1
|
Hassan M, Elzallat M, Mohammed DM, Balata M, El-Maadawy WH. Exploiting regulatory T cells (Tregs): Cutting-edge therapy for autoimmune diseases. Int Immunopharmacol 2025; 155:114624. [PMID: 40215774 DOI: 10.1016/j.intimp.2025.114624] [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: 02/01/2025] [Revised: 03/11/2025] [Accepted: 04/05/2025] [Indexed: 04/29/2025]
Abstract
Regulatory T cells (Tregs) are a specialized subset of suppressive T cells that are essential for maintaining self-tolerance, regulating effector T cells, managing microbial infections, preventing tumors, allergies, and autoimmune disorders, and facilitating allograft transplantation. Disruptions in Treg function or abundance contribute to an imbalance between pathogenic and protective immune cells in autoimmune diseases. Recently, one promising treatment strategy to restore immune balance involves the selective expansion or manipulation of Tregs using low-dose IL-2 therapy, adoptive Treg cell transfer, and chimeric antigen receptor (CAR)-Treg approaches. Tregs have been shown in an increasing number of research studies to prevent or even treat a variety of disorders, such as tumors, autoimmune and allergic diseases, transplant rejection, and graft-versus-host disease. A thorough comprehension of Treg function is anticipated to provide clear prospects for effective Treg immunotherapy in the treatment of a wide range of diseases. This review provides an overview of Tregs biology, including their functions, suppressive mechanisms, phenotypic markers, as well as their involvement in disease settings. Furthermore, we discuss the therapeutic potential of different Treg subpopulations and their translational applications in the treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Marwa Hassan
- Immunology Department, Theodor Bilharz Research Institute, Kornaish El Nile, Warrak El Hadar, Imbaba, P.O. 30, Giza 12411, Egypt
| | - Mohamed Elzallat
- Immunology Department, Theodor Bilharz Research Institute, Kornaish El Nile, Warrak El Hadar, Imbaba, P.O. 30, Giza 12411, Egypt
| | - Dina Mostafa Mohammed
- Nutrition and Food Sciences Department, National Research Centre, Dokki, Giza, 12622, Egypt.
| | - Mahmoud Balata
- University hospital bonn. Venusberg-Campus 1, 53127 Bonn, Germany.
| | - Walaa H El-Maadawy
- Pharmacology Department, Theodor Bilharz Research Institute, Kornaish El Nile, Warrak El-Hadar, Imbaba, P.O. 30, Giza, 12411, Egypt
| |
Collapse
|
2
|
Ji Y, Xu M, Zhao H, Cai H, Chen K, Zhang L, Mao H, Wang F, Zhu J, Fang X. Genetic mechanisms underlying gray matter atrophy in Parkinson's disease: a combined transcriptome and neuroimaging study. Cereb Cortex 2025; 35:bhaf097. [PMID: 40302614 DOI: 10.1093/cercor/bhaf097] [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: 02/04/2025] [Revised: 03/13/2025] [Accepted: 04/02/2025] [Indexed: 05/02/2025] Open
Abstract
Extensive studies have demonstrated significant gray matter atrophy in patients with Parkinson's disease (PD); however, the underlying gene expression mechanisms remain largely unknown. To comprehensively characterize the gray matter volume alterations in PD patients, we conducted a neuroimaging meta-analysis and validated the observed atrophic phenotypes in an independent dataset. Leveraging the Allen Human Brain Atlas (AHBA), we linked brain transcriptomic data to neuroimaging phenotypes to identify genes associated with PD-related gray matter atrophy. Further enrichment analyses and functional characterization explored the potential roles of these correlated genes in disease pathology. Both the neuroimaging meta-analysis and independent dataset analysis consistently revealed significant gray matter atrophy in PD, particularly in the superior temporal gyrus, highly associated with sensory and motor functions. Spatial transcriptome-neuroimaging correlation analysis identified 1,952 overlapping genes whose expression levels were significantly correlated with the spatial distribution of gray matter atrophy in PD patients. These genes were enriched in several key biological processes and molecular pathways, exhibiting region- and cell type-specific expression, particularly in dopaminergic receptor neurons of brain tissue. This study delineates the spatial distribution of gray matter atrophy in PD and suggests that this neurodegenerative phenotype may result from complex interactions among multiple functionally relevant genes.
Collapse
Affiliation(s)
- Yi Ji
- Department of Radiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, No. 299, Qingyang Road, Wuxi City 214023, Jiangsu Province, China
- Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, No. 299, Qingyang Road, Wuxi City 214023, Jiangsu Province, China
| | - Min Xu
- Department of Radiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, No. 299, Qingyang Road, Wuxi City 214023, Jiangsu Province, China
- Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, No. 299, Qingyang Road, Wuxi City 214023, Jiangsu Province, China
| | - Han Zhao
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Shushan District, Hefei 230022, China
- Research Center of Clinical Medical Imaging, No. 218, Jixi Road, Shushan District, Hefei 230022, China
- Anhui Provincial Institute of Translational Medicine, No. 218, Jixi Road, Shushan District, Hefei 230022, China
| | - Huanhuan Cai
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Shushan District, Hefei 230022, China
- Research Center of Clinical Medical Imaging, No. 218, Jixi Road, Shushan District, Hefei 230022, China
- Anhui Provincial Institute of Translational Medicine, No. 218, Jixi Road, Shushan District, Hefei 230022, China
| | - Kaidong Chen
- Department of Radiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, No. 299, Qingyang Road, Wuxi City 214023, Jiangsu Province, China
- Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, No. 299, Qingyang Road, Wuxi City 214023, Jiangsu Province, China
| | - Li Zhang
- Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, No. 299, Qingyang Road, Wuxi City 214023, Jiangsu Province, China
- Department of Neurology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, No. 299, Qingyang Road, Wuxi City 214023, Jiangsu Province, China
| | - Haixia Mao
- Department of Radiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, No. 299, Qingyang Road, Wuxi City 214023, Jiangsu Province, China
- Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, No. 299, Qingyang Road, Wuxi City 214023, Jiangsu Province, China
| | - Feng Wang
- Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, No. 299, Qingyang Road, Wuxi City 214023, Jiangsu Province, China
- Department of Neurology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, No. 299, Qingyang Road, Wuxi City 214023, Jiangsu Province, China
| | - Jiajia Zhu
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Shushan District, Hefei 230022, China
- Research Center of Clinical Medical Imaging, No. 218, Jixi Road, Shushan District, Hefei 230022, China
- Anhui Provincial Institute of Translational Medicine, No. 218, Jixi Road, Shushan District, Hefei 230022, China
| | - Xiangming Fang
- Department of Radiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, No. 299, Qingyang Road, Wuxi City 214023, Jiangsu Province, China
- Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, No. 299, Qingyang Road, Wuxi City 214023, Jiangsu Province, China
| |
Collapse
|
3
|
Wang G, Wang Y, Tang X, Li D, Zhao Y, Zhang F. Identification and validation of Atp5f1c in CD4 + T cell as a hub protein in Parkinson's disease. Int J Biol Macromol 2025; 297:139858. [PMID: 39814280 DOI: 10.1016/j.ijbiomac.2025.139858] [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: 11/16/2024] [Revised: 01/12/2025] [Accepted: 01/12/2025] [Indexed: 01/18/2025]
Abstract
Parkinson's disease (PD) is an age-related and progressive neurodegenerative disease. Growing evidences indicate that CD4+ T cell dysfunction plays an essential role in the progress of PD. Here, in LPS-induced PD mice, we isolated midbrain CD4+ T cell and peripheral CD4+ T cell to perform proteomics, and then screened a total of 167 co-expression proteins via integrated bioinformatics analysis. In addition, the subcellular localization, GO analysis, KEGG pathways and protein-protein interaction of 167 co-expression proteins were assessed. Furthermore, GeneMANIA searched the hub proteins and their co-expression genes and found 13 overlapping hub proteins, including Ndufa3, Cox5b, Mrpl21, Ndufab1, Idh3g, Ndufb7, Cyc1, Cisd1, Atp5f1c, Sdhc, Ndufb9, Mtnd1 and Mrpl17. Next, GO analysis and KEGG analysis of the 13 overlapping hub proteins were also exhibited. Further analysis identified that 4 hub proteins (Idh3g, Cisd1, Atp5f1c and Mtnd1) were downregulated both in midbrain and peripheral CD4+ T cell from proteomics. Identification and rescue experiment analysis showed that only Atp5f1c was decreased in LPS- and 6-OHDA-induced PD mice and dopamine (DA) neuronal loss and ATP production decrease were disappeared after Atp5f1c over-expression/Atp5f1c reinfusion both in vivo and in vitro. In conclusion, Atp5f1c was verified as a potential CD4+ T cell-related hub protein for PD.
