1
|
Schrier MS, Smirnova MI, Nemeth DP, Deth RC, Quan N. Flavins and Flavoproteins in the Neuroimmune Landscape of Stress Sensitization and Major Depressive Disorder. J Inflamm Res 2025; 18:681-699. [PMID: 39839188 PMCID: PMC11748166 DOI: 10.2147/jir.s501652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 01/08/2025] [Indexed: 01/23/2025] Open
Abstract
Major Depressive Disorder (MDD) is a common and severe neuropsychiatric condition resulting in irregular alterations in affect, mood, and cognition. Besides the well-studied neurotransmission-related etiologies of MDD, several biological systems and phenomena, such as the hypothalamic-pituitary-adrenal (HPA) axis, reactive oxygen species (ROS) production, and cytokine signaling, have been implicated as being altered and contributing to depressive symptoms. However, the manner in which these factors interact with each other to induce their effects on MDD development has been less clear, but is beginning to be understood. Flavins are potent biomolecules that regulate many redox activities, including ROS generation and energy production. Studies have found that circulating flavin levels are modulated during stress and MDD. Flavins are also known for their importance in immune responses. This review offers a unique perspective that considers the redox-active cofactors, flavin mononucleotide (FMN) and flavin adenine dinucleotide (FAD), as vital substrates for linking MDD-related maladaptive processes together, by permitting stress-induced enhancement of microglial interleukin-1 beta (IL-1β) signaling.
Collapse
Affiliation(s)
- Matt Scott Schrier
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, USA
| | - Maria Igorevna Smirnova
- The International Max Planck Research School (IMPRS) for Synapses and Circuits, Jupiter, FL, USA
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, USA
- Department of Biological Sciences, Charles E. Schmidt College of Science, Florida Atlantic University, Jupiter, FL, USA
| | - Daniel Paul Nemeth
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, USA
| | - Richard Carlton Deth
- Department of Pharmaceutical Sciences, Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Ft. Lauderdale, FL, USA
| | - Ning Quan
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, USA
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| |
Collapse
|
2
|
Li S, Guo Z, Liu J, Ma Y, Zhang X, Hou L, Wang Q, Jiang W, Wang Q. CD11b-NOX2 mutual regulation-mediated microglial exosome release contributes to rotenone-induced inflammation and neurotoxicity in BV2 microglia and primary cultures. Free Radic Biol Med 2024; 224:436-446. [PMID: 39265792 DOI: 10.1016/j.freeradbiomed.2024.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 08/15/2024] [Accepted: 09/09/2024] [Indexed: 09/14/2024]
Abstract
Epidemiological studies have revealed a potent association between chronic exposure to rotenone, a commonly used pesticide, in individuals and the incidence of Parkinson's disease (PD). We previously identified the contribution of the activation of microglial NADPH oxidase (NOX2) in rotenone-induced neurotoxicity. However, the regulation of NOX2 activation remains unexplored. Integrins are known to be bidirectionally regulated in the plasma membrane through the inside-out and outside-in signaling. CD11b is the α-chain of integrin macrophage antigen complex-1. This study aimed to investigate whether CD11b mediates rotenone-induced NOX2 activation. We observed that rotenone exposure increased NOX2 activation in BV2 microglia, which was associated with elevated CD11b expression. Silencing CD11b significantly reduced rotenone-induced ROS production and p47phox phosphorylation, a key step for NOX2 activation. Furthermore, the Src-FAK-PKB and Syk-Vav1-Rac1 signaling pathways downstream of CD11b were found to be essential for CD11b-mediated NOX2 activation in rotenone-intoxicated microglia. Interestingly, we also found that inhibition of NOX2 decreased rotenone-induced CD11b expression, indicating a crosstalk between CD11b and NOX2. Subsequently, the inhibition of the CD11b-NOX2 axis suppressed rotenone-induced microglial activation and exosome release. Furthermore, inhibiting exosome synthesis in microglia blocked rotenone-induced gene expression of proinflammatory factors and related neurotoxicity. Finally, blocking the CD11b-NOX2 axis and exosome synthesis or endocytosis mitigated microglial activation and dopaminergic neurodegeneration in rotenone-intoxicated midbrain primary cultures. Our findings highlight the crucial involvement of the CD11b-NOX2 axis in rotenone-induced inflammation and neurotoxicity, offering fresh perspectives on the underlying mechanisms of pesticide-induced neuronal damage.
Collapse
Affiliation(s)
- Su Li
- School of Public Health, Dalian Medical University, Dalian, 116044, China; Department of Anesthesiology, Affiliated Zhongshan Hospital of Dalian University, Dalian, 116023, China
| | - Ziyang Guo
- School of Public Health, Dalian Medical University, Dalian, 116044, China
| | - Jianing Liu
- School of Public Health, Dalian Medical University, Dalian, 116044, China
| | - Yu Ma
- School of Public Health, Dalian Medical University, Dalian, 116044, China
| | - Xiaomeng Zhang
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044, China
| | - Liyan Hou
- Dalian Medical University Library, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian, 116044, China
| | - Qinghui Wang
- Department of Anesthesiology, Affiliated Zhongshan Hospital of Dalian University, Dalian, 116023, China
| | - Wanwei Jiang
- Department of Anesthesiology, Affiliated Zhongshan Hospital of Dalian University, Dalian, 116023, China.
| | - Qingshan Wang
- School of Public Health, Dalian Medical University, Dalian, 116044, China; National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044, China.
