
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Chao Jiang
- Department of Neurology, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yan Xu
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Ruochen Hao
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Zili Xu
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Yiyang Yu
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Yousef Rastegar-Kashkooli
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- School of International Education, Zhengzhou University, Zhengzhou, Henan, China
| | - Leo Huang
- Department of Psychology, University of Toronto, Toronto, Ontario, Canada
| | - Tom J Wang
- Program in Behavioral Biology, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Qiao Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Songxue Su
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Limin Wang
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Junyang Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Menglu Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Yun Tai Kim
- Division of Functional Food Research, Korea Food Research Institute, Wanju-gun, Wanju Jeollabuk-do, Korea (the Republic of)
- Department of Food Biotechnology, Korea University of Science & Technology, Daejeon, Korea (the Republic of)
| | - Ujjal K Bhawal
- Center for Global Health Research, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil nadu, India
- Research Institute of Oral Science, Nihon University School of Dentistry at Matsudo, Chiba, Japan
| | - Fushun Wang
- Department of Psychology, Sichuan Normal University, Chengdu, Sichuan, China
| | - Ting Zhao
- Department of Neurology, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Junmin Wang
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Xuemei Chen
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Jian Wang
- Department of Pain Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Human Anatomy, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
39
|
Samriti, Kaur A, Kaur A, Goel RK. Ameliorative effect of diclofenac in rotenone corneal kindling model of drug-resistant epilepsy: Edge of dual COX and KMO inhibition. Brain Res 2024; 1846:149246. [PMID: 39304107 DOI: 10.1016/j.brainres.2024.149246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 09/15/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024]
Abstract
Epilepsy affects millions of people worldwide, about one-third patients with epilepsy exhibits resistance to available antiseizures medications, known as drug-resistant epilepsy (DRE). Mitochondrial dysfunction has been implicated as a hallmark in drug-resistant epilepsy via activation of microglial kynurenine 3-monooxygenase (KMO) and cyclooxygenase (COX) enzymes, leading to neuroinflammation and oxidative stress. Diclofenac, an equipotent non selective cyclooxygenase inhibitor, has inhibitory action on KMO enzyme and has also shown anti-inflammatory and antioxidant properties in animal models of epilepsy. These properties make it a suitable candidate for amelioration of DRE. However, its potential in drug-resistant epilepsy remained unexplored till date. In this study, dose dependent effect of diclofenac (5 mg/kg, 10 mg/kg, 20 mg/kg) has been explored in rotenone corneal kindling model of mitochondrial DRE. The results of our study revealed the induction of drug resistance to antiseizure medications and induced kynurenine 3-monooxygenase activity in rotenone corneal kindled epileptic mice in comparison to naive mice. Treatment of rotenone corneal kindled epileptic mice with diclofenac resulted in a significant decrease in drug resistance to antiseizure medications as evident by a reduction in seizure score in the treatment groups as compared to control group, in post-treatment resistance validation. The kynurenine 3-monooxygenase inhibitory activity (as evidenced by decreased levels of neurotoxic quinolinic acid) and the antioxidant effect (as evident by significantly reduced oxidative stress) in the diclofenac treated groups, emerged as a major contributor for its ameliorative action. Findings of this study suggests, diclofenac can be used as an adjunct therapy in amelioration of drug-resistant epilepsy.
Collapse
Affiliation(s)
- Samriti
- Department of Pharmaceutical Sciences & Drug Research, Punjabi University, Patiala, 147002 India.
| | - Arvinder Kaur
- Department of Pharmaceutical Sciences & Drug Research, Punjabi University, Patiala, 147002 India.
| | - Arshbir Kaur
- Department of Pharmaceutical Sciences & Drug Research, Punjabi University, Patiala, 147002 India.
| | - R K Goel
- Department of Pharmaceutical Sciences & Drug Research, Punjabi University, Patiala, 147002 India.
| |
Collapse
|
40
|
Liu Y, Zhao P, Cai Z, He P, Wang J, He H, Zhu Z, Guo X, Ma K, Peng K, Zhao J. Buqi-Huoxue-Tongnao decoction drives gut microbiota-derived indole lactic acid to attenuate ischemic stroke via the gut-brain axis. Chin Med 2024; 19:126. [PMID: 39278929 PMCID: PMC11403783 DOI: 10.1186/s13020-024-00991-1] [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: 05/30/2024] [Accepted: 08/28/2024] [Indexed: 09/18/2024] Open
Abstract
BACKGROUND Ischemic stroke belongs to "apoplexy" and its pathogenesis is characterized by qi deficiency and blood stasis combining with phlegm-damp clouding orifices. Buqi-Huoxue-Tongnao decoction (BHTD) is a traditional Chinese medicine formula for qi deficiency, blood stasis and phlegm obstruction syndrome. However, its efficacy and potential mechanism on ischemic stroke are still unclear. This study aims to investigate the protective effect and potential mechanism of BHTD against ischemic stroke. MATERIALS AND METHODS Middle cerebral artery occlusion (MCAO) surgery was carried out to establish an ischemic stroke model in rats. Subsequently, the rats were gavaged with different doses of BHTD (2.59, 5.175, 10.35 g/kg) for 14 days. The protective effects of BHTD on the brain and gut were evaluated by neurological function scores, cerebral infarction area, levels of brain injury markers (S-100B, NGB), indicators of gut permeability (FD-4) and bacterial translocation (DAO, LPS, D-lactate), and tight junction proteins (Occludin, Claudin-1, ZO-1) in brain and colon. 16S rRNA gene sequencing and metabolomic analysis were utilized to analyze the effects on gut microecology and screen for marker metabolites to explore potential mechanisms of BHTD protection against ischemic stroke. RESULTS BHTD could effectively mitigate brain impairment, including reducing neurological damage, decreasing cerebral infarction and repairing the blood-brain barrier, and BHTD showed the best effect at the dose of 10.35 g/kg. Moreover, BHTD reversed gut injury induced by ischemic stroke, as evidenced by decreased intestinal permeability, reduced intestinal bacterial translocation, and enhanced intestinal barrier integrity. In addition, BHTD rescued gut microbiota dysbiosis by increasing the abundance of beneficial bacteria, including Turicibacter and Faecalibaculum. Transplantation of the gut microbiota remodeled by BHTD into ischemic stroke rats recapitulated the protective effects of BHTD. Especially, BHTD upregulated tryptophan metabolism, which promoted gut microbiota to produce more indole lactic acid (ILA). Notably, supplementation with ILA by gavage could alleviate stroke injury, which suggested that driving the production of ILA in the gut might be a novel treatment for ischemic stroke. CONCLUSION BHTD could increase gut microbiota-derived indole lactic acid to attenuate ischemic stroke via the gut-brain axis. Our current finding provides evidence that traditional Chinese medicine can ameliorate central diseases through regulating the gut microbiology.