Collapse
Affiliation(s)
- Guoqing Wang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province and Laboratory Animal Center, Zunyi Medical University, Zunyi, Guizhou, China
| | - Yuanyuan Wang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province and Laboratory Animal Center, Zunyi Medical University, Zunyi, Guizhou, China
| | - Xianjin Tang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province and Laboratory Animal Center, Zunyi Medical University, Zunyi, Guizhou, China
| | - Daidi Li
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province and Laboratory Animal Center, Zunyi Medical University, Zunyi, Guizhou, China
| | - Yujia Zhao
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province and Laboratory Animal Center, Zunyi Medical University, Zunyi, Guizhou, China
| | - Feng Zhang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province and Laboratory Animal Center, Zunyi Medical University, Zunyi, Guizhou, China.
| |
Collapse
|
4
|
Li Q, Han X, Dong M, Bai L, Zhang W, Liu W, Wang F, Zhu X. FDA-Approved Secukinumab Alleviates Glial Activation and Immune Cell Infiltration in MPTP-Induced Mouse Model of Parkinson's Disease. Inflammation 2025:10.1007/s10753-025-02267-8. [PMID: 40011292 DOI: 10.1007/s10753-025-02267-8] [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: 10/23/2024] [Revised: 01/27/2025] [Accepted: 02/05/2025] [Indexed: 02/28/2025]
Abstract
Interleukin-17A (IL-17A) has been implicated in the progression of neurodegenerative diseases such as Alzheimer's disease (AD) and Parkinson's disease (PD). However, the effect of the FDA-approved Secukinumab (SEC), an IL-17A inhibitor, on PD remains unclear. This study aimed to investigate the neuroprotective effect of SEC and its potential mechanisms in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mouse model of PD. Male C57BL/6 J mice were mainly assigned to three groups: Sham, MPTP, and MPTP + SEC. Motor coordination was assessed using the climbing rod and rotarod tests. Dopaminergic neurons (TH +) and glial cells (Iba-1 + , GFAP +) in the substantia nigra were evaluated using immunohistochemistry and immunofluorescence. Flow cytometry was used to analyze immune cell populations in the brain and spleen. Inflammatory cytokines and chemokines were quantified using RT-PCR. SEC treatment significantly alleviated the loss of dopaminergic neurons and improved motor coordination in MPTP mice. It also reduced the infiltration of peripheral immune cells, including CD4 + T cells, NK cells, and monocyte-macrophages into the brain. SEC attenuated glial activation (Iba-1 + , GFAP +) and decreased the expression of pro-inflammatory cytokines and chemokines (CCL2, CXCL9), which recruit immune cells into the brain. These results suggest that Secukinumab protects dopaminergic neurons and attenuates neuroinflammation in MPTP-induced model. SEC treatment in PD might be an effective therapeutic approach for clinical application in the future. HIGHLIGHTS: • Secukinumab reduces the loss of dopaminergic neurons and axons in MPTP mice. • Secukinumab inhibits the infiltration of peripheral immune cells into the brain in MPTP mice. • Secukinumab inhibits the activation of glial cells and reduces neuroinflammation in MPTP mice.
Collapse
Affiliation(s)
- Qi Li
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Anshan Road 154#, Tianjin, 300052, China
| | - Xiaoxuan Han
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Anshan Road 154#, Tianjin, 300052, China
| | - Mengmeng Dong
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Anshan Road 154#, Tianjin, 300052, China
| | - Lipeng Bai
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Anshan Road 154#, Tianjin, 300052, China
| | - Wei Zhang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Anshan Road 154#, Tianjin, 300052, China
| | - Wei Liu
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Anshan Road 154#, Tianjin, 300052, China
| | - Fei Wang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Anshan Road 154#, Tianjin, 300052, China
| | - Xiaodong Zhu
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Anshan Road 154#, Tianjin, 300052, China.
| |
Collapse
|
5
|
Song W, Zhou ZM, Zhang LL, Shu HF, Xia JR, Qin X, Hua R, Zhang YM. Infiltrating peripheral monocyte TREM-1 mediates dopaminergic neuron injury in substantia nigra of Parkinson's disease model mice. Cell Death Dis 2025; 16:18. [PMID: 39809747 PMCID: PMC11733277 DOI: 10.1038/s41419-025-07333-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 11/20/2024] [Accepted: 01/07/2025] [Indexed: 01/16/2025]
Abstract
Neuroinflammation is a key factor in the pathogenesis of Parkinson's disease (PD). Activated microglia in the central nervous system (CNS) and infiltration of peripheral immune cells contribute to dopaminergic neuron loss. However, the role of peripheral immune responses, particularly triggering receptor expressed on myeloid cells-1 (TREM-1), in PD remains unclear. Using a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine hydrochloride (MPTP)-induced PD mouse model, we examined TREM-1 expression and monocyte infiltration in the substantia nigra pars compacta (SNpc). We found that MPTP increased peripheral monocytes, and deletion of peripheral monocytes protected against MPTP neurotoxicity in the SNpc. TREM-1 inhibition, both genetically and pharmacologically, reduced monocyte infiltration, alleviated neuroinflammation, and preserved dopaminergic neurons, resulting in improved motor function. Furthermore, adoptive transfer of TREM-1-expressing monocytes from PD model mice to naive mice induced neuronal damage and motor deficits. These results underscore the critical role of peripheral monocytes and TREM-1 in PD progression, suggesting that targeting TREM-1 could be a promising therapeutic approach to prevent dopaminergic neurodegeneration and motor dysfunction in PD. Schematic diagram of monocyte TREM-1-mediated dopaminergic neuron damage. The figure illustrates that in experimental MPTP-induced PD model mice, the number of inflammatory monocytes in the peripheral blood increases, after which the monocytes infiltrate the CNS through the Blood-Brain Barrier(BBB). These infiltrating monocytes increase the release of inflammatory cytokines and eventually cause neuronal injury. TREM-1 gene deletion and pharmacological blockade limit inflammatory monocyte recruitment into the SNpc and ameliorate neuroinflammatory events and the loss of dopaminergic neurons.
Collapse
Affiliation(s)
- Wei Song
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
- Yancheng Stomatological Hospital, Yancheng, China
| | - Zi-Ming Zhou
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
| | - Le-le Zhang
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
| | - Hai-Feng Shu
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
| | - Jin-Ru Xia
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
| | - Xia Qin
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China
| | - Rong Hua
- Department of Emergency, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.
| | - Yong-Mei Zhang
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou, China.
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, China.
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, China.
| |
Collapse
|
6
|
Zhang Y, Zou M, Wu H, Zhu J, Jin T. The cGAS-STING pathway drives neuroinflammation and neurodegeneration via cellular and molecular mechanisms in neurodegenerative diseases. Neurobiol Dis 2024; 202:106710. [PMID: 39490400 DOI: 10.1016/j.nbd.2024.106710] [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: 07/03/2024] [Revised: 09/27/2024] [Accepted: 10/18/2024] [Indexed: 11/05/2024] Open
Abstract
Neurodegenerative diseases (NDs) are a type of common chronic progressive disorders characterized by progressive damage to specific cell populations in the nervous system, ultimately leading to disability or death. Effective treatments for these diseases are still lacking, due to a limited understanding of their pathogeneses, which involve multiple cellular and molecular pathways. The triggering of an immune response is a common feature in neurodegenerative disorders. A critical challenge is the intricate interplay between neuroinflammation, neurodegeneration, and immune responses, which are not yet fully characterized. In recent years, the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon gene (STING) pathway, a crucial immune response for intracellular DNA sensing, has gradually gained attention. However, the specific roles of this pathway within cellular types such as immune cells, glial and neuronal cells, and its contribution to ND pathogenesis, remain not fully elucidated. In this review, we systematically explore how the cGAS-STING signaling links various cell types with related cellular effector pathways under the context of NDs for multifaceted therapeutic directions. We emphasize the discovery of condition-dependent cellular heterogeneity in the cGAS-STING pathway, which is integral for understanding the diverse cellular responses and potential therapeutic targets. Additionally, we review the pathogenic role of cGAS-STING activation in Parkinson's disease, ataxia-telangiectasia, and amyotrophic lateral sclerosis. We focus on the complex bidirectional roles of the cGAS-STING pathway in Alzheimer's disease, Huntington's disease, and multiple sclerosis, revealing their double-edged nature in disease progression. The objective of this review is to elucidate the pivotal role of the cGAS-STING pathway in ND pathogenesis and catalyze new insights for facilitating the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Yuxin Zhang
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Meijuan Zou
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Hao Wu
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Jie Zhu
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun, China; Department of Neurobiology, Care Sciences & Society, Karolinska Institute, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Tao Jin
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
7
|
Roodveldt C, Bernardino L, Oztop-Cakmak O, Dragic M, Fladmark KE, Ertan S, Aktas B, Pita C, Ciglar L, Garraux G, Williams-Gray C, Pacheco R, Romero-Ramos M. The immune system in Parkinson's disease: what we know so far. Brain 2024; 147:3306-3324. [PMID: 38833182 PMCID: PMC11449148 DOI: 10.1093/brain/awae177] [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: 02/20/2024] [Revised: 05/02/2024] [Accepted: 05/13/2024] [Indexed: 06/06/2024] Open
Abstract
Parkinson's disease is characterized neuropathologically by the degeneration of dopaminergic neurons in the ventral midbrain, the accumulation of α-synuclein (α-syn) aggregates in neurons and chronic neuroinflammation. In the past two decades, in vitro, ex vivo and in vivo studies have consistently shown the involvement of inflammatory responses mediated by microglia and astrocytes, which may be elicited by pathological α-syn or signals from affected neurons and other cell types, and are directly linked to neurodegeneration and disease development. Apart from the prominent immune alterations seen in the CNS, including the infiltration of T cells into the brain, more recent studies have demonstrated important changes in the peripheral immune profile within both the innate and adaptive compartments, particularly involving monocytes, CD4+ and CD8+ T cells. This review aims to integrate the consolidated understanding of immune-related processes underlying the pathogenesis of Parkinson's disease, focusing on both central and peripheral immune cells, neuron-glia crosstalk as well as the central-peripheral immune interaction during the development of Parkinson's disease. Our analysis seeks to provide a comprehensive view of the emerging knowledge of the mechanisms of immunity in Parkinson's disease and the implications of this for better understanding the overall pathogenesis of this disease.