| |
Collapse
|
3
|
Wang Q, Liu J, Zhang Y, Li Z, Zhao Z, Jiang W, Zhao J, Hou L, Wang Q. Microglial CR3 promotes neuron ferroptosis via NOX2-mediated iron deposition in rotenone-induced experimental models of Parkinson's disease. Redox Biol 2024; 77:103369. [PMID: 39357423 PMCID: PMC11471230 DOI: 10.1016/j.redox.2024.103369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 09/22/2024] [Accepted: 09/22/2024] [Indexed: 10/04/2024] Open
Abstract
The activation of complement receptor 3 (CR3) in microglia contributes to neurodegeneration in neurological disorders, including Parkinson's disease (PD). However, it remains unclear for mechanistic knowledge on how CR3 mediates neuronal damage. In this study, the expression of CR3 and its ligands iC3b and ICAM-1 was found to be up-regulated in the midbrain of rotenone PD mice, which was associated with elevation of iron content and disruption of balance of iron metabolism proteins. Interestingly, genetic deletion of CR3 blunted iron accumulation and recovered the expression of iron metabolism markers in response to rotenone. Furthermore, reduced lipid peroxidation, ferroptosis of dopaminergic neurons and neuroinflammation were detected in rotenone-lesioned CR3-/- mice compared with WT mice. The regulatory effect of CR3 on ferroptotic death of dopaminergic neurons was also mirrored in vitro. Mechanistic study revealed that iron accumulation in neuron but not the physiological contact between microglia and neurons was essential for microglial CR3-regulated neuronal ferroptosis. In a cell-culture system, microglial CR3 silence significantly dampened iron deposition in neuron in response to rotenone, which was accompanied by mitigated lipid peroxidation and neurodegeneration. Furthermore, ROS released from activated microglia via NOX2 was identified to couple microglial CR3-mediated iron accumulation and subsequent neuronal ferroptosis. Finally, supplementation with exogenous iron was found to recover the sensitivity of CR3-/- mice to rotenone-induced neuronal ferroptosis. Altogether, our findings suggested that microglial CR3 regulates neuron ferroptosis through NOX2 -mediated iron accumulation in experimental Parkinsonism, providing novel points of the immunopathogenesis of neurological disorders.
Collapse
Affiliation(s)
- Qinghui Wang
- School of Public Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian, 116044, China; Department of Anesthesiology, Affiliated Zhongshan Hospital of Dalian University, Dalian, 116023, China
| | - Jianing Liu
- School of Public Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian, 116044, China
| | - Yu Zhang
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian, 116044, China
| | - Zhen Li
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian, 116044, China
| | - Zirui Zhao
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian, 116044, China
| | - Wanwei Jiang
- Department of Anesthesiology, Affiliated Zhongshan Hospital of Dalian University, Dalian, 116023, China
| | - Jie Zhao
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian, 116044, China
| | - Liyan Hou
- The Library of Dalian Medical University, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian, 116044, China.
| | - Qingshan Wang
- School of Public Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian, 116044, China; National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian, 116044, China.
| |
Collapse
|
4
|
Leak RK, Clark RN, Abbas M, Xu F, Brodsky JL, Chen J, Hu X, Luk KC. Current insights and assumptions on α-synuclein in Lewy body disease. Acta Neuropathol 2024; 148:18. [PMID: 39141121 PMCID: PMC11324801 DOI: 10.1007/s00401-024-02781-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 07/28/2024] [Accepted: 08/04/2024] [Indexed: 08/15/2024]
Abstract
Lewy body disorders are heterogeneous neurological conditions defined by intracellular inclusions composed of misshapen α-synuclein protein aggregates. Although α-synuclein aggregates are only one component of inclusions and not strictly coupled to neurodegeneration, evidence suggests they seed the propagation of Lewy pathology within and across cells. Genetic mutations, genomic multiplications, and sequence polymorphisms of the gene encoding α-synuclein are also causally linked to Lewy body disease. In nonfamilial cases of Lewy body disease, the disease trigger remains unidentified but may range from industrial/agricultural toxicants and natural sources of poisons to microbial pathogens. Perhaps due to these peripheral exposures, Lewy inclusions appear at early disease stages in brain regions connected with cranial nerves I and X, which interface with inhaled and ingested environmental elements in the nasal or gastrointestinal cavities. Irrespective of its identity, a stealthy disease trigger most likely shifts soluble α-synuclein (directly or indirectly) into insoluble, cross-β-sheet aggregates. Indeed, β-sheet-rich self-replicating α-synuclein multimers reside in patient plasma, cerebrospinal fluid, and other tissues, and can be subjected to α-synuclein seed amplification assays. Thus, clinicians should be able to capitalize on α-synuclein seed amplification assays to stratify patients into potential responders versus non-responders in future clinical trials of α-synuclein targeted therapies. Here, we briefly review the current understanding of α-synuclein in Lewy body disease and speculate on pathophysiological processes underlying the potential transmission of α-synucleinopathy across the neuraxis.
Collapse
Affiliation(s)
- Rehana K Leak
- Graduate School of Pharmaceutical Sciences, Duquesne University, 418C Mellon Hall, 913 Bluff Street, Pittsburgh, PA, 15219, USA.