Collapse
Affiliation(s)
- Yarui Liu
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism and Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Peng Zhao
- Peng Kang National Famous Traditional Chinese Medicine Expert Inheritance Studio, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510315, Guangdong, China
| | - Zheng Cai
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism and Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, Guangdong, China
- Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510315, Guangdong, China
| | - Peishi He
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism and Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Jiahan Wang
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism and Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Haoqing He
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism and Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Zhibo Zhu
- Peng Kang National Famous Traditional Chinese Medicine Expert Inheritance Studio, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510315, Guangdong, China
| | - Xiaowen Guo
- Peng Kang National Famous Traditional Chinese Medicine Expert Inheritance Studio, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510315, Guangdong, China
| | - Ke Ma
- Peng Kang National Famous Traditional Chinese Medicine Expert Inheritance Studio, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510315, Guangdong, China
| | - Kang Peng
- Peng Kang National Famous Traditional Chinese Medicine Expert Inheritance Studio, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510315, Guangdong, China.
| | - Jie Zhao
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism and Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, Guangdong, China.
- Peng Kang National Famous Traditional Chinese Medicine Expert Inheritance Studio, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510315, Guangdong, China.
- Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510315, Guangdong, China.
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| |
Collapse
|
41
|
Alfaro-Rodríguez A, Reyes-Long S, Roldan-Valadez E, González-Torres M, Bonilla-Jaime H, Bandala C, Avila-Luna A, Bueno-Nava A, Cabrera-Ruiz E, Sanchez-Aparicio P, González Maciel A, Dotor-Llerena AL, Cortes-Altamirano JL. Association of the Serotonin and Kynurenine Pathways as Possible Therapeutic Targets to Modulate Pain in Patients with Fibromyalgia. Pharmaceuticals (Basel) 2024; 17:1205. [PMID: 39338367 PMCID: PMC11434812 DOI: 10.3390/ph17091205] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 08/27/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
Fibromyalgia (FM) is a disorder characterized by widespread chronic pain, significant depression, and various neural abnormalities. Recent research suggests a reciprocal exacerbation mechanism between chronic pain and depression. In patients with FM, dysregulation of tryptophan (Trp) metabolism has been identified. Trp, an essential amino acid, serves as a precursor to serotonin (5-HT), a neuromodulator that influences mood, appetite, sleep, and pain perception through the receptors 5-HT1, 5-HT2, and 5-HT3. Additionally, Trp is involved in the kynurenine pathway, a critical route in the immune response, inflammation, and production of neuroactive substances and nicotinamide adenine dinucleotide (NAD+). The activation of this pathway by pro-inflammatory cytokines, such as tumor necrosis factor α (TNF-α) and interferon gamma (IFN-γ), leads to the production of kynurenic acid (KYNA), which has neuroprotective properties, and quinolinic acid (QA), which is neurotoxic. These findings underscore the crucial balance between Trp metabolism, 5-HT, and kynurenine, where an imbalance can contribute to the dual burden of pain and depression in patients with FM. This review proposes a novel therapeutic approach for FM pain management, focusing on inhibiting QA synthesis while co-administering selective serotonin reuptake inhibitors to potentially increase KYNA levels, thus dampening pain perception and improving patient outcomes.