Collapse
Affiliation(s)
- Cintia Roodveldt
- Centre for Molecular Biology and Regenerative Medicine-CABIMER, University of Seville-CSIC, Seville 41092, Spain
- Department of Medical Biochemistry, Molecular Biology and Immunology, Faculty of Medicine, University of Seville, Seville 41009, Spain
| | - Liliana Bernardino
- Health Sciences Research Center (CICS-UBI), Faculty of Health Sciences, University of Beira Interior, 6200-506, Covilhã, Portugal
| | - Ozgur Oztop-Cakmak
- Department of Neurology, Faculty of Medicine, Koç University, Istanbul 34010, Turkey
| | - Milorad Dragic
- Laboratory for Neurobiology, Department of General Physiology and Biophysics, Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia
- Department of Molecular Biology and Endocrinology, ‘VINČA’ Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia
| | - Kari E Fladmark
- Department of Biological Science, University of Bergen, 5006 Bergen, Norway
| | - Sibel Ertan
- Department of Neurology, Faculty of Medicine, Koç University, Istanbul 34010, Turkey
| | - Busra Aktas
- Department of Molecular Biology and Genetics, Burdur Mehmet Akif Ersoy University, Burdur 15200, Turkey
| | - Carlos Pita
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal
| | - Lucia Ciglar
- Center Health & Bioresources, Competence Unit Molecular Diagnostics, AIT Austrian Institute of Technology GmbH, 1210 Vienna, Austria
| | - Gaetan Garraux
- Movere Group, Faculty of Medicine, GIGA Institute, University of Liège, Liège 4000, Belgium
| | | | - Rodrigo Pacheco
- Laboratorio de Neuroinmunología, Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Huechuraba 8580702, Santiago, Chile
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Providencia 7510156, Santiago, Chile
| | - Marina Romero-Ramos
- Department of Biomedicine & The Danish Research Institute of Translational Neuroscience—DANDRITE, Aarhus University, DK-8000 Aarhus C, Denmark
| |
Collapse
|
8
|
Campos J, Osorio-Barrios F, Villanelo F, Gutierrez-Maldonado SE, Vargas P, Pérez-Acle T, Pacheco R. Chemokinergic and Dopaminergic Signalling Collaborates through the Heteromer Formed by CCR9 and Dopamine Receptor D5 Increasing the Migratory Speed of Effector CD4 + T-Cells to Infiltrate the Colonic Mucosa. Int J Mol Sci 2024; 25:10022. [PMID: 39337509 PMCID: PMC11432204 DOI: 10.3390/ijms251810022] [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: 08/12/2024] [Revised: 09/10/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
Inflammatory bowel diseases (IBDs) involve chronic inflammation of the gastrointestinal tract, where effector CD4+ T-cells play a central role. Thereby, the recruitment of T-cells into the colonic mucosa represents a key process in IBD. We recently found that CCR9 and DRD5 might form a heteromeric complex on the T-cell surface. The increase in CCL25 production and the reduction in dopamine levels associated with colonic inflammation represent a dual signal stimulating the CCR9:DRD5 heteromer, which promotes the recruitment of CD4+ T-cells into the colonic lamina propria. Here, we aimed to analyse the molecular requirements involved in the heteromer assembly as well as to determine the underlying cellular mechanisms involved in the colonic tropism given by the stimulation of the CCR9:DRD5 complex. The results show that dual stimulation of the CCR9:DRD5 heteromer potentiates the phosphorylation of the myosin light chain 2 (MLC2) and the migration speed in confined microchannels. Accordingly, disrupting the CCR9:DRD5 assembly induced a sharp reduction in the pMLC2 in vitro, decreased the migratory speed in confined microchannels, and dampened the recruitment of CD4+ T-cells into the inflamed colonic mucosa. Furthermore, in silico analysis confirmed that the interface of interaction of CCR9:DRD5 is formed by the transmembrane segments 5 and 6 from each protomer. Our findings demonstrated that the CCR9:DRD5 heteromeric complex plays a fundamental role in the migration of CD4+ T-cells into the colonic mucosa upon inflammation. Thereby, the present study encourages the design of strategies for disassembling the formation of the CCR9:DRD5 as a therapeutic opportunity to treat IBD.
Collapse
Affiliation(s)
- Javier Campos
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Huechuraba 8580704, Santiago, Chile; (J.C.); (F.V.); (S.E.G.-M.); (T.P.-A.)
| | - Francisco Osorio-Barrios
- Gut Microbiology, Institute for Infectious Diseases, University of Bern, Friedbühlstrasse 25, 3001 Bern, Switzerland
| | - Felipe Villanelo
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Huechuraba 8580704, Santiago, Chile; (J.C.); (F.V.); (S.E.G.-M.); (T.P.-A.)
- Escuela de Ingeniería, Facultad de Ingeniería Arquitectura y Diseño, Universidad San Sebastián, Recoleta 8420524, Santiago, Chile
| | - Sebastian E. Gutierrez-Maldonado
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Huechuraba 8580704, Santiago, Chile; (J.C.); (F.V.); (S.E.G.-M.); (T.P.-A.)
- Escuela de Ingeniería, Facultad de Ingeniería Arquitectura y Diseño, Universidad San Sebastián, Recoleta 8420524, Santiago, Chile
| | - Pablo Vargas
- Institut Curie, PSL Research University, CNRS, UMR144, F-75005 Paris, France;
- Université Paris Cité, CNRS, INSERM, Inserm, INEM, F-75015 Paris, France
| | - Tomás Pérez-Acle
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Huechuraba 8580704, Santiago, Chile; (J.C.); (F.V.); (S.E.G.-M.); (T.P.-A.)
- Escuela de Ingeniería, Facultad de Ingeniería Arquitectura y Diseño, Universidad San Sebastián, Recoleta 8420524, Santiago, Chile
| | - Rodrigo Pacheco
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Huechuraba 8580704, Santiago, Chile; (J.C.); (F.V.); (S.E.G.-M.); (T.P.-A.)
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Providencia 7510157, Santiago, Chile
| |
Collapse
|
9
|
Guevara‐Salinas A, Netzahualcoyotzi C, Álvarez‐Luquín DD, Pérez‐Figueroa E, Sevilla‐Reyes EE, Castellanos‐Barba C, Vega‐Ángeles VT, Terán‐Dávila E, Estudillo E, Velasco I, Adalid‐Peralta L. Treating activated regulatory T cells with pramipexole protects human dopaminergic neurons from 6-OHDA-induced degeneration. CNS Neurosci Ther 2024; 30:e14883. [PMID: 39097919 PMCID: PMC11298200 DOI: 10.1111/cns.14883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 06/17/2024] [Accepted: 07/15/2024] [Indexed: 08/06/2024] Open
Abstract
BACKGROUND Parkinson's disease (PD) is a chronic neurodegenerative disorder characterized by the loss of dopaminergic neurons in the substantia nigra, which promotes a sustained inflammatory environment in the central nervous system. Regulatory T cells (Tregs) play an important role in the control of inflammation and might play a neuroprotective role. Indeed, a decrease in Treg number and function has been reported in PD. In this context, pramipexole, a dopaminergic receptor agonist used to treat PD symptoms, has been shown to increase peripheral levels of Treg cells and improve their suppressive function. The aim of this work was to determine the effect of pramipexole on immunoregulatory Treg cells and its possible neuroprotective effect on human dopaminergic neurons differentiated from human embryonic stem cells. METHODS Treg cells were sorted from white blood cells of healthy human donors. Assays were performed with CD3/CD28-activated and non-activated Treg cells treated with pramipexole at concentrations of 2 or 200 ng/mL. These regulatory cells were co-cultured with in vitro-differentiated human dopaminergic neurons in a cytotoxicity assay with 6-hydroxydopamine (6-OHDA). The role of interleukin-10 (IL-10) was investigated by co-culturing activated IL-10-producing Treg cells with neurons. To further investigate the effect of treatment on Tregs, gene expression in pramipexole-treated, CD3/CD28-activated Treg cells was determined by Fluidigm analysis. RESULTS Pramipexole-treated CD3/CD28-activated Treg cells showed significant protective effects on dopaminergic neurons when challenged with 6-OHDA. Pramipexole-treated activated Treg cells showed neuroprotective capacity through mechanisms involving IL-10 release and the activation of genes associated with regulation and neuroprotection. CONCLUSION Anti-CD3/CD28-activated Treg cells protect dopaminergic neurons against 6-OHDA-induced damage. In addition, activated, IL-10-producing, pramipexole-treated Tregs also induced a neuroprotective effect, and the supernatants of these co-cultures promoted axonal growth. Pramipexole-treated, activated Tregs altered their gene expression in a concentration-dependent manner, and enhanced TGFβ-related dopamine receptor regulation and immune-related pathways. These findings open new perspectives for the development of immunomodulatory therapies for the treatment of PD.