| | - Rachel N Clark
- Graduate School of Pharmaceutical Sciences, Duquesne University, 418C Mellon Hall, 913 Bluff Street, Pittsburgh, PA, 15219, USA
| | - Muslim Abbas
- Graduate School of Pharmaceutical Sciences, Duquesne University, 418C Mellon Hall, 913 Bluff Street, Pittsburgh, PA, 15219, USA
| | - Fei Xu
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jeffrey L Brodsky
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jun Chen
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, Pennsylvania, USA
| | - Xiaoming Hu
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kelvin C Luk
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Pennsylvania, PA, USA
| |
Collapse
|
5
|
Hu R, Wang R, Yuan J, Lin Z, Hutchins E, Landin B, Liao Z, Liu G, Scherzer CR, Dong X. Transcriptional pathobiology and multi-omics predictors for Parkinson's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.18.599639. [PMID: 38948706 PMCID: PMC11212969 DOI: 10.1101/2024.06.18.599639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Early diagnosis and biomarker discovery to bolster the therapeutic pipeline for Parkinson's disease (PD) are urgently needed. In this study, we leverage the large-scale whole-blood total RNA-seq dataset from the Accelerating Medicine Partnership in Parkinson's Disease (AMP PD) program to identify PD-associated RNAs, including both known genes and novel circular RNAs (circRNA) and enhancer RNAs (eRNAs). There were 1,111 significant marker RNAs, including 491 genes, 599 eRNAs, and 21 circRNAs, that were first discovered in the PPMI cohort (FDR < 0.05) and confirmed in the PDBP/BioFIND cohorts (nominal p < 0.05). Functional enrichment analysis showed that the PD-associated genes are involved in neutrophil activation and degranulation, as well as the TNF-alpha signaling pathway. We further compare the PD-associated genes in blood with those in post-mortem brain dopamine neurons in our BRAINcode cohort. 44 genes show significant changes with the same direction in both PD brain neurons and PD blood, including neuroinflammation-associated genes IKBIP, CXCR2, and NFKBIB. Finally, we built a novel multi-omics machine learning model to predict PD diagnosis with high performance (AUC = 0.89), which was superior to previous studies and might aid the decision-making for PD diagnosis in clinical practice. In summary, this study delineates a wide spectrum of the known and novel RNAs linked to PD and are detectable in circulating blood cells in a harmonized, large-scale dataset. It provides a generally useful computational framework for further biomarker development and early disease prediction.
Collapse
Affiliation(s)
- Ruifeng Hu
- APDA Center for Advanced Parkinson Research, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Precision Neurology Program, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Genomics and Bioinformatics Hub, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Ruoxuan Wang
- APDA Center for Advanced Parkinson Research, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Precision Neurology Program, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Genomics and Bioinformatics Hub, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Jie Yuan
- APDA Center for Advanced Parkinson Research, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Precision Neurology Program, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Genomics and Bioinformatics Hub, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Zechuan Lin
- APDA Center for Advanced Parkinson Research, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Precision Neurology Program, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Elizabeth Hutchins
- Neurogenomics Division, Translational Genomics Research Institute (TGen), Phoenix, AZ, USA
| | | | - Zhixiang Liao
- APDA Center for Advanced Parkinson Research, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Precision Neurology Program, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Ganqiang Liu
- APDA Center for Advanced Parkinson Research, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Shenzhen Key Laboratory of Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Clemens R. Scherzer
- APDA Center for Advanced Parkinson Research, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Precision Neurology Program, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Xianjun Dong
- APDA Center for Advanced Parkinson Research, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Precision Neurology Program, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Genomics and Bioinformatics Hub, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| |
Collapse
|
6
|
Tan LY, Cunliffe G, Hogan MP, Yeo XY, Oh C, Jin B, Kang J, Park J, Kwon MS, Kim M, Jung S. Emergence of the brain-border immune niches and their contribution to the development of neurodegenerative diseases. Front Immunol 2024; 15:1380063. [PMID: 38863704 PMCID: PMC11165048 DOI: 10.3389/fimmu.2024.1380063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 05/14/2024] [Indexed: 06/13/2024] Open
Abstract
Historically, the central nervous system (CNS) was regarded as 'immune-privileged', possessing its own distinct immune cell population. This immune privilege was thought to be established by a tight blood-brain barrier (BBB) and blood-cerebrospinal-fluid barrier (BCSFB), which prevented the crossing of peripheral immune cells and their secreted factors into the CNS parenchyma. However, recent studies have revealed the presence of peripheral immune cells in proximity to various brain-border niches such as the choroid plexus, cranial bone marrow (CBM), meninges, and perivascular spaces. Furthermore, emerging evidence suggests that peripheral immune cells may be able to infiltrate the brain through these sites and play significant roles in driving neuronal cell death and pathology progression in neurodegenerative disease. Thus, in this review, we explore how the brain-border immune niches may contribute to the pathogenesis of neurodegenerative disorders such as Alzheimer's disease (AD), Parkinson's disease (PD), and multiple sclerosis (MS). We then discuss several emerging options for harnessing the neuroimmune potential of these niches to improve the prognosis and treatment of these debilitative disorders using novel insights from recent studies.
Collapse
Affiliation(s)
- Li Yang Tan
- Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Grace Cunliffe
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Michael Patrick Hogan
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Xin Yi Yeo
- Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Chansik Oh
- Department of Medical Science, College of Medicine, CHA University, Seongnam, Republic of Korea
| | - Bohwan Jin
- Department of Medical Science, College of Medicine, CHA University, Seongnam, Republic of Korea
| | - Junmo Kang
- Department of Medical Science, College of Medicine, CHA University, Seongnam, Republic of Korea
| | - Junho Park
- Department of Pharmacology, Research Institute for Basic Medical Science, School of Medicine, CHA University, Seongnam, Republic of Korea
| | - Min-Soo Kwon
- Department of Pharmacology, Research Institute for Basic Medical Science, School of Medicine, CHA University, Seongnam, Republic of Korea
| | - MinYoung Kim
- Rehabilitation and Regeneration Research Center, CHA University School of Medicine, Seongnam, Republic of Korea
- Department of Biomedical Science, CHA University School of Medicine, Seongnam, Republic of Korea
- Department of Rehabilitation Medicine, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Sangyong Jung
- Department of Medical Science, College of Medicine, CHA University, Seongnam, Republic of Korea
| |
Collapse
|
7
|
Kou L, Chi X, Sun Y, Yin S, Wu J, Zou W, Wang Y, Jin Z, Huang J, Xiong N, Xia Y, Wang T. Circadian regulation of microglia function: Potential targets for treatment of Parkinson's Disease. Ageing Res Rev 2024; 95:102232. [PMID: 38364915 DOI: 10.1016/j.arr.2024.102232] [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: 07/17/2023] [Revised: 02/11/2024] [Accepted: 02/11/2024] [Indexed: 02/18/2024]
Abstract
Circadian rhythms are involved in the regulation of many aspects of the body, including cell function, physical activity and disease. Circadian disturbance often predates the typical symptoms of neurodegenerative diseases and is not only a non-motor symptom, but also one of the causes of their occurrence and progression. Glial cells possess circadian clocks that regulate their function to maintain brain development and homeostasis. Emerging evidence suggests that the microglial circadian clock is involved in the regulation of many physiological processes, such as cytokine release, phagocytosis, and nutritional and metabolic support, and that disruption of the microglia clock may affect multiple aspects of Parkinson's disease, especially neuroinflammation and α-synuclein processes. Herein, we review recent advances in the circadian control of microglia function in health and disease, and discuss novel pharmacological interventions for microglial clocks in neurodegenerative disorders.