Collapse
Affiliation(s)
- Alfonso Alfaro-Rodríguez
- Division of Basic Neurosciences, Instituto Nacional de Rehabilitación, "Luis Guillermo Ibarra Ibarra", Mexico City 14389, Mexico
| | - Samuel Reyes-Long
- Division of Basic Neurosciences, Instituto Nacional de Rehabilitación, "Luis Guillermo Ibarra Ibarra", Mexico City 14389, Mexico
| | - Ernesto Roldan-Valadez
- Division of Basic Neurosciences, Instituto Nacional de Rehabilitación, "Luis Guillermo Ibarra Ibarra", Mexico City 14389, Mexico
- Department of Radiology, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119992 Moscow, Russia
| | - Maykel González-Torres
- Conahcyt & Biotechnology Laboratory, Instituto Nacional de Rehabilitación, "Luis Guillermo Ibarra Ibarra", Mexico City 03940, Mexico
| | - Herlinda Bonilla-Jaime
- Department of Reproductive Biology, Universidad Autónoma Metropolitana Iztapalapa, Mexico City 09340, Mexico
| | - Cindy Bandala
- Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Alberto Avila-Luna
- Division of Basic Neurosciences, Instituto Nacional de Rehabilitación, "Luis Guillermo Ibarra Ibarra", Mexico City 14389, Mexico
| | - Antonio Bueno-Nava
- Division of Basic Neurosciences, Instituto Nacional de Rehabilitación, "Luis Guillermo Ibarra Ibarra", Mexico City 14389, Mexico
| | - Elizabeth Cabrera-Ruiz
- Division of Basic Neurosciences, Instituto Nacional de Rehabilitación, "Luis Guillermo Ibarra Ibarra", Mexico City 14389, Mexico
| | - Pedro Sanchez-Aparicio
- Pharmacology Department, Facultad de Medicina Veterinaria, Universidad Autónoma del Estado de México, Toluca 50090, Mexico
| | - Angélica González Maciel
- Laboratory of Cell and Tissue Morphology, Instituto Nacional de Pediatría, Mexico City 04530, Mexico
| | - Ana Lilia Dotor-Llerena
- Division of Clinic Neurosciences, Instituto Nacional de Rehabilitación, "Luis Guillermo Ibarra Ibarra", Mexico City 14389, Mexico
| | - José Luis Cortes-Altamirano
- Division of Basic Neurosciences, Instituto Nacional de Rehabilitación, "Luis Guillermo Ibarra Ibarra", Mexico City 14389, Mexico
- Department of Chiropractic, Universidad Estatal del Valle de Ecatepec, Ecatepec de Morelos 55210, Mexico
| |
Collapse
|
42
|
Li Y, Wu C, Yang R, Tang J, Li Z, Yi X, Fan Z. Application and Development of Cell Membrane Functionalized Biomimetic Nanoparticles in the Treatment of Acute Ischemic Stroke. Int J Mol Sci 2024; 25:8539. [PMID: 39126107 PMCID: PMC11313357 DOI: 10.3390/ijms25158539] [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: 05/25/2024] [Revised: 08/01/2024] [Accepted: 08/02/2024] [Indexed: 08/12/2024] Open
Abstract
Ischemic stroke is a serious neurological disease involving multiple complex physiological processes, including vascular obstruction, brain tissue ischemia, impaired energy metabolism, cell death, impaired ion pump function, and inflammatory response. In recent years, there has been significant interest in cell membrane-functionalized biomimetic nanoparticles as a novel therapeutic approach. This review comprehensively explores the mechanisms and importance of using these nanoparticles to treat acute ischemic stroke with a special emphasis on their potential for actively targeting therapies through cell membranes. We provide an overview of the pathophysiology of ischemic stroke and present advances in the study of biomimetic nanoparticles, emphasizing their potential for drug delivery and precision-targeted therapy. This paper focuses on bio-nanoparticles encapsulated in bionic cell membranes to target ischemic stroke treatment. It highlights the mechanism of action and research progress regarding different types of cell membrane-functionalized bi-onic nanoparticles such as erythrocytes, neutrophils, platelets, exosomes, macrophages, and neural stem cells in treating ischemic stroke while emphasizing their potential to improve brain tissue's ischemic state and attenuate neurological damage and dysfunction. Through an in-depth exploration of the potential benefits provided by cell membrane-functionalized biomimetic nanoparticles to improve brain tissue's ischemic state while reducing neurological injury and dysfunction, this study also provides comprehensive research on neural stem cells' potential along with that of cell membrane-functionalized biomimetic nanoparticles to ameliorate neurological injury and dysfunction. However, it is undeniable that there are still some challenges and limitations in terms of biocompatibility, safety, and practical applications for clinical translation.
Collapse
Affiliation(s)
- Ying Li
- Xiamen Key Laboratory of Traditional Chinese Bio-Engineering, Xiamen Medical College, Xiamen 361021, China
| | - Chuang Wu
- Xiamen Key Laboratory of Traditional Chinese Bio-Engineering, Xiamen Medical College, Xiamen 361021, China
| | - Rui Yang
- Xiamen Key Laboratory of Traditional Chinese Bio-Engineering, Xiamen Medical College, Xiamen 361021, China
| | - Jiannan Tang
- Xiamen Key Laboratory of Traditional Chinese Bio-Engineering, Xiamen Medical College, Xiamen 361021, China
| | - Zhanqing Li
- Xiamen Key Laboratory of Traditional Chinese Bio-Engineering, Xiamen Medical College, Xiamen 361021, China
| | - Xue Yi
- Xiamen Key Laboratory of Traditional Chinese Bio-Engineering, Xiamen Medical College, Xiamen 361021, China
| | - Zhongxiong Fan
- School of Pharmaceutical Sciences and Institute of Materia Medica, Xinjiang University, Urumqi 830017, China
| |
Collapse
|
43
|
Plaatjie ON, van Furth AMT, van der Kuip M, Mason S. LC-MS metabolomics and lipidomics in cerebrospinal fluid from viral and bacterial CNS infections: a review. Front Neurol 2024; 15:1403312. [PMID: 39161867 PMCID: PMC11330781 DOI: 10.3389/fneur.2024.1403312] [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: 03/19/2024] [Accepted: 07/25/2024] [Indexed: 08/21/2024] Open
Abstract
There is compelling evidence that a dysregulated immune inflammatory response in neuroinfectious diseases results in modifications in metabolic processes and altered metabolites, directly or indirectly influencing lipid metabolism within the central nervous system (CNS). The challenges in differential diagnosis and the provision of effective treatment in many neuroinfectious diseases are, in part, due to limited understanding of the pathophysiology underlying the disease. Although there are numerous metabolomics studies, there remains a deficit in neurolipidomics research to provide a comprehensive understanding of the connection between altered metabolites and changes in lipid metabolism. The brain is an inherently high-lipid organ; hence, understanding neurolipidomics is the key to future breakthroughs. This review aims to provide an integrative summary of altered cerebrospinal fluid (CSF) metabolites associated with neurolipid metabolism in bacterial and viral CNS infections, with a particular focus on studies that used liquid chromatography-mass spectrometry (LC-MS). Lipid components (phospholipids) and metabolites (carnitine and tryptophan) appear to be the most significant indicators in both bacterial and viral infections. On the basis of our analysis of the literature, we recommend employing neurolipidomics in conjunction with existing neurometabolomics data as a prospective method to enhance our understanding of the cross link between dysregulated metabolites and lipid metabolism in neuroinfectious diseases.