Collapse
Affiliation(s)
- Adrián Guevara‐Salinas
- Laboratorio de Reprogramación CelularInstituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”Mexico CityMexico
| | - Citlalli Netzahualcoyotzi
- Laboratorio de Reprogramación CelularInstituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”Mexico CityMexico
- Instituto de Fisiología Celular – NeurocienciasUniversidad Nacional Autónoma de MéxicoMexico CityMexico
| | - Diana Denisse Álvarez‐Luquín
- Laboratorio de Reprogramación CelularInstituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”Mexico CityMexico
| | - Erandi Pérez‐Figueroa
- Laboratorio de Reprogramación CelularInstituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”Mexico CityMexico
| | - Edgar E. Sevilla‐Reyes
- Centro de Investigación en Enfermedades InfecciosasInstituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”Mexico CityMexico
- Laboratorio de Transcriptómica e Inmunología MolecularInstituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas"Mexico CityMexico
| | - Carlos Castellanos‐Barba
- Laboratorio Nacional de Citometría de FlujoInstituto de Investigaciones Biomédicas UNAMMexico CityMexico
| | - Vera Teresa Vega‐Ángeles
- Laboratorio de Reprogramación CelularInstituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”Mexico CityMexico
| | - Edgar Terán‐Dávila
- Laboratorio de Reprogramación CelularInstituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”Mexico CityMexico
| | - Enrique Estudillo
- Laboratorio de Reprogramación CelularInstituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”Mexico CityMexico
| | - Iván Velasco
- Laboratorio de Reprogramación CelularInstituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”Mexico CityMexico
- Instituto de Fisiología Celular – NeurocienciasUniversidad Nacional Autónoma de MéxicoMexico CityMexico
| | - Laura Adalid‐Peralta
- Laboratorio de Reprogramación CelularInstituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez”Mexico CityMexico
| |
Collapse
|
10
|
Jackson ER, Persson ML, Fish CJ, Findlay IJ, Mueller S, Nazarian J, Hulleman E, van der Lugt J, Duchatel RJ, Dun MD. A review of current therapeutics targeting the mitochondrial protease ClpP in diffuse midline glioma, H3 K27-altered. Neuro Oncol 2024; 26:S136-S154. [PMID: 37589388 PMCID: PMC11066926 DOI: 10.1093/neuonc/noad144] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Indexed: 08/18/2023] Open
Abstract
Diffuse midline gliomas (DMGs) are devastating pediatric brain tumors recognized as the leading cause of cancer-related death in children. DMGs are high-grade gliomas (HGGs) diagnosed along the brain's midline. Euchromatin is the hallmark feature of DMG, caused by global hypomethylation of H3K27 either through point mutations in histone H3 genes (H3K27M), or by overexpression of the enhancer of zeste homolog inhibitory protein. In a clinical trial for adults with progressive HGGs, a 22-year-old patient with a thalamic DMG, H3 K27-altered, showed a remarkable clinical and radiological response to dordaviprone (ONC201). This response in an H3 K27-altered HGG patient, coupled with the lack of response of patients harboring wildtype-H3 tumors, has increased the clinical interest in dordaviprone for the treatment of DMG. Additional reports of clinical benefit have emerged, but research defining mechanisms of action (MOA) fall behind dordaviprone's clinical use, with biomarkers of response unresolved. Here, we summarize dordaviprone's safety, interrogate its preclinical MOA identifying the mitochondrial protease "ClpP" as a biomarker of response, and discuss other ClpP agonists, expanding the arsenal of potential weapons in the fight against DMG. Finally, we discuss combination strategies including ClpP agonists, and their immunomodulatory effects suggestive of a role for the tumor microenvironment in DMG patient response.
Collapse
Affiliation(s)
- Evangeline R Jackson
- Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, New South Wales, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales , Australia
| | - Mika L Persson
- Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, New South Wales, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales , Australia
| | - Cameron J Fish
- Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, New South Wales, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales , Australia
| | - Izac J Findlay
- Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, New South Wales, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales , Australia
| | - Sabine Mueller
- DIPG/DMG Center Zurich, University Children’s Hospital Zürich, Zurich, Switzerland
- Department of Neurology, Neurosurgery and Pediatric, UCSF, San Francisco, California, USA
| | - Javad Nazarian
- DIPG/DMG Center Zurich, University Children’s Hospital Zürich, Zurich, Switzerland
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, District of Columbia, USA
- The George Washington University, School of Medicine and Health Sciences, Washington, District of Columbia, USA
| | - Esther Hulleman
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands, Utrecht, Netherlands
| | - Jasper van der Lugt
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands, Utrecht, Netherlands
| | - Ryan J Duchatel
- Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, New South Wales, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales , Australia
| | - Matthew D Dun
- Cancer Signalling Research Group, School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, New South Wales, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales , Australia
- Paediatric Program, Mark Hughes Foundation Centre for Brain Cancer Research, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, New South Wales, Australia
| |
Collapse
|
11
|
Wang X, Zhang L, Zhou Y, Wang Y, Wang X, Zhang Y, Quan A, Mao Y, Zhang Y, Qi J, Ren Z, Gu L, Yu R, Zhou X. Chronic Stress Exacerbates the Immunosuppressive Microenvironment and Progression of Gliomas by Reducing Secretion of CCL3. Cancer Immunol Res 2024; 12:516-529. [PMID: 38437646 DOI: 10.1158/2326-6066.cir-23-0378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 09/17/2023] [Accepted: 02/28/2024] [Indexed: 03/06/2024]
Abstract
As understanding of cancer has deepened, increasing attention has been turned to the roles of psychological factors, especially chronic stress-induced depression, in the occurrence and development of tumors. However, whether and how depression affects the progression of gliomas are still unclear. In this study, we have revealed that chronic stress inhibited the recruitment of tumor-associated macrophages (TAM) and other immune cells, especially M1-type TAMs and CD8+ T cells, and decreased the level of proinflammatory cytokines in gliomas, leading to an immunosuppressive microenvironment and glioma progression. Mechanistically, by promoting the secretion of stress hormones, chronic stress inhibited the secretion of the chemokine CCL3 and the recruitment of M1-type TAMs in gliomas. Intratumoral administration of CCL3 reprogrammed the immune microenvironment of gliomas and abolished the progression of gliomas induced by chronic stress. Moreover, levels of CCL3 and M1-type TAMs were decreased in the tumor tissues of glioma patients with depression, and CCL3 administration enhanced the antitumor effect of anti-PD-1 therapy in orthotopic models of gliomas undergoing chronic stress. In conclusion, our study has revealed that chronic stress exacerbates the immunosuppressive microenvironment and progression of gliomas by reducing the secretion of CCL3. CCL3 alone or in combination with an anti-PD-1 may be an effective immunotherapy for the treatment of gliomas with depression. See related Spotlight by Cui and Kang, p. 514.
Collapse
Affiliation(s)
- Xu Wang
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Long Zhang
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yi Zhou
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yan Wang
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xiang Wang
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yining Zhang
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ankang Quan
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yufei Mao
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yu Zhang
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Ji Qi
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zhongyu Ren
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Linbo Gu
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Rutong Yu
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xiuping Zhou
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
12
|
Gu R, Pan J, Awan MUN, Sun X, Yan F, Bai L, Bai J. The major histocompatibility complex participates in Parkinson's disease. Pharmacol Res 2024; 203:107168. [PMID: 38583689 DOI: 10.1016/j.phrs.2024.107168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 03/23/2024] [Accepted: 04/02/2024] [Indexed: 04/09/2024]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disease characterized by progressive loss of dopaminergic neurons in the substantia nigra and the aggregation of alpha-synuclein (α-syn). The central nervous system (CNS) has previously been considered as an immune-privileged area. However, studies have shown that the immune responses are involved in PD. The major histocompatibility complex (MHC) presents antigens from antigen-presenting cells (APCs) to T lymphocytes, immune responses will be induced. MHCs are expressed in microglia, astrocytes, and dopaminergic neurons. Single nucleotide polymorphisms in MHC are related to the risk of PD. The aggregated α-syn triggers the expression of MHCs by activating glia cells. CD4+ and CD8+ T lymphocytes responses and microglia activation are detected in brains of PD patients. In addiction immune responses further increase blood-brain barrier (BBB) permeability and T cell infiltration in PD. Thus, MHCs are involved in PD through participating in immune and inflammatory responses.