Collapse
Affiliation(s)
- Liang Kou
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiaosa Chi
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yadi Sun
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Sijia Yin
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jiawei Wu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Wenkai Zou
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yiming Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zongjie Jin
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jinsha Huang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Nian Xiong
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yun Xia
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Tao Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
8
|
Miao Y, Meng H. The involvement of α-synucleinopathy in the disruption of microglial homeostasis contributes to the pathogenesis of Parkinson's disease. Cell Commun Signal 2024; 22:31. [PMID: 38216911 PMCID: PMC10785555 DOI: 10.1186/s12964-023-01402-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 11/18/2023] [Indexed: 01/14/2024] Open
Abstract
The intracellular deposition and intercellular transmission of α-synuclein (α-syn) are shared pathological characteristics among neurodegenerative disorders collectively known as α-synucleinopathies, including Parkinson's disease (PD). Although the precise triggers of α-synucleinopathies remain unclear, recent findings indicate that disruption of microglial homeostasis contributes to the pathogenesis of PD. Microglia play a crucial role in maintaining optimal neuronal function by ensuring a homeostatic environment, but this function is disrupted during the progression of α-syn pathology. The involvement of microglia in the accumulation, uptake, and clearance of aggregated proteins is critical for managing disease spread and progression caused by α-syn pathology. This review summarizes current knowledge on the interrelationships between microglia and α-synucleinopathies, focusing on the remarkable ability of microglia to recognize and internalize extracellular α-syn through diverse pathways. Microglia process α-syn intracellularly and intercellularly to facilitate the α-syn neuronal aggregation and cell-to-cell propagation. The conformational state of α-synuclein distinctly influences microglial inflammation, which can affect peripheral immune cells such as macrophages and lymphocytes and may regulate the pathogenesis of α-synucleinopathies. We also discuss ongoing research efforts to identify potential therapeutic approaches targeting both α-syn accumulation and inflammation in PD. Video Abstract.
Collapse
Affiliation(s)
- Yongzhen Miao
- Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, China
| | - Hongrui Meng
- Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, China.
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
9
|
Niederberger E, Möller M, Mungo E, Hass M, Wilken-Schmitz A, Manderscheid C, Möser CV, Geisslinger G. Distinct molecular mechanisms contribute to the reduction of melanoma growth and tumor pain after systemic and local depletion of alpha-Synuclein in mice. FASEB J 2023; 37:e23287. [PMID: 37930651 DOI: 10.1096/fj.202301489r] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/29/2023] [Accepted: 10/18/2023] [Indexed: 11/07/2023]
Abstract
Epidemiological studies show a coincidence between Parkinson's disease (PD) and malignant melanoma. It has been suggested that this relationship is due, at least in part, to modulation of alpha-Synuclein (αSyn/Snca). αSyn oligomers accumulate in PD, which triggers typical PD symptoms, and in malignant melanoma, which increases the proliferation of tumor cells. In addition, αSyn contributes to non-motor symptoms of PD, including pain. In this study, we investigated the role of αSyn in melanoma growth and melanoma-induced pain in a mouse model using systemic and local depletion of αSyn. B16BL6 wild-type as well as αSyn knock-down melanoma cells were inoculated into the paws of αSyn knock-out mice and wild-type mice, respectively. Tumor growth and tumor-induced pain hypersensitivity were assessed over a period of 21 days. Molecular mechanisms were analyzed by RT-PCR and Western Blot in tumors, spinal cord, and sciatic nerve. Our results indicate that both global and local ablation of Snca contribute to reduced tumor growth and to a reduction of tumor-induced mechanical allodynia, though mechanisms contributing to these effects differ. While injection of wild-type cells in Snca knock-out mice strongly increased the immune response in the tumor, local Snca knock-down decreased autophagy mechanisms and the inflammatory reaction in the tumor. In conclusion, a knockdown of αSyn might constitute a promising approach to inhibiting the progression of melanoma and reducing tumor-induced pain.