Collapse
Affiliation(s)
- Ontefetse Neo Plaatjie
- Department of Biochemistry, Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom, South Africa
| | - A. Marceline Tutu van Furth
- Department of Pediatric Infectious Diseases and Immunology, Pediatric Infectious Diseases and Immunology, Amsterdam University Medical Center, Emma Children’s Hospital, Amsterdam, Netherlands
| | - Martijn van der Kuip
- Department of Pediatric Infectious Diseases and Immunology, Pediatric Infectious Diseases and Immunology, Amsterdam University Medical Center, Emma Children’s Hospital, Amsterdam, Netherlands
| | - Shayne Mason
- Department of Biochemistry, Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom, South Africa
| |
Collapse
|
44
|
Zhu L, Shang J, Li Y, Zhang Z, Fu P, Zong Y, Chen S, Wang J, Zhang J, Wang J, Jiang C. Toll-Like Receptors Mediate Opposing Dendritic Cell Effects on Treg/Th17 Balance in Mice With Intracerebral Hemorrhage. Stroke 2024; 55:2126-2138. [PMID: 38920054 DOI: 10.1161/strokeaha.124.046394] [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: 01/02/2024] [Accepted: 05/23/2024] [Indexed: 06/27/2024]
Abstract
BACKGROUND Dendritic cells (DCs) regulate the immune response associated with T lymphocytes, but their role in stroke remains unclear. In this study, we investigated the causal relationship between DCs and T-cell response in intracerebral hemorrhage (ICH) by focusing on TLRs (toll-like receptors) that may modulate the function of DCs. METHODS We studied the effects of TLR4, TLR2, and TLR9 on DC-mediated T-cell response and the outcomes of ICH using male C57BL/6 and CD11c-DTx (diphtheria toxin) receptor mice. We administered specific agents intraperitoneally or orally and evaluated the results using flow cytometry, real-time polymerase chain reaction, Western blotting, immunofluorescence staining, histopathology, and behavioral tests. RESULTS TLR4 and TLR2 activation induces DC maturation and reduces the ratio of regulatory T to T-helper 17 cells in the brain and periphery after ICH. When either of these receptors is activated, it can worsen neuroinflammation and exacerbate ICH outcomes. TLR9 also promotes DC maturation, stabilizing the number of DCs, particularly conventional DCs. TLR9 has the opposite effects on regulatory T/T-helper 17 balance, neuroinflammation, and ICH outcomes compared with TLR4 and TLR2. Upon stimulation, TLR4 and TLR9 may achieve these effects through the p38-MAPK (p38-mitogen-activated protein kinase)/MyD88 (myeloid differentiation primary response gene 88) and indoleamine 2,3-dioxygenase 1 (IDO1)/GCN2 (general control nonderepressible 2) signaling pathways, respectively. DCs act as intermediaries for TLR-mediated T-cell response. CONCLUSIONS TLR-mediated opposing effects of DCs on T-cell response may provide novel strategies to treat ICH.
Collapse
Affiliation(s)
- Li Zhu
- Department of Neurology (L.Z., Y.L., Z.Z., P.F., Y.Z., S.C., C.J.), The Fifth Affiliated Hospital of Zhengzhou University, China
- The Laboratory of Cerebrovascular Diseases and Neuroimmunology (L.Z., Y.L., Z.Z., P.F., Y.Z., S.C., C.J.), The Fifth Affiliated Hospital of Zhengzhou University, China
| | - Junkui Shang
- Department of Neurology, People's Hospital of Zhengzhou University, China (J.S., J.Z., C.J.)
| | - Yinuo Li
- Department of Neurology (L.Z., Y.L., Z.Z., P.F., Y.Z., S.C., C.J.), The Fifth Affiliated Hospital of Zhengzhou University, China
- The Laboratory of Cerebrovascular Diseases and Neuroimmunology (L.Z., Y.L., Z.Z., P.F., Y.Z., S.C., C.J.), The Fifth Affiliated Hospital of Zhengzhou University, China
| | - Zhiying Zhang
- Department of Neurology (L.Z., Y.L., Z.Z., P.F., Y.Z., S.C., C.J.), The Fifth Affiliated Hospital of Zhengzhou University, China
- The Laboratory of Cerebrovascular Diseases and Neuroimmunology (L.Z., Y.L., Z.Z., P.F., Y.Z., S.C., C.J.), The Fifth Affiliated Hospital of Zhengzhou University, China
| | - Peiji Fu
- Department of Neurology (L.Z., Y.L., Z.Z., P.F., Y.Z., S.C., C.J.), The Fifth Affiliated Hospital of Zhengzhou University, China
- The Laboratory of Cerebrovascular Diseases and Neuroimmunology (L.Z., Y.L., Z.Z., P.F., Y.Z., S.C., C.J.), The Fifth Affiliated Hospital of Zhengzhou University, China
| | - Yan Zong
- Department of Neurology (L.Z., Y.L., Z.Z., P.F., Y.Z., S.C., C.J.), The Fifth Affiliated Hospital of Zhengzhou University, China
- The Laboratory of Cerebrovascular Diseases and Neuroimmunology (L.Z., Y.L., Z.Z., P.F., Y.Z., S.C., C.J.), The Fifth Affiliated Hospital of Zhengzhou University, China
| | - Shuai Chen
- Department of Neurology (L.Z., Y.L., Z.Z., P.F., Y.Z., S.C., C.J.), The Fifth Affiliated Hospital of Zhengzhou University, China
- The Laboratory of Cerebrovascular Diseases and Neuroimmunology (L.Z., Y.L., Z.Z., P.F., Y.Z., S.C., C.J.), The Fifth Affiliated Hospital of Zhengzhou University, China
| | - Junmin Wang
- Department of Anatomy, School of Basic Medical Sciences, Zhengzhou University, China (Junmin Wang, Jian Wang)
| | - Jiewen Zhang
- Department of Neurology, People's Hospital of Zhengzhou University, China (J.S., J.Z., C.J.)