Collapse
Affiliation(s)
- Rou Gu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China; Medical School, Kunming University of Science and Technology, Kunming 650500, China
| | - Jianyu Pan
- Medical School, Kunming University of Science and Technology, Kunming 650500, China
| | - Maher Un Nisa Awan
- Medical School, Kunming University of Science and Technology, Kunming 650500, China; Department of Neurology, The Affiliated Hospital of Yunnan University, Kunming 650500, China
| | - Xiaowei Sun
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China; Medical School, Kunming University of Science and Technology, Kunming 650500, China
| | - Fang Yan
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China; Medical School, Kunming University of Science and Technology, Kunming 650500, China
| | - Liping Bai
- Medical School, Kunming University of Science and Technology, Kunming 650500, China
| | - Jie Bai
- Medical School, Kunming University of Science and Technology, Kunming 650500, China.
| |
Collapse
|
13
|
Manjarres Z, Calvo M, Pacheco R. Regulation of Pain Perception by Microbiota in Parkinson Disease. Pharmacol Rev 2023; 76:7-36. [PMID: 37863655 DOI: 10.1124/pharmrev.122.000674] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 10/03/2023] [Accepted: 10/10/2023] [Indexed: 10/22/2023] Open
Abstract
Pain perception involves current stimulation in peripheral nociceptive nerves and the subsequent stimulation of postsynaptic excitatory neurons in the spinal cord. Importantly, in chronic pain, the neural activity of both peripheral nociceptors and postsynaptic neurons in the central nervous system is influenced by several inflammatory mediators produced by the immune system. Growing evidence has indicated that the commensal microbiota plays an active role in regulating pain perception by either acting directly on nociceptors or indirectly through the modulation of the inflammatory activity on immune cells. This symbiotic relationship is mediated by soluble bacterial mediators or intrinsic structural components of bacteria that act on eukaryotic cells, including neurons, microglia, astrocytes, macrophages, T cells, enterochromaffin cells, and enteric glial cells. The molecular mechanisms involve bacterial molecules that act directly on neurons, affecting their excitability, or indirectly on non-neuronal cells, inducing changes in the production of proinflammatory or anti-inflammatory mediators. Importantly, Parkinson disease, a neurodegenerative and inflammatory disorder that affects mainly the dopaminergic neurons implicated in the control of voluntary movements, involves not only a motor decline but also nonmotor symptomatology, including chronic pain. Of note, several recent studies have shown that Parkinson disease involves a dysbiosis in the composition of the gut microbiota. In this review, we first summarize, integrate, and classify the molecular mechanisms implicated in the microbiota-mediated regulation of chronic pain. Second, we analyze the changes on the commensal microbiota associated to Parkinson disease and propose how these changes affect the development of chronic pain in this pathology. SIGNIFICANCE STATEMENT: The microbiota regulates chronic pain through the action of bacterial signals into two main locations: the peripheral nociceptors and the postsynaptic excitatory neurons in the spinal cord. The dysbiosis associated to Parkinson disease reveals increased representation of commensals that potentially exacerbate chronic pain and reduced levels of bacteria with beneficial effects on pain. This review encourages further research to better understand the signals involved in bacteria-bacteria and bacteria-host communication to get the clues for the development of probiotics with therapeutic potential.
Collapse
Affiliation(s)
- Zulmary Manjarres
- Laboratorio de Neuroinmunología, Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Santiago, Chile (Z.M., R.P.); Facultad de Ciencias Biológicas (Z.M., M.C.) and División de Anestesiología, Escuela de Medicina (M.C.), Pontificia Universidad Católica de Chile, Santiago, Chile; Millennium Nucleus for the Study of Pain, Santiago, Chile (Z.M., M.C.); and Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile (R.P.)
| | - Margarita Calvo
- Laboratorio de Neuroinmunología, Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Santiago, Chile (Z.M., R.P.); Facultad de Ciencias Biológicas (Z.M., M.C.) and División de Anestesiología, Escuela de Medicina (M.C.), Pontificia Universidad Católica de Chile, Santiago, Chile; Millennium Nucleus for the Study of Pain, Santiago, Chile (Z.M., M.C.); and Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile (R.P.)
| | - Rodrigo Pacheco
- Laboratorio de Neuroinmunología, Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Santiago, Chile (Z.M., R.P.); Facultad de Ciencias Biológicas (Z.M., M.C.) and División de Anestesiología, Escuela de Medicina (M.C.), Pontificia Universidad Católica de Chile, Santiago, Chile; Millennium Nucleus for the Study of Pain, Santiago, Chile (Z.M., M.C.); and Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile (R.P.)
| |
Collapse
|
14
|
Gustavsson J, Johansson J, Falahati F, Andersson M, Papenberg G, Avelar-Pereira B, Bäckman L, Kalpouzos G, Salami A. The iron-dopamine D1 coupling modulates neural signatures of working memory across adult lifespan. Neuroimage 2023; 279:120323. [PMID: 37582419 DOI: 10.1016/j.neuroimage.2023.120323] [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: 03/07/2023] [Revised: 08/09/2023] [Accepted: 08/10/2023] [Indexed: 08/17/2023] Open
Abstract
Brain iron overload and decreased integrity of the dopaminergic system have been independently reported as brain substrates of cognitive decline in aging. Dopamine (DA), and iron are co-localized in high concentrations in the striatum and prefrontal cortex (PFC), but follow opposing age-related trajectories across the lifespan. DA contributes to cellular iron homeostasis and the activation of D1-like DA receptors (D1DR) alleviates oxidative stress-induced inflammatory responses, suggesting a mutual interaction between these two fundamental components. Still, a direct in-vivo study testing the iron-D1DR relationship and their interactions on brain function and cognition across the lifespan is rare. Using PET and MRI data from the DyNAMiC study (n=180, age=20-79, %50 female), we showed that elevated iron content was related to lower D1DRs in DLPFC, but not in striatum, suggesting that dopamine-rich regions are less susceptible to elevated iron. Critically, older individuals with elevated iron and lower D1DR exhibited less frontoparietal activations during the most demanding task, which in turn was related to poorer working-memory performance. Together, our findings suggest that the combination of elevated iron load and reduced D1DR contribute to disturbed PFC-related circuits in older age, and thus may be targeted as two modifiable factors for future intervention.
Collapse
Affiliation(s)
- Jonatan Gustavsson
- Aging Research Center, Karolinska Institutet and Stockholm University, Sweden.
| | - Jarkko Johansson
- Faculty of Medicine, Department of Radiation Sciences, Umeå University, Sweden; Umeå Center for Functional Brain Imaging, Umeå University, Umeå, Sweden
| | - Farshad Falahati
- Aging Research Center, Karolinska Institutet and Stockholm University, Sweden
| | - Micael Andersson
- Umeå Center for Functional Brain Imaging, Umeå University, Umeå, Sweden; Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | - Goran Papenberg
- Aging Research Center, Karolinska Institutet and Stockholm University, Sweden
| | - Bárbara Avelar-Pereira
- Aging Research Center, Karolinska Institutet and Stockholm University, Sweden; Department of Psychiatry and Behavioural Sciences, School of Medicine, Stanford University, Stanford, California 94304, USA
| | - Lars Bäckman
- Aging Research Center, Karolinska Institutet and Stockholm University, Sweden
| | - Grégoria Kalpouzos
- Aging Research Center, Karolinska Institutet and Stockholm University, Sweden
| | - Alireza Salami
- Aging Research Center, Karolinska Institutet and Stockholm University, Sweden; Umeå Center for Functional Brain Imaging, Umeå University, Umeå, Sweden; Department of Integrative Medical Biology, Umeå University, Umeå, Sweden; Wallenberg Center for Molecular Medicine, Umeå University, Sweden
| |
Collapse
|
15
|
Xia X, Li H, Xu X, Zhao G, Du M. Facilitating Pro-survival Mitophagy for Alleviating Parkinson's Disease via Sequence-Targeted Lycopene Nanodots. ACS NANO 2023; 17:17979-17995. [PMID: 37714739 DOI: 10.1021/acsnano.3c04308] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/17/2023]
Abstract
The pathogenesis of Parkinson's disease is closely linked to impaired mitochondrial function and abnormal mitophagy. Biocompatible natural antioxidants effectively protect dopaminergic neurons. However, the main challenge in using natural antioxidants for Parkinson's disease therapy is creating a delivery platform to achieve neuron-targeted enrichment. Herein, we synthesized rationally sequence-targeted lycopene nanodots using recombinant human H-ferritin nanocages with lycopene loading into the cavity and lipophilic triphenylphosphonium (TPP) coupling on the outer surface. The nanodots allow for the neural enrichment and mitochondrial regulation of lycopene through blood-brain barrier transcytosis and neuronal mitochondria-targeting capability. These anti-ROS nanodots protect neuronal mitochondrial function and promote PINK1/Parkin-mediated mitophagy in MPTP toxicity-induced neurons in vivo and in vitro, which favors the secretory efflux of pathogenic α-synuclein and the survival of dopaminergic neurons. Moreover, these nanodots restore the Parkinson-like motor symptoms in Parkinson's model mice. This noninvasive sequence-targeted delivery strategy with excellent biocompatibility for pro-survival mitophagy-mediated pathology alleviation makes it a promising approach for treating and preventing Parkinson's disease.