Collapse
Affiliation(s)
- Ellen Niederberger
- Goethe-Universität Frankfurt, Universitätsklinikum, pharmazentrum frankfurt/ZAFES, Institut für Klinische Pharmakologie, Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine & Pharmacology ITMP, Frankfurt am Main, Germany
| | - Moritz Möller
- Goethe-Universität Frankfurt, Universitätsklinikum, pharmazentrum frankfurt/ZAFES, Institut für Klinische Pharmakologie, Frankfurt am Main, Germany
| | - Eleonora Mungo
- Goethe-Universität Frankfurt, Universitätsklinikum, pharmazentrum frankfurt/ZAFES, Institut für Klinische Pharmakologie, Frankfurt am Main, Germany
| | - Michelle Hass
- Goethe-Universität Frankfurt, Universitätsklinikum, pharmazentrum frankfurt/ZAFES, Institut für Klinische Pharmakologie, Frankfurt am Main, Germany
| | - Annett Wilken-Schmitz
- Goethe-Universität Frankfurt, Universitätsklinikum, pharmazentrum frankfurt/ZAFES, Institut für Klinische Pharmakologie, Frankfurt am Main, Germany
| | - Christine Manderscheid
- Goethe-Universität Frankfurt, Universitätsklinikum, pharmazentrum frankfurt/ZAFES, Institut für Klinische Pharmakologie, Frankfurt am Main, Germany
| | - Christine V Möser
- Goethe-Universität Frankfurt, Universitätsklinikum, pharmazentrum frankfurt/ZAFES, Institut für Klinische Pharmakologie, Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine & Pharmacology ITMP, Frankfurt am Main, Germany
| | - Gerd Geisslinger
- Goethe-Universität Frankfurt, Universitätsklinikum, pharmazentrum frankfurt/ZAFES, Institut für Klinische Pharmakologie, Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine & Pharmacology ITMP, Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Frankfurt am Main, Germany
| |
Collapse
|
10
|
McFleder RL, Makhotkina A, Groh J, Keber U, Imdahl F, Peña Mosca J, Peteranderl A, Wu J, Tabuchi S, Hoffmann J, Karl AK, Pagenstecher A, Vogel J, Beilhack A, Koprich JB, Brotchie JM, Saliba AE, Volkmann J, Ip CW. Brain-to-gut trafficking of alpha-synuclein by CD11c + cells in a mouse model of Parkinson's disease. Nat Commun 2023; 14:7529. [PMID: 37981650 PMCID: PMC10658151 DOI: 10.1038/s41467-023-43224-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 11/03/2023] [Indexed: 11/21/2023] Open
Abstract
Inflammation in the brain and gut is a critical component of several neurological diseases, such as Parkinson's disease (PD). One trigger of the immune system in PD is aggregation of the pre-synaptic protein, α-synuclein (αSyn). Understanding the mechanism of propagation of αSyn aggregates is essential to developing disease-modifying therapeutics. Using a brain-first mouse model of PD, we demonstrate αSyn trafficking from the brain to the ileum of male mice. Immunohistochemistry revealed that the ileal αSyn aggregations are contained within CD11c+ cells. Using single-cell RNA sequencing, we demonstrate that ileal CD11c+ cells are microglia-like and the same subtype of cells is activated in the brain and ileum of PD mice. Moreover, by utilizing mice expressing the photo-convertible protein, Dendra2, we show that CD11c+ cells traffic from the brain to the ileum. Together these data provide a mechanism of αSyn trafficking between the brain and gut.
Collapse
Affiliation(s)
- Rhonda L McFleder
- Department of Neurology, University Hospital of Würzburg, Würzburg, Germany
| | | | - Janos Groh
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
| | - Ursula Keber
- Department of Neuropathology, Philipps University of Marburg, Marburg, Germany
| | - Fabian Imdahl
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz-Center for Infection Research (HZI), Würzburg, Germany
| | - Josefina Peña Mosca
- Department of Internal Medicine II, Center for Experimental Molecular Medicine (ZEMM), Würzburg University Hospital, Würzburg, Germany
| | - Alina Peteranderl
- Department of Neurology, University Hospital of Würzburg, Würzburg, Germany
| | - Jingjing Wu
- Department of Neurology, University Hospital of Würzburg, Würzburg, Germany
| | - Sawako Tabuchi
- Department of Neurology, University Hospital of Würzburg, Würzburg, Germany
| | - Jan Hoffmann
- Department of Neurology, University Hospital of Würzburg, Würzburg, Germany
| | - Ann-Kathrin Karl
- Department of Neurology, University Hospital of Würzburg, Würzburg, Germany
| | - Axel Pagenstecher
- Department of Neuropathology, Philipps University of Marburg, Marburg, Germany
| | - Jörg Vogel
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz-Center for Infection Research (HZI), Würzburg, Germany
| | - Andreas Beilhack
- Department of Internal Medicine II, Center for Experimental Molecular Medicine (ZEMM), Würzburg University Hospital, Würzburg, Germany
| | - James B Koprich
- Atuka Inc., Toronto, ON, Canada
- Krembil Research Institute, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
| | - Jonathan M Brotchie
- Atuka Inc., Toronto, ON, Canada
- Krembil Research Institute, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
| | - Antoine-Emmanuel Saliba
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz-Center for Infection Research (HZI), Würzburg, Germany
- Faculty of Medicine, Institute of Molecular Infection Biology (IMIB), University of Würzburg, Josef-Schneider-Str. 2, 97080, Würzburg, Germany
| | - Jens Volkmann
- Department of Neurology, University Hospital of Würzburg, Würzburg, Germany
| | - Chi Wang Ip
- Department of Neurology, University Hospital of Würzburg, Würzburg, Germany.