| | - Jian Wang
- Department of Anatomy, School of Basic Medical Sciences, Zhengzhou University, China (Junmin Wang, Jian Wang)
| | - Chao Jiang
- Department of Neurology (L.Z., Y.L., Z.Z., P.F., Y.Z., S.C., C.J.), The Fifth Affiliated Hospital of Zhengzhou University, China
- The Laboratory of Cerebrovascular Diseases and Neuroimmunology (L.Z., Y.L., Z.Z., P.F., Y.Z., S.C., C.J.), The Fifth Affiliated Hospital of Zhengzhou University, China
- Department of Neurology, People's Hospital of Zhengzhou University, China (J.S., J.Z., C.J.)
| |
Collapse
|
45
|
Jia P, Peng Q, Fan X, Zhang Y, Xu H, Li J, Sonita H, Liu S, Le A, Hu Q, Zhao T, Zhang S, Wang J, Zille M, Jiang C, Chen X, Wang J. Immune-mediated disruption of the blood-brain barrier after intracerebral hemorrhage: Insights and potential therapeutic targets. CNS Neurosci Ther 2024; 30:e14853. [PMID: 39034473 PMCID: PMC11260770 DOI: 10.1111/cns.14853] [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: 05/17/2024] [Revised: 06/21/2024] [Accepted: 07/02/2024] [Indexed: 07/23/2024] Open
Abstract
AIMS Intracerebral hemorrhage (ICH) is a condition that arises due to the rupture of cerebral blood vessels, leading to the flow of blood into the brain tissue. One of the pathological alterations that occurs during an acute ICH is an impairment of the blood-brain barrier (BBB), which leads to severe perihematomal edema and an immune response. DISCUSSION A complex interplay between the cells of the BBB, for example, pericytes, astrocytes, and brain endothelial cells, with resident and infiltrating immune cells, such as microglia, monocytes, neutrophils, T lymphocytes, and others accounts for both damaging and protective mechanisms at the BBB following ICH. However, the precise immunological influence of BBB disruption has yet to be richly ascertained, especially at various stages of ICH. CONCLUSION This review summarizes the changes in different cell types and molecular components of the BBB associated with immune-inflammatory responses during ICH. Furthermore, it highlights promising immunoregulatory therapies to protect the integrity of the BBB after ICH. By offering a comprehensive understanding of the mechanisms behind BBB damage linked to cellular and molecular immunoinflammatory responses after ICH, this article aimed to accelerate the identification of potential therapeutic targets and expedite further translational research.
Collapse
Affiliation(s)
- Peijun Jia
- Department of Pain MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Department of Human AnatomySchool of Basic Medical Sciences of Zhengzhou UniversityZhengzhouChina
- School of Life SciencesZhengzhou UniversityZhengzhouChina
| | - Qinfeng Peng
- Department of Human AnatomySchool of Basic Medical Sciences of Zhengzhou UniversityZhengzhouChina
| | - Xiaochong Fan
- Department of Pain MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Yumeng Zhang
- Department of Human AnatomySchool of Basic Medical Sciences of Zhengzhou UniversityZhengzhouChina
| | - Hanxiao Xu
- Department of Human AnatomySchool of Basic Medical Sciences of Zhengzhou UniversityZhengzhouChina
| | - Jiaxin Li
- Department of Human AnatomySchool of Basic Medical Sciences of Zhengzhou UniversityZhengzhouChina
| | - Houn Sonita
- Department of Human AnatomySchool of Basic Medical Sciences of Zhengzhou UniversityZhengzhouChina
| | - Simon Liu
- David Geffen School of MedicineUniversity of California Los AngelesLos AngelesCaliforniaUSA
| | - Anh Le
- George Washington School of Medicine and Health SciencesWashingtonDCUSA
| | - Qiongqiong Hu
- Department of NeurologyZhengzhou Central Hospital Affiliated to Zhengzhou UniversityZhengzhouHenanChina
| | - Ting Zhao
- Department of NeurologyPeople's Hospital of Zhengzhou UniversityZhengzhouChina
| | - Shijie Zhang
- School of Life SciencesZhengzhou UniversityZhengzhouChina
| | - Junmin Wang
- Department of Human AnatomySchool of Basic Medical Sciences of Zhengzhou UniversityZhengzhouChina
| | - Marietta Zille
- Division of Pharmacology and Toxicology, Department of Pharmaceutical SciencesUniversity of ViennaViennaAustria
| | - Chao Jiang
- Department of NeurologyPeople's Hospital of Zhengzhou UniversityZhengzhouChina
| | - Xuemei Chen
- Department of Human AnatomySchool of Basic Medical Sciences of Zhengzhou UniversityZhengzhouChina
| | - Jian Wang
- Department of Pain MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Department of Human AnatomySchool of Basic Medical Sciences of Zhengzhou UniversityZhengzhouChina
| |
Collapse
|
46
|
Yang Y, Liu X, Liu X, Xie C, Shi J. The role of the kynurenine pathway in cardiovascular disease. Front Cardiovasc Med 2024; 11:1406856. [PMID: 38883986 PMCID: PMC11176437 DOI: 10.3389/fcvm.2024.1406856] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 05/23/2024] [Indexed: 06/18/2024] Open
Abstract