Collapse
Affiliation(s)
- Xiaoyu Xia
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, China
- Collaborative Innovation Centre of Provincial and Ministerial Co-construction for Seafood Deep Processing, Dalian 116034, China
| | - Han Li
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, China
- Collaborative Innovation Centre of Provincial and Ministerial Co-construction for Seafood Deep Processing, Dalian 116034, China
| | - Xianbing Xu
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, China
- Collaborative Innovation Centre of Provincial and Ministerial Co-construction for Seafood Deep Processing, Dalian 116034, China
| | - Guanghua Zhao
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Ming Du
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, China
- Collaborative Innovation Centre of Provincial and Ministerial Co-construction for Seafood Deep Processing, Dalian 116034, China
| |
Collapse
|
16
|
Załuski M, Karcz T, Drabczyńska A, Vielmuth C, Olejarz-Maciej A, Głuch-Lutwin M, Mordyl B, Siwek A, Satała G, Müller CE, Kieć-Kononowicz K. Xanthine-Dopamine Hybrid Molecules as Multitarget Drugs with Potential for the Treatment of Neurodegenerative Diseases. Biomolecules 2023; 13:1079. [PMID: 37509114 PMCID: PMC10377586 DOI: 10.3390/biom13071079] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/28/2023] [Accepted: 06/30/2023] [Indexed: 07/30/2023] Open
Abstract
Multitarget drugs based on a hybrid dopamine-xanthine core were designed as potential drug candidates for the treatment of neurodegenerative diseases. Monoamine oxidase B (MAO-B) inhibitors with significant ancillary A2A adenosine receptor (A2AAR) antagonistic properties were further developed to exhibit additional phosphodiesterase-4 and -10 (PDE4/10) inhibition and/or dopamine D2 receptor (D2R) agonistic activity. While all of the designed compounds showed MAO-B inhibition in the nanomolar range mostly combined with submicromolar A2AAR affinity, significant enhancement of PDE-inhibitory and D2R-agonistic activity was additionally reached for some compounds through various structural modifications. The final multitarget drugs also showed promising antioxidant properties in vitro. In order to evaluate their potential neuroprotective effect, representative ligands were tested in a cellular model of toxin-induced neurotoxicity. As a result, protective effects against oxidative stress in neuroblastoma cells were observed, confirming the utility of the applied strategy. Further evaluation of the newly developed multitarget ligands in preclinical models of Alzheimer's and Parkinson's diseases is warranted.
Collapse
Affiliation(s)
- Michał Załuski
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, 30-688 Krakow, Poland
| | - Tadeusz Karcz
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, 30-688 Krakow, Poland
| | - Anna Drabczyńska
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, 30-688 Krakow, Poland
| | - Christin Vielmuth
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, D-53121 Bonn, Germany
| | - Agnieszka Olejarz-Maciej
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, 30-688 Krakow, Poland
| | - Monika Głuch-Lutwin
- Department of Pharmacobiology, Faculty of Pharmacy, Jagiellonian University Medical College, 30-688 Krakow, Poland
| | - Barbara Mordyl
- Department of Pharmacobiology, Faculty of Pharmacy, Jagiellonian University Medical College, 30-688 Krakow, Poland
| | - Agata Siwek
- Department of Pharmacobiology, Faculty of Pharmacy, Jagiellonian University Medical College, 30-688 Krakow, Poland
| | - Grzegorz Satała
- Department of Medicinal Chemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Krakow, Poland
| | - Christa E Müller
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, D-53121 Bonn, Germany
| | - Katarzyna Kieć-Kononowicz
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, 30-688 Krakow, Poland
| |
Collapse
|
17
|
Wang XQ, Cai HH, Deng QW, Chang YZ, Peng YP, Qiu YH. Dopamine D2 receptor on CD4 + T cells is protective against inflammatory responses and signs in a mouse model of rheumatoid arthritis. Arthritis Res Ther 2023; 25:87. [PMID: 37237413 DOI: 10.1186/s13075-023-03071-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 05/20/2023] [Indexed: 05/28/2023] Open
Abstract
BACKGROUND Dopamine is a neurotransmitter and has been found to regulate lymphocytes by acting on dopamine receptors (DRs). CD4+ T cells express all the five subtypes of DRs, D1R to D5R. Although CD4+ T cells have been involved in pathogenesis of rheumatoid arthritis (RA), roles of DRs expressed on these cells in RA are poorly understood. This study determined whether D2R expressed on CD4+ T cells regulates inflammatory responses and signs in collagen type II (CII)-induced arthritis (CIA), a mouse model of RA. METHODS DBA/1 mice and C57BL/6 mice with global D1r or D2r deficiency (D1r-/- or D2r-/-) or CD4+ T cell-specific D2r deletion (D2rfl/fl/CD4Cre) were used to prepare CIA model by intradermal injection of CII. D2R agonist sumanirole was intraperitoneally administered in CIA mice. CD4+ T cells obtained from CIA mice were exposed to sumanirole or/and D2R antagonist L-741,626 in vitro. Arthritic symptoms were assessed by clinical arthritis scores. Flow cytometric assay measured frequencies of CD4+ T cell subsets (Th1, Th2, Th17 and Treg cells). Expression of specific transcription factors for the CD4+ T cell subsets was tested by Western blot. Cytokine production was estimated by quantitative PCR and ELISA. RESULTS CIA mice manifested a bias of CD4+ T cells towards Th1 and Th17 cells. D2r-/- CIA mice showed a stronger bias towards Th1 and Th17 phenotypes than CIA mice, while D1r-/- CIA mice did not show the changes. CD4+ T cell-specific D2r deletion exacerbated both the polarization towards Th1 and Th17 cells and the symptoms of arthritis. Sumanirole administration in CIA mice ameliorated the bias of CD4+ T cells towards Th1 and Th17 phenotypes as well as arthritic symptoms. Sumanirole treatment of in vitro CD4+ T cells obtained from CIA mice promoted the shift to Treg cells, and the effect of sumanirole was blocked by L-741,626. CONCLUSIONS D2R expressed on CD4+ T cells is protective against imbalance between pro-inflammatory and anti-inflammatory T cells and arthritic symptoms in CIA.
Collapse
Affiliation(s)
- Xiao-Qin Wang
- Department of Physiology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong, 226001, China
| | - Huan-Huan Cai
- Department of Physiology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong, 226001, China
| | - Qiao-Wen Deng
- Department of Physiology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong, 226001, China
| | - Ya-Zhou Chang
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Yu-Ping Peng
- Department of Physiology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong, 226001, China.
| | - Yi-Hua Qiu
- Department of Physiology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong, 226001, China.
| |
Collapse
|
18
|
Channer B, Matt SM, Nickoloff-Bybel EA, Pappa V, Agarwal Y, Wickman J, Gaskill PJ. Dopamine, Immunity, and Disease. Pharmacol Rev 2023; 75:62-158. [PMID: 36757901 PMCID: PMC9832385 DOI: 10.1124/pharmrev.122.000618] [Citation(s) in RCA: 103] [Impact Index Per Article: 51.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 08/02/2022] [Accepted: 08/04/2022] [Indexed: 12/14/2022] Open
Abstract
The neurotransmitter dopamine is a key factor in central nervous system (CNS) function, regulating many processes including reward, movement, and cognition. Dopamine also regulates critical functions in peripheral organs, such as blood pressure, renal activity, and intestinal motility. Beyond these functions, a growing body of evidence indicates that dopamine is an important immunoregulatory factor. Most types of immune cells express dopamine receptors and other dopaminergic proteins, and many immune cells take up, produce, store, and/or release dopamine, suggesting that dopaminergic immunomodulation is important for immune function. Targeting these pathways could be a promising avenue for the treatment of inflammation and disease, but despite increasing research in this area, data on the specific effects of dopamine on many immune cells and disease processes remain inconsistent and poorly understood. Therefore, this review integrates the current knowledge of the role of dopamine in immune cell function and inflammatory signaling across systems. We also discuss the current understanding of dopaminergic regulation of immune signaling in the CNS and peripheral tissues, highlighting the role of dopaminergic immunomodulation in diseases such as Parkinson's disease, several neuropsychiatric conditions, neurologic human immunodeficiency virus, inflammatory bowel disease, rheumatoid arthritis, and others. Careful consideration is given to the influence of experimental design on results, and we note a number of areas in need of further research. Overall, this review integrates our knowledge of dopaminergic immunology at the cellular, tissue, and disease level and prompts the development of therapeutics and strategies targeted toward ameliorating disease through dopaminergic regulation of immunity. SIGNIFICANCE STATEMENT: Canonically, dopamine is recognized as a neurotransmitter involved in the regulation of movement, cognition, and reward. However, dopamine also acts as an immune modulator in the central nervous system and periphery. This review comprehensively assesses the current knowledge of dopaminergic immunomodulation and the role of dopamine in disease pathogenesis at the cellular and tissue level. This will provide broad access to this information across fields, identify areas in need of further investigation, and drive the development of dopaminergic therapeutic strategies.