| |
Collapse
|
11
|
Carroll JA, Striebel JF, Baune C, Chesebro B, Race B. CD11c is not required by microglia to convey neuroprotection after prion infection. PLoS One 2023; 18:e0293301. [PMID: 37910561 PMCID: PMC10619787 DOI: 10.1371/journal.pone.0293301] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 10/10/2023] [Indexed: 11/03/2023] Open
Abstract
Prion diseases are caused by the misfolding of a normal host protein that leads to gliosis, neuroinflammation, neurodegeneration, and death. Microglia have been shown to be critical for neuroprotection during prion infection of the central nervous system (CNS), and their presence extends survival in mice. How microglia impart these benefits to the infected host are unknown. Previous transcriptomics and bioinformatics studies suggested that signaling through the heterodimeric integrin receptor CD11c/CD18, expressed by microglia in the brain, might be important to microglial function during prion disease. Herein, we intracerebrally challenged CD11c-/- mice with prion strain RML and compared them to similarly infected C57BL/6 mice as controls. We initially assessed changes in the brain that are associated with disease such as astrogliosis, microgliosis, prion accumulation, and survival. Targeted qRT-PCR arrays were used to determine alterations in transcription in mice in response to prion infection. We demonstrate that expression of Itgax (CD11c) and Itgb2 (CD18) increases in the CNS in correlation with advancing prion infection. Gliosis, neuropathology, prion deposition, and disease progression in prion infected CD11c deficient mice were comparable to infected C57BL/6 mice. Additionally, both CD11c deficient and C57BL/6 prion-infected mouse cohorts had a similar consortium of inflammatory- and phagocytosis-associated genes that increased as disease progressed to clinical stages. Ingenuity Pathway Analysis of upregulated genes in infected C57BL/6 mice suggested numerous cell-surface transmembrane receptors signal through Spleen Tyrosine Kinase, a potential key regulator of phagocytosis and innate immune activation in the prion infected brain. Ultimately, the deletion of CD11c did not influence prion pathogenesis in mice and CD11c signaling is not involved in the neuroprotection provided by microglia, but our analysis identified a conspicuous phagocytosis pathway in the CNS of infected mice that appeared to be activated during prion pathogenesis.
Collapse
Affiliation(s)
- James A. Carroll
- Laboratory of Neurological Infections and Immunity, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - James F. Striebel
- Laboratory of Neurological Infections and Immunity, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Chase Baune
- Laboratory of Neurological Infections and Immunity, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Bruce Chesebro
- Laboratory of Neurological Infections and Immunity, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Brent Race
- Laboratory of Neurological Infections and Immunity, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| |
Collapse
|
12
|
Chu CH, Chen JS, Chan YL, Lu WJ, Huang YT, Mao PC, Sze CI, Sun HS. TIAM2S-positive microglia enhance inflammation and neurotoxicity through soluble ICAM-1-mediated immune priming. FASEB J 2023; 37:e23242. [PMID: 37801065 DOI: 10.1096/fj.202300462rr] [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: 03/10/2023] [Revised: 09/05/2023] [Accepted: 09/22/2023] [Indexed: 10/07/2023]
Abstract
TIAM Rac1-associated GEF 2 short form (TIAM2S) as an oncoprotein alters the immunity of peripheral immune cells to construct an inflammatory tumor microenvironment. However, its role in the activation of microglia, the primary innate immune cells of the brain, and neuroinflammation remains unknown. This study investigated the mechanism underlying TIAM2S shapes immune properties of microglia to facilitate neuron damage. Human microglial clone 3 cell line (HMC3) and human brain samples were applied to determine the presence of TIAM2S in microglia by western blots and double immunostaining. Furthermore, TIAM2S transgenic mice combined with multiple reconstituted primary neuron-glial culture systems and a cytokine array were performed to explore how TIAM2S shaped immune priming of microglia and participated in lipopolysaccharide (LPS)-induced neuron damage. TIAM2S protein was detectable in HMC3 cells and presented in a small portion (~11.1%) of microglia in human brains referred to as TIAM2S-positive microglia. With the property of secreted soluble factor-mediated immune priming, TIAM2S-positive microglia enhanced LPS-induced neuroinflammation and neural damage in vivo and in vitro. The gain- and loss-of-function experiments showed soluble intercellular adhesion molecule-1 (sICAM-1) participated in neurotoxic immune priming of TIAM2S+ microglia. Together, this study demonstrated a novel TIAM2S-positive microglia subpopulation enhances inflammation and neurotoxicity through sICAM-1-mediated immune priming.
Collapse
Affiliation(s)
- Chun-Hsien Chu
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Jia-Shing Chen
- School of Medicine for International Students, I-Shou University, Kaohsiung, Taiwan
| | - Ya-Ling Chan
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Wei-Jen Lu
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Te Huang
- Department of Geriatrics and Gerontology, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Pin-Cheng Mao
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chun-I Sze
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - H Sunny Sun
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
13
|
Kang YJ, Diep YN, Tran M, Tran VTA, Ambrin G, Ngo H, Cho H. Three-dimensional human neural culture on a chip recapitulating neuroinflammation and neurodegeneration. Nat Protoc 2023; 18:2838-2867. [PMID: 37542184 DOI: 10.1038/s41596-023-00861-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 05/30/2023] [Indexed: 08/06/2023]
Abstract
Neuroinflammation has either beneficial or detrimental effects, depending on risk factors and neuron-glia interactions in neurological disorders. However, studying neuroinflammation has been challenging due to the complexity of cell-cell interactions and lack of physio-pathologically relevant neuroinflammatory models. Here, we describe our three-dimensional microfluidic multicellular human neural culture model, referred to as a 'brain-on-a-chip' (BoC). This elucidates neuron-glia interactions in a controlled manner and recapitulates pathological signatures of the major neurological disorders: dementia, brain tumor and brain edema. This platform includes a chemotaxis module offering a week-long, stable chemo-gradient compared with the few hours in other chemotaxis models. Additionally, compared with conventional brain models cultured with mixed phenotypes of microglia, our BoC can separate the disease-associated microglia out of heterogeneous population and allow selective neuro-glial engagement in three dimensions. This provides benefits of interpreting the neuro-glia interactions while revealing that the prominent activation of innate immune cells is the risk factor leading to synaptic impairment and neuronal loss, validated in our BoC models of disorders. This protocol describes how to fabricate and implement our human BoC, manipulate in real time and perform end-point analyses. It takes 2 d to set up the device and cell preparations, 1-9 weeks to develop brain models under disease conditions and 2-3 d to carry out analyses. This protocol requires at least 1 month training for researchers with basic molecular biology techniques. Taken together, our human BoCs serve as reliable and valuable platforms to investigate pathological mechanisms involving neuroinflammation and to assess therapeutic strategies modulating neuroinflammation in neurological disorders.