The kynurenine pathway (KP) serves as the primary route for tryptophan metabolism in most mammalian organisms, with its downstream metabolites actively involved in various physiological and pathological processes. Indoleamine 2,3-dioxygenase (IDO) and tryptophan 2,3-dioxygenase (TDO) serve as the initial and pivotal enzymes of the KP, with IDO playing important and intricate roles in cardiovascular diseases. Multiple metabolites of KP have been observed to exhibit elevated concentrations in plasma across various cardiovascular diseases, such as atherosclerosis, hypertension, and acute myocardial infarction. Multiple studies have indicated that kynurenine (KYN) may serve as a potential biomarker for several adverse cardiovascular events. Furthermore, Kynurenine and its downstream metabolites have complex roles in inflammation, exhibiting both inhibitory and stimulatory effects on inflammatory responses under different conditions. In atherosclerosis, upregulation of IDO stimulates KYN production, mediating aromatic hydrocarbon receptor (AhR)-induced exacerbation of vascular inflammation and promotion of foam cell formation. Conversely, in arterial calcification, this mediation alleviates osteogenic differentiation of vascular smooth muscle cells. Additionally, in cardiac remodeling, KYN-mediated AhR activation exacerbates pathological left ventricular hypertrophy and fibrosis. Interventions targeting components of the KP, such as IDO inhibitors, 3-hydroxyanthranilic acid, and anthranilic acid, demonstrate cardiovascular protective effects. This review outlines the mechanistic roles of KP in coronary atherosclerosis, arterial calcification, and myocardial diseases, highlighting the potential diagnostic, prognostic, and therapeutic value of KP in cardiovascular diseases, thus providing novel insights for the development and application of related drugs in future research.
Collapse
Affiliation(s)
- Yuehang Yang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xing Liu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinyi Liu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chiyang Xie
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiawei Shi
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
47
|
Gonçalves M, Rodrigues-Santos P, Januário C, Cosentino M, Pereira FC. Indoleamine 2,3-dioxygenase (IDO1) - Can dendritic cells and monocytes expressing this moonlight enzyme change the phase of Parkinson's Disease? Int Immunopharmacol 2024; 133:112062. [PMID: 38652967 DOI: 10.1016/j.intimp.2024.112062] [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: 02/23/2024] [Revised: 03/31/2024] [Accepted: 04/08/2024] [Indexed: 04/25/2024]
Abstract
Parkinson's Disease (PD) is the second most common neurodegenerative disease where central and peripheral immune dysfunctions have been pointed out as a critical component of susceptibility and progression of this disease. Dendritic cells (DCs) and monocytes are key players in promoting immune response regulation and can induce the enzyme indoleamine 2,3-dioxygenase 1 (IDO1) under pro-inflammatory environments. This enzyme with catalytic and signaling activity supports the axis IDO1-KYN-aryl hydrocarbon receptor (AhR), promoting disease-specific immunomodulatory effects. IDO1 is a rate-limiting enzyme of the kynurenine pathway (KP) that begins tryptophan (Trp) catabolism across this pathway. The immune functions of the pathway, which are extensively described in cancer, have been forgotten so far in neurodegenerative diseases, where a chronic inflammatory environment underlines the progression of the disease. Despite dysfunctions of KP have been described in PD, these are mainly associated with neurotoxic functions. With this review, we aim to focus on the immune properties of IDO1+DCs and IDO1+monocytes as a possible strategy to balance the pro-inflammatory profile described in PD. We also highlight the importance of exploring the role of dopaminergic therapeutics in IDO1 modulation to possibly optimize current PD therapeutic strategies.
Collapse
Affiliation(s)
- Milene Gonçalves
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal; Univ Coimbra, Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, Coimbra, Portugal; Univ Coimbra, CIBB - Centre for Innovative Biomedicine and Biotechnology, Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal; University of Coimbra, Institute for Interdisciplinary Research, Doctoral Programme in Experimental Biology and Biomedicine (PDBEB), Portugal
| | - Paulo Rodrigues-Santos
- Univ Coimbra, Institute of Immunology, Faculty of Medicine, Coimbra, Portugal; Univ Coimbra, Center for Neuroscience and Cell Biology, Coimbra, Portugal
| | - Cristina Januário
- Univ Coimbra, CIBIT - Coimbra Institute for Biomedical Imaging and Translational Research, Coimbra, Portugal
| | - Marco Cosentino
- Univ Insubria, Center for Research in Medical Pharmacology, Varese, Italy
| | - Frederico C Pereira
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal; Univ Coimbra, Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, Coimbra, Portugal; Univ Coimbra, CIBB - Centre for Innovative Biomedicine and Biotechnology, Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal.