Collapse
Affiliation(s)
- Breana Channer
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Stephanie M Matt
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Emily A Nickoloff-Bybel
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Vasiliki Pappa
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Yash Agarwal
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Jason Wickman
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Peter J Gaskill
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| |
Collapse
|
19
|
Mechanisms of Autoimmune Cell in DA Neuron Apoptosis of Parkinson's Disease: Recent Advancement. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7965433. [PMID: 36567855 PMCID: PMC9771667 DOI: 10.1155/2022/7965433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 11/19/2022] [Accepted: 11/21/2022] [Indexed: 12/23/2022]
Abstract
Parkinson's disease (PD) is a prevalent neurodegenerative disorder that manifests as motor and nonmotor symptoms due to the selective loss of midbrain DArgic (DA) neurons. More and more studies have shown that pathological reactions initiated by autoimmune cells play an essential role in the progression of PD. Autoimmune cells exist in the brain parenchyma, cerebrospinal fluid, and meninges; they are considered inducers of neuroinflammation and regulate the immune in the human brain in PD. For example, T cells can recognize α-synuclein presented by antigen-presenting cells to promote neuroinflammation. In addition, B cells will accelerate the apoptosis of DA neurons in the case of PD-related gene mutations. Activation of microglia and damage of DA neurons even form the self-degeneration cycle to deteriorate PD. Numerous autoimmune cells have been considered regulators of apoptosis, α-synuclein misfolding and aggregation, mitochondrial dysfunction, autophagy, and neuroinflammation of DA neurons in PD. The evidence is mounting that autoimmune cells promote DA neuron apoptosis. In this review, we discuss the current knowledge regarding the regulation and function of B cell, T cell, and microglia as well as NK cell in PD pathogenesis, focusing on DA neuron apoptosis to understand the disease better and propose potential target identification for the treatment in the early stages of PD. However, there are still some limitations in our work, for example, the specific mechanism of PD progression caused by autoimmune cells in mitochondrial dysfunction, ferroptosis, and autophagy has not been clarified in detail, which needs to be summarized in further work.
Collapse
|
20
|
Borowicz-Reutt K, Banach M. Trimetazidine, an Anti-Ischemic Drug, Reduces the Antielectroshock Effects of Certain First-Generation Antiepileptic Drugs. Int J Mol Sci 2022; 23:ijms231911328. [PMID: 36232629 PMCID: PMC9570019 DOI: 10.3390/ijms231911328] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 09/14/2022] [Accepted: 09/21/2022] [Indexed: 11/16/2022] Open
Abstract
Trimetazidine (TMZ), an anti-ischemic drug for improving cellular metabolism, is mostly administered to patients with poorly controlled ischemic heart disease (IHD). Since IHD is considered the most frequent causative factor of cardiac arrhythmias, and these often coexist with seizure disorders, we decided to investigate the effect of TMZ in the electroconvulsive threshold test (ECT) and its influence on the action of four first-generation antiepileptic drugs in the maximal electroshock test (MES) in mice. The TMZ (up to 120 mg/kg) did not affect the ECT, but applied at doses of 20–120 mg/kg it decreased the antielectroshock action of phenobarbital. The TMZ (50–120 mg/kg) reduced the effect of phenytoin, and, when administered at a dose of 120 mg/kg, it diminished the action of carbamazepine. All of these revealed interactions seem to be pharmacodynamic, since the TMZ did not affect the brain levels of antiepileptic drugs. Furthermore, the combination of TMZ with valproate (but not with other antiepileptic drugs) significantly impaired motor coordination, evaluated using the chimney test. Long-term memory, assessed with a passive-avoidance task, was not affected by either the TMZ or its combinations with antiepileptic drugs. The obtained results suggest that TMZ may not be beneficial as an add-on therapy in patients with IHD and epilepsy.
Collapse
|
21
|
Contaldi E, Magistrelli L, Cosentino M, Marino F, Comi C. Lymphocyte Count and Neutrophil-to-Lymphocyte Ratio Are Associated with Mild Cognitive Impairment in Parkinson’s Disease: A Single-Center Longitudinal Study. J Clin Med 2022; 11:jcm11195543. [PMID: 36233411 PMCID: PMC9571051 DOI: 10.3390/jcm11195543] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/13/2022] [Accepted: 09/20/2022] [Indexed: 11/24/2022] Open
Abstract
Lymphocyte count and neutrophil-to-lymphocyte ratio (NLR) may represent useful biomarkers of Parkinson’s disease (PD), but their role in PD-related mild cognitive impairment (MCI) has not been fully elucidated. The present study aimed to confirm whether these immunological measures can discriminate PD patients from healthy controls (HC) and establish their feasibility as prognostic biomarkers of MCI in PD. Immunological data at baseline were analyzed in 58 drug-naïve PD patients and 58 HC matched 1:1 for age, sex, and cardiovascular comorbidities. We selected a subgroup of 51 patients from this initial cohort who underwent longitudinal neuropsychological assessments through the Addenbrooke’s Cognitive Examination Revised (ACE-R) test. We considered the last examination available to analyze the relationship between ACE-R test scores and immunological measures. We found that lymphocyte count was lower and NLR higher in PD than HC (p = 0.006, p = 0.044), with AUC = 0.649 and 0.608, respectively. Secondly, in PD-MCI there were significantly higher levels of circulating lymphocytes (p = 0.002) and lower NLR (p = 0.020) than PD with normal cognitive status (PD-NC). Correlations between lymphocyte count and ACE-R total score and memory subitem (r = −0.382, p = 0.006; r = −0.362, p = 0.01), as well as between NLR and ACE-R total score and memory subitem (r = 0.325, p = 0.02; r = 0.374, p = 0.007), were also found. ROC curve analysis showed that lymphocyte count and NLR displayed acceptable discrimination power of PD-MCI with AUC = 0.759 and 0.691, respectively. In conclusion, we suggest that an altered peripheral immune phenotype could foster cognitive decline development in PD, thus opening the possibility of immune-targeting strategies to tackle this disabling non-motor feature.
Collapse
Affiliation(s)
- Elena Contaldi
- Movement Disorders Centre, Neurology Unit, Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy
- PhD Program in Medical Sciences and Biotechnology, University of Piemonte Orientale, 28100 Novara, Italy
| | - Luca Magistrelli
- Movement Disorders Centre, Neurology Unit, Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy
- PhD Program in Clinical and Experimental Medicine and Medical Humanities, University of Insubria, 21100 Varese, Italy
| | - Marco Cosentino
- Center of Research in Medical Pharmacology, University of Insubria, 21100 Varese, Italy
- Center for Research in Neuroscience, University of Insubria, 21100 Varese, Italy
| | - Franca Marino
- Center of Research in Medical Pharmacology, University of Insubria, 21100 Varese, Italy
- Center for Research in Neuroscience, University of Insubria, 21100 Varese, Italy
| | - Cristoforo Comi
- Center of Research in Medical Pharmacology, University of Insubria, 21100 Varese, Italy
- Department of Translational Medicine, Neurology Unit, S. Andrea Hospital, University of Piemonte Orientale, 13100 Vercelli, Italy
- Correspondence:
| |
Collapse
|
22
|
Liu W, Fan M, Lu W, Zhu W, Meng L, Lu S. Emerging Roles of T Helper Cells in Non-Infectious Neuroinflammation: Savior or Sinner. Front Immunol 2022; 13:872167. [PMID: 35844577 PMCID: PMC9280647 DOI: 10.3389/fimmu.2022.872167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 05/30/2022] [Indexed: 12/03/2022] Open
Abstract
CD4+ T cells, also known as T helper (Th) cells, contribute to the adaptive immunity both in the periphery and in the central nervous system (CNS). At least seven subsets of Th cells along with their signature cytokines have been identified nowadays. Neuroinflammation denotes the brain’s immune response to inflammatory conditions. In recent years, various CNS disorders have been related to the dysregulation of adaptive immunity, especially the process concerning Th cells and their cytokines. However, as the functions of Th cells are being discovered, it’s also found that their roles in different neuroinflammatory conditions, or even the participation of a specific Th subset in one CNS disorder may differ, and sometimes contrast. Based on those recent and contradictory evidence, the conflicting roles of Th cells in multiple sclerosis, Alzheimer’s disease, Parkinson’s disease, epilepsy, traumatic brain injury as well as some typical mental disorders will be reviewed herein. Research progress, limitations and novel approaches concerning different neuroinflammatory conditions will also be mentioned and compared.