Collapse
Affiliation(s)
- You Jung Kang
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon, Republic of Korea
| | - Yen N Diep
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, Republic of Korea
| | - Minh Tran
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, Republic of Korea
| | - Van Thi Ai Tran
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon, Republic of Korea
| | - Ghuncha Ambrin
- Department of Psychiatry, School of Medicine, University of California San Diego, San Diego, CA, USA
| | - Huyen Ngo
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, Republic of Korea
| | - Hansang Cho
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Republic of Korea.
- Department of Biophysics, Sungkyunkwan University, Suwon, Republic of Korea.
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, Republic of Korea.
| |
Collapse
|
14
|
Liu Z, Lemus J, Smirnova IV, Liu W. Rehabilitation for non-motor symptoms for patients with Parkinson's disease from an α-synuclein perspective: a narrative review. EXPLORATION OF NEUROPROTECTIVE THERAPY 2023; 3:235-257. [PMID: 37920444 PMCID: PMC10621781 DOI: 10.37349/ent.2023.00049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 06/22/2023] [Indexed: 11/04/2023]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder affecting aged population around the world. PD is characterized by neuronal Lewy bodies present in the substantia nigra of the midbrain and the loss of dopaminergic neurons with various motor and non-motor symptoms associated with the disease. The protein α-synuclein has been extensively studied for its contribution to PD pathology, as α-synuclein aggregates form the major component of Lewy bodies, a hallmark of PD. In this narrative review, the authors first focus on a brief explanation of α-synuclein aggregation and circumstances under which aggregation can occur, then present a hypothesis for PD pathogenesis in the peripheral nervous system (PNS) and how PD can spread to the central nervous system from the PNS via the transport of α-synuclein aggregates. This article presents arguments both for and against this hypothesis. It also presents various non-pharmacological rehabilitation approaches and management techniques for both motor and non-motor symptoms of PD and the related pathology. This review seeks to examine a possible hypothesis of PD pathogenesis and points to a new research direction focus on rehabilitation therapy for patients with PD. As various non-motor symptoms of PD appear to occur earlier than motor symptoms, more focus on the treatment of non-motor symptoms as well as a better understanding of the biochemical mechanisms behind those non-motor symptoms may lead to better long-term outcomes for patients with PD.
Collapse
Affiliation(s)
- Zhaoyang Liu
- Department of Physical Therapy, Rehabilitation Science, and Athletic Training, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Department of Orthopedic Surgery and Sports Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Jessica Lemus
- Department of Physical Therapy, Rehabilitation Science, and Athletic Training, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Irina V. Smirnova
- Department of Physical Therapy, Rehabilitation Science, and Athletic Training, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Wen Liu
- Department of Physical Therapy, Rehabilitation Science, and Athletic Training, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
15
|
Heiden DL, Monogue B, Ali MDH, Beckham JD. A functional role for alpha-synuclein in neuroimmune responses. J Neuroimmunol 2023; 376:578047. [PMID: 36791583 PMCID: PMC10022478 DOI: 10.1016/j.jneuroim.2023.578047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 01/30/2023] [Accepted: 02/04/2023] [Indexed: 02/11/2023]
Abstract
Alpha-synuclein is a neuronal protein with unclear function but is associated with the pathogenesis of Parkinson's disease and other synucleinopathies. In this review, we discuss the emerging functional role of alpha-synuclein in support of the unique immune responses in the nervous system. Recent data now show that alpha-synuclein functions to support interferon signaling within neurons and is released from neurons to support chemoattraction and activation of local glial cells and infiltrating immune cells. Inflammatory activation and interferon signaling also induce post-translational modifications of alpha-synuclein that are commonly associated with Parkinson's disease pathogenesis. Taken together, emerging data implicate complex interactions between alpha-synuclein and host immune responses that may contribute to the pathogenesis of Parkinson's disease. Additional study of the function of alpha-synuclein in the brain's immune response may provide disease-modifying therapeutic targets for Parkinson's disease in the future.
Collapse
Affiliation(s)
- Dustin L Heiden
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Brendan Monogue
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - M D Haider Ali
- Department of Medicine, Division of Infectious Diseases, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - J David Beckham
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Medicine, Division of Infectious Diseases, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Rocky Mountain Regional VA Medical Center, Aurora, CO, USA.
| |
Collapse
|
16
|
Lv QK, Tao KX, Wang XB, Yao XY, Pang MZ, Liu JY, Wang F, Liu CF. Role of α-synuclein in microglia: autophagy and phagocytosis balance neuroinflammation in Parkinson's disease. Inflamm Res 2023; 72:443-462. [PMID: 36598534 DOI: 10.1007/s00011-022-01676-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 11/27/2022] [Accepted: 12/12/2022] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Parkinson's disease (PD) is the second most common neurodegenerative disease, and is characterized by accumulation of α-synuclein (α-syn). Neuroinflammation driven by microglia is an important pathological manifestation of PD. α-Syn is a crucial marker of PD, and its accumulation leads to microglia M1-like phenotype polarization, activation of NLRP3 inflammasomes, and impaired autophagy and phagocytosis in microglia. Autophagy of microglia is related to degradation of α-syn and NLRP3 inflammasome blockage to relieve neuroinflammation. Microglial autophagy and phagocytosis of released α-syn or fragments from apoptotic neurons maintain homeostasis in the brain. A variety of PD-related genes such as LRRK2, GBA and DJ-1 also contribute to this stability process. OBJECTIVES Further studies are needed to determine how α-syn works in microglia. METHODS A keyword-based search was performed using the PubMed database for published articles. CONCLUSION In this review, we discuss the interaction between microglia and α-syn in PD pathogenesis and the possible mechanism of microglial autophagy and phagocytosis in α-syn clearance and inhibition of neuroinflammation. This may provide a novel insight into treatment of PD.