| |
Collapse
|
48
|
Yue X, Stauff E, Boyapati S, Langhans SA, Xu W, Makrogiannis S, Okorie UJ, Okorie AM, Kandula VVR, Kecskemethy HH, Nikam RM, Averill LW, Shaffer TH. PET Imaging of Neurofibromatosis Type 1 with a Fluorine-18 Labeled Tryptophan Radiotracer. Pharmaceuticals (Basel) 2024; 17:685. [PMID: 38931352 PMCID: PMC11206478 DOI: 10.3390/ph17060685] [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: 04/12/2024] [Revised: 05/14/2024] [Accepted: 05/22/2024] [Indexed: 06/28/2024] Open
Abstract
Neurofibromatosis type 1 (NF1) is a neurocutaneous disorder. Plexiform neurofibromas (PNFs) are benign tumors commonly formed in patients with NF1. PNFs have a high incidence of developing into malignant peripheral nerve sheath tumors (MPNSTs) with a 5-year survival rate of only 30%. Therefore, the accurate diagnosis and differentiation of MPNSTs from benign PNFs are critical to patient management. We studied a fluorine-18 labeled tryptophan positron emission tomography (PET) radiotracer, 1-(2-[18F]fluoroethyl)-L-tryptophan (L-[18F]FETrp), to detect NF1-associated tumors in an animal model. An ex vivo biodistribution study of L-[18F]FETrp showed a similar tracer distribution and kinetics between the wild-type and triple mutant mice with the highest uptake in the pancreas. Bone uptake was stable. Brain uptake was low during the 90-min uptake period. Static PET imaging at 60 min post-injection showed L-[18F]FETrp had a comparable tumor uptake with [1⁸F]fluorodeoxyglucose (FDG). However, L-[18F]FETrp showed a significantly higher tumor-to-brain ratio than FDG (n = 4, p < 0.05). Sixty-minute-long dynamic PET scans using the two radiotracers showed similar kidney, liver, and lung kinetics. A dysregulated tryptophan metabolism in NF1 mice was further confirmed using immunohistostaining. L-[18F]FETrp is warranted to further investigate differentiating malignant NF1 tumors from benign PNFs. The study may reveal the tryptophan-kynurenine pathway as a therapeutic target for treating NF1.
Collapse
Affiliation(s)
- Xuyi Yue
- Department of Radiology, Nemours Children’s Health, Delaware, Wilmington, DE 19803, USA; (E.S.); (S.B.); (W.X.); (V.V.R.K.); (H.H.K.); (R.M.N.); (L.W.A.)
- Diagnostic & Research PET/MR Center, Nemours Children’s Health, Delaware, Wilmington, DE 19803, USA;
| | - Erik Stauff
- Department of Radiology, Nemours Children’s Health, Delaware, Wilmington, DE 19803, USA; (E.S.); (S.B.); (W.X.); (V.V.R.K.); (H.H.K.); (R.M.N.); (L.W.A.)
- Diagnostic & Research PET/MR Center, Nemours Children’s Health, Delaware, Wilmington, DE 19803, USA;
| | - Shriya Boyapati
- Department of Radiology, Nemours Children’s Health, Delaware, Wilmington, DE 19803, USA; (E.S.); (S.B.); (W.X.); (V.V.R.K.); (H.H.K.); (R.M.N.); (L.W.A.)
| | - Sigrid A. Langhans
- Diagnostic & Research PET/MR Center, Nemours Children’s Health, Delaware, Wilmington, DE 19803, USA;
- Division of Neurology, Nemours Children’s Health, Delaware, Wilmington, DE 19803, USA
| | - Wenqi Xu
- Department of Radiology, Nemours Children’s Health, Delaware, Wilmington, DE 19803, USA; (E.S.); (S.B.); (W.X.); (V.V.R.K.); (H.H.K.); (R.M.N.); (L.W.A.)
- Diagnostic & Research PET/MR Center, Nemours Children’s Health, Delaware, Wilmington, DE 19803, USA;
| | - Sokratis Makrogiannis
- Division of Physics, Engineering, Mathematics, and Computer Science, Delaware State University, Dover, DE 19901, USA; (S.M.); (U.J.O.); (A.M.O.)
| | - Uchenna J. Okorie
- Division of Physics, Engineering, Mathematics, and Computer Science, Delaware State University, Dover, DE 19901, USA; (S.M.); (U.J.O.); (A.M.O.)
| | - Azubuike M. Okorie
- Division of Physics, Engineering, Mathematics, and Computer Science, Delaware State University, Dover, DE 19901, USA; (S.M.); (U.J.O.); (A.M.O.)
| | - Vinay V. R. Kandula
- Department of Radiology, Nemours Children’s Health, Delaware, Wilmington, DE 19803, USA; (E.S.); (S.B.); (W.X.); (V.V.R.K.); (H.H.K.); (R.M.N.); (L.W.A.)
| | - Heidi H. Kecskemethy
- Department of Radiology, Nemours Children’s Health, Delaware, Wilmington, DE 19803, USA; (E.S.); (S.B.); (W.X.); (V.V.R.K.); (H.H.K.); (R.M.N.); (L.W.A.)
- Diagnostic & Research PET/MR Center, Nemours Children’s Health, Delaware, Wilmington, DE 19803, USA;
| | - Rahul M. Nikam
- Department of Radiology, Nemours Children’s Health, Delaware, Wilmington, DE 19803, USA; (E.S.); (S.B.); (W.X.); (V.V.R.K.); (H.H.K.); (R.M.N.); (L.W.A.)
- Diagnostic & Research PET/MR Center, Nemours Children’s Health, Delaware, Wilmington, DE 19803, USA;
| | - Lauren W. Averill
- Department of Radiology, Nemours Children’s Health, Delaware, Wilmington, DE 19803, USA; (E.S.); (S.B.); (W.X.); (V.V.R.K.); (H.H.K.); (R.M.N.); (L.W.A.)