Collapse
Affiliation(s)
- Wenbin Liu
- Institute of Molecular and Translational Medicine, and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
- Department of Neurosurgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Meiyang Fan
- Institute of Molecular and Translational Medicine, and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Wen Lu
- Department of Psychiatry, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Wenhua Zhu
- Institute of Molecular and Translational Medicine, and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
- National Joint Engineering Research Center of Biodiagnostics and Biotherapy, Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
- *Correspondence: Wenhua Zhu, ; Liesu Meng,
| | - Liesu Meng
- Institute of Molecular and Translational Medicine, and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
- National Joint Engineering Research Center of Biodiagnostics and Biotherapy, Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
- Key Laboratory of Environment and Genes Related to Diseases (Xi’an Jiaotong University), Ministry of Education, Xi’an, China
- *Correspondence: Wenhua Zhu, ; Liesu Meng,
| | - Shemin Lu
- Institute of Molecular and Translational Medicine, and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
- National Joint Engineering Research Center of Biodiagnostics and Biotherapy, Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
- Key Laboratory of Environment and Genes Related to Diseases (Xi’an Jiaotong University), Ministry of Education, Xi’an, China
| |
Collapse
|
23
|
Russo T, Riessland M. Age-Related Midbrain Inflammation and Senescence in Parkinson’s Disease. Front Aging Neurosci 2022; 14:917797. [PMID: 35721008 PMCID: PMC9204626 DOI: 10.3389/fnagi.2022.917797] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 05/04/2022] [Indexed: 11/13/2022] Open
Abstract
Immune responses are arising as a common feature of several neurodegenerative diseases, such as Parkinson’s disease (PD), Alzheimer’s disease (AD), and Amyotrophic Lateral Sclerosis (ALS), but their role as either causative or consequential remains debated. It is evident that there is local inflammation in the midbrain in PD patients even before symptom onset, but the underlying mechanisms remain elusive. In this mini-review, we discuss this midbrain inflammation in the context of PD and argue that cellular senescence may be the cause for this immune response. We postulate that to unravel the relationship between inflammation and senescence in PD, it is crucial to first understand the potential causative roles of various cell types of the midbrain and determine how the possible paracrine spreading of senescence between them may lead to observed local immune responses. We hypothesize that secretion of pro-inflammatory factors by senescent cells in the midbrain triggers neuroinflammation resulting in immune cell-mediated killing of midbrain dopaminergic (DA) neurons in PD.
Collapse
Affiliation(s)
- Taylor Russo
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, United States
- Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY, United States
| | - Markus Riessland
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, United States
- Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY, United States
- *Correspondence: Markus Riessland,
| |
Collapse
|
24
|
Gustavsson J, Papenberg G, Falahati F, Laukka EJ, Kalpouzos G. Contributions of the Catechol-O-Methyltransferase Val158Met Polymorphism to Changes in Brain Iron Across Adulthood and Their Relationships to Working Memory. Front Hum Neurosci 2022; 16:838228. [PMID: 35571998 PMCID: PMC9091601 DOI: 10.3389/fnhum.2022.838228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 03/16/2022] [Indexed: 11/13/2022] Open
Abstract
Ageing is associated with excessive free brain iron, which may induce oxidative stress and neuroinflammation, likely causing cognitive deficits. Lack of dopamine may be a factor behind the increase of iron with advancing age, as it has an important role in cellular iron homoeostasis. We investigated the effect of COMT Val 158 Met (rs4680), a polymorphism crucial for dopamine degradation and proxy for endogenous dopamine, on iron accumulation and working memory in a longitudinal lifespan sample (n = 208, age 20–79 at baseline, mean follow-up time = 2.75 years) using structural equation modelling. Approximation of iron content was assessed using quantitative susceptibility mapping in striatum and dorsolateral prefrontal cortex (DLPFC). Iron accumulated in both striatum and DLPFC during the follow-up period. Greater iron accumulation in DLPFC was associated with more deleterious change in working memory. Older (age 50–79) Val homozygotes (with presumably lower endogenous dopamine) accumulated more iron than older Met carriers in both striatum and DLPFC, no such differences were observed among younger adults (age 20–49). In conclusion, individual differences in genetic predisposition related to low dopamine levels increase iron accumulation, which in turn may trigger deleterious change in working memory. Future studies are needed to better understand how dopamine may modulate iron accumulation across the human lifespan.
Collapse
Affiliation(s)
- Jonatan Gustavsson
- Aging Research Center, Karolinska Institutet and Stockholm University, Stockholm, Sweden
- *Correspondence: Jonatan Gustavsson,
| | - Goran Papenberg
- Aging Research Center, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| | - Farshad Falahati
- Aging Research Center, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| | - Erika J. Laukka
- Aging Research Center, Karolinska Institutet and Stockholm University, Stockholm, Sweden
- Stockholm Gerontology Research Center, Stockholm, Sweden
| | - Grégoria Kalpouzos
- Aging Research Center, Karolinska Institutet and Stockholm University, Stockholm, Sweden
- Grégoria Kalpouzos,
| |
Collapse
|
25
|
Fu J, Huang Y, Bao T, Liu C, Liu X, Chen X. The role of Th17 cells/IL-17A in AD, PD, ALS and the strategic therapy targeting on IL-17A. J Neuroinflammation 2022; 19:98. [PMID: 35459141 PMCID: PMC9034482 DOI: 10.1186/s12974-022-02446-6] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Accepted: 03/29/2022] [Indexed: 02/08/2023] Open
Abstract
Neurodegenerative diseases are a group of disorders characterized by progressive loss of certain populations of neurons, which eventually lead to dysfunction. These diseases include Alzheimer’s disease (AD), Parkinson’s disease (PD), and amyotrophic lateral sclerosis (ALS). Immune pathway dysregulation is one of the common features of neurodegeneration. Recently, there is growing interest in the specific role of T helper Th 17 cells and Interleukin-17A (IL-17A), the most important cytokine of Th 17 cells, in the pathogenesis of the central nervous system (CNS) of neurodegenerative diseases. In the present study, we summarized current knowledge about the function of Th17/IL-17A, the physiology of Th17/IL-17A in diseases, and the contribution of Th17/IL-17A in AD, PD, and ALS. We also update the findings on IL-17A-targeting drugs as potentially immunomodulatory therapeutic agents for neurodegenerative diseases. Although the specific mechanism of Th17/IL-17A in this group of diseases is still controversial, uncovering the molecular pathways of Th17/IL-17A in neurodegeneration allows the identification of suitable targets to modulate these cellular processes. Therapeutics targeting IL-17A might represent potentially novel anti-neurodegeneration drugs.
Collapse
Affiliation(s)
- Jiajia Fu
- Department of Neurology, West China Hospital, Sichuan University, Wai Nan Guo Xue Xiang 37#, Chengdu, Sichuan, China
| | - Yan Huang
- Management Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ting Bao
- Management Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chengcheng Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Xi Liu
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xueping Chen
- Department of Neurology, West China Hospital, Sichuan University, Wai Nan Guo Xue Xiang 37#, Chengdu, Sichuan, China.
| |
Collapse
|
26
|
Contaldi E, Magistrelli L, Comi C. T Lymphocytes in Parkinson's Disease. JOURNAL OF PARKINSON'S DISEASE 2022; 12:S65-S74. [PMID: 35253782 PMCID: PMC9535550 DOI: 10.3233/jpd-223152] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
T cells are key mediators of both humoral and cellular adaptive immune responses, and their role in Parkinson’s disease (PD) is being increasingly recognized. Several lines of evidence have highlighted how T cells are involved in both the central nervous system and the periphery, leading to a profound imbalance in the immune network in PD patients. This review discusses the involvement of T cells in both preclinical and clinical studies, their importance as feasible biomarkers of motor and non-motor progression of the disease, and recent therapeutic strategies addressing the modulation of T cell response.
Collapse
Affiliation(s)
- Elena Contaldi
- Department of Translational Medicine, Movement Disorders Centre, "Maggiore della Caritá" University Hospital, University of Piemonte Orientale, Novara, Italy.,Program in Medical Sciences and Biotechnology, University of Piemonte Orientale, Novara, Italy
| | - Luca Magistrelli
- Department of Translational Medicine, Movement Disorders Centre, "Maggiore della Caritá" University Hospital, University of Piemonte Orientale, Novara, Italy.,Program in Clinical and Experimental Medicine and Medical Humanities, University of Insubria, Varese, Italy
| | - Cristoforo Comi
- Department of Translational Medicine, Neurology Unit, S. Andrea Hospital, University of Piemonte Orientale, Vercelli, Italy
| |
Collapse
|