- Diagnostic & Research PET/MR Center, Nemours Children’s Health, Delaware, Wilmington, DE 19803, USA;
| | - Thomas H. Shaffer
- Nemours Biomedical Research, Nemours Children’s Health, Delaware, Wilmington, DE 19803, USA;
| |
Collapse
|
49
|
Duan A, Qiu Y, Song B, Tao Y, Wang M, Yin Z, Xie M, Chen Z, Wang Z, Sun X. Metabolome-Wide Mendelian Randomization Assessing the Causal Role of Serum and Cerebrospinal Metabolites in Traumatic Brain Injury. Biomedicines 2024; 12:1178. [PMID: 38927385 PMCID: PMC11201266 DOI: 10.3390/biomedicines12061178] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/10/2024] [Accepted: 05/14/2024] [Indexed: 06/28/2024] Open
Abstract
Previous studies have identified metabolites as biomarkers or potential therapeutic targets for traumatic brain injury (TBI). However, the causal association between them remains unknown. Therefore, we investigated the causal effect of serum metabolites and cerebrospinal fluid (CSF) metabolites on TBI susceptibility through Mendelian randomization (MR). Genetic variants related to metabolites and TBI were extracted from a corresponding genome-wide association study (GWAS). Causal effects were estimated through the inverse variance weighted approach, supplemented by a weighted median, weight mode, and the MR-Egger test. In addition, sensitivity analyses were further performed to evaluate the stability of the MR results, including the MR-Egger intercept, leave-one-out analysis, Cochrane's Q-test, and the MR-PRESSO global test. Metabolic pathway analysis was applied to uncover the underlying pathways of the significant metabolites in TBI. In blood metabolites, substances such as 4-acetaminophen sulfate and kynurenine showed positive links, whereas beta-hydroxyisovalerate and creatinine exhibited negative correlations. CSF metabolites such as N-formylanthranilic acid were positively related, while kynurenate showed negative associations. The metabolic pathway analysis highlighted the potential biological pathways involved in TBI. Of these 16 serum metabolites, 11 CSF metabolites and metabolic pathways may serve as useful circulating biomarkers in clinical screening and prevention, and may be candidate molecules for the exploration of mechanisms and drug targets.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Zhong Wang
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, China; (A.D.); (Y.Q.)
| | - Xiaoou Sun
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, China; (A.D.); (Y.Q.)
| |
Collapse
|
50
|
Eggertsen PP, Palmfeldt J, Pedersen AR, Frederiksen OV, Olsen RKJ, Nielsen JF. Serum neurofilament light chain, inflammatory markers, and kynurenine metabolites in patients with persistent post-concussion symptoms: A cohort study. J Neurol Sci 2024; 460:123016. [PMID: 38636323 DOI: 10.1016/j.jns.2024.123016] [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: 01/22/2024] [Revised: 02/29/2024] [Accepted: 04/10/2024] [Indexed: 04/20/2024]
Abstract
BACKGROUND Concussion leads to persistent post-concussion symptoms (PPCS) in up to one-third of those affected. While previous research has linked the initial trauma to elevated serum levels of neurofilament light chain (NFL), inflammatory markers, and neurotoxic metabolites within the kynurenine pathway, few studies have explored their relevance in PPCS. This study aims to investigate these biomarkers in PPCS patients, elucidating their relevance in the prolonged phase of concussion. METHODS Serum samples from 86 PPCS individuals aged 18-30 years, 2-6 months post-trauma were analyzed, with 54 providing follow-up samples after seven months. NFL was measured using single-molecule array (Simoa) technology, 13 inflammatory markers via a Luminex immunoassay, and five kynurenine metabolites using liquid chromatography-mass spectrometry. A control group of 120 healthy anonymous blood donors was recruited for comparison. RESULTS No significant NFL differences were found in PPCS participants compared with healthy individuals (p = 0.22). Intriguingly, a subset (9.3%) of PPCS participants initially exhibited abnormally high NFL levels (>9.7 pg/mL), which normalized upon follow-up (p = 0.032). Additionally, serum levels of the inflammatory markers, monocyte chemoattractant protein-1 (MCP-1/CCL2), and eotaxin-1/CCL11 were 25-40% lower than in healthy individuals (p ≤ 0.001). As hypothesized, PPCS participants exhibited a 22% reduction in the ratio of kynurenic acid to quinolinic acid (neuroprotective index) (p < 0.0001), indicating a shift towards the formation of neurotoxic metabolites. CONCLUSION NFL may serve as a biomarker to monitor recovery, and future studies should investigate the potential therapeutic benefits of modulating the kynurenine pathway to improve PPCS.
Collapse
Affiliation(s)
- Peter Preben Eggertsen
- Hammel Neurorehabilitation Centre and University Research Clinic, Department of Clinical Medicine, Aarhus University, Voldbyvej 15A, Hammel 8450, Denmark; Research Unit for Molecular Medicine, Department of Clinical Medicine, Aarhus University Hospital and Aarhus University, Palle Juul-Jensens Boulevard 99, Aarhus N 8200, Denmark.
| | - Johan Palmfeldt
- Research Unit for Molecular Medicine, Department of Clinical Medicine, Aarhus University Hospital and Aarhus University, Palle Juul-Jensens Boulevard 99, Aarhus N 8200, Denmark
| | - Asger Roer Pedersen
- University Research Clinic for Innovative Patient Pathways, Diagnostic Centre, Silkeborg Regional Hospital, Falkevej 1, Silkeborg 8600, Denmark
| | | | - Rikke Katrine Jentoft Olsen
- Research Unit for Molecular Medicine, Department of Clinical Medicine, Aarhus University Hospital and Aarhus University, Palle Juul-Jensens Boulevard 99, Aarhus N 8200, Denmark
| | - Jørgen Feldbæk Nielsen
- Hammel Neurorehabilitation Centre and University Research Clinic, Department of Clinical Medicine, Aarhus University, Voldbyvej 15A, Hammel 8450, Denmark
| |
Collapse
|