Collapse
Affiliation(s)
- Qian-Kun Lv
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Kang-Xin Tao
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Xiao-Bo Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Xiao-Yu Yao
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Meng-Zhu Pang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Jun-Yi Liu
- Department of Neurology, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, China
| | - Fen Wang
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China.
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, 215123, China.
| | - Chun-Feng Liu
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China.
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, 215123, China.
| |
Collapse
|
17
|
Kumari S, Taliyan R, Dubey SK. Comprehensive Review on Potential Signaling Pathways Involving the Transfer of α-Synuclein from the Gut to the Brain That Leads to Parkinson's Disease. ACS Chem Neurosci 2023; 14:590-602. [PMID: 36724408 DOI: 10.1021/acschemneuro.2c00730] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Parkinson's disease is the second most prevalent neurological disease after Alzheimer's. Primarily, old age males are more affected than females. The aggregates of oligomeric forms of α-synuclein cause the loss of dopaminergic neurons in the substantia nigra pars compacta. Further, it leads to dopamine shortage in the striatum region. According to recent preclinical studies, environmental factors like pesticides, food supplements, pathogens, etc. enter the body through the mouth or nose and ultimately reach the gut. Further, these factors get accumulated in enteric nervous system which leads to misfolding of α-synuclein gene, and aggregation of this gene results in Lewy pathology in the gut and reaches to the brain through the vagus nerve. This evidence showed a strong bidirectional connection between the gut and the brain, which leads to gastrointestinal problems in Parkinson patients. Moreover, several studies reveal that patients with Parkinson experience more gastrointestinal issues in the early stages of the disease, such as constipation, increased motility, gut inflammation, etc. This review article focuses on the transmission of α-synuclein and the mechanisms involved in the link between the gut and the brain in Parkinson's disease. Also, this review explores the various pathways involved in Parkinson and current therapeutic approaches for the improvement of Parkinson's disease.
Collapse
Affiliation(s)
- Shobha Kumari
- Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science─Pilani, Pilani, 333031 Rajasthan, India
| | - Rajeev Taliyan
- Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science─Pilani, Pilani, 333031 Rajasthan, India
| | | |
Collapse
|
18
|
Wang Q, Zheng J, Pettersson S, Reynolds R, Tan EK. The link between neuroinflammation and the neurovascular unit in synucleinopathies. SCIENCE ADVANCES 2023; 9:eabq1141. [PMID: 36791205 PMCID: PMC9931221 DOI: 10.1126/sciadv.abq1141] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 01/19/2023] [Indexed: 05/28/2023]
Abstract
The neurovascular unit (NVU) is composed of vascular cells, glial cells, and neurons. As a fundamental functional module in the central nervous system, the NVU maintains homeostasis in the microenvironment and the integrity of the blood-brain barrier. Disruption of the NVU and interactions among its components are involved in the pathophysiology of synucleinopathies, which are characterized by the pathological accumulation of α-synuclein. Neuroinflammation contributes to the pathophysiology of synucleinopathies, including Parkinson's disease, multiple system atrophy, and dementia with Lewy bodies. This review aims to summarize the neuroinflammatory response of glial cells and vascular cells in the NVU. We also review neuroinflammation in the context of the cross-talk between glial cells and vascular cells, between glial cells and pericytes, and between microglia and astroglia. Last, we discuss how α-synuclein affects neuroinflammation and how neuroinflammation influences the aggregation and spread of α-synuclein and analyze different properties of α-synuclein in synucleinopathies.
Collapse
Affiliation(s)
- Qing Wang
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Jialing Zheng
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Sven Pettersson
- ASEAN Microbiome Nutrition Centre, National Neuroscience Institute, Singapore 308433, Singapore
- Karolinska Institutet, Department of Odontology, 171 77 Solna, Sweden
- Faculty of Medical Sciences, Sunway University, Subang Jaya, 47500 Selangor, Malaysia
- Department of Microbiology and Immunology, National University Singapore, Singapore 117545, Singapore
| | - Richard Reynolds
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital Campus, Burlington Danes Building, Du Cane Road, London W12 0NN, UK
- Centre for Molecular Neuropathology, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Eng-King Tan
- Department of Neurology, National Neuroscience Institute, Singapore General Hospital, Duke-NUS Medical School, Singapore, Singapore
| |
Collapse
|
19
|
Balzano T, Esteban-García N, Blesa J. Neuroinflammation, immune response and α-synuclein pathology: how animal models are helping us to connect dots. Expert Opin Drug Discov 2023; 18:13-23. [PMID: 36538833 DOI: 10.1080/17460441.2023.2160440] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022]
Abstract
INTRODUCTION A key pathological event occurring in Parkinson's disease (PD) is the transneuronal spreading of alpha-synuclein (α-syn). Other hallmarks of PD include neurodegeneration, glial activation, and immune cell infiltration in susceptible brain regions. Although preclinical models can mimic most of the key characteristics of PD, it is crucial to know the biological bases of individual differences between them when choosing one over another, to ensure proper interpretation of the results and to positively influence the outcome of the experiments. AREAS COVERED This review provides an overview of current preclinical models actively used to study the interplay between α-syn pathology, neuroinflammation and immune response in PD but also to explore new potential preclinical models or emerging therapeutic strategies intended to fulfill the unmet medical needs in this disease. Lastly, this review also considers the current state of the ongoing clinical trials of new drugs designed to target these processes and delay the initiation or progression of the disease. EXPERT OPINION Anti-inflammatory and immunomodulatory agents have been demonstrated to be very promising candidates for reducing disease progression; however, more efforts are needed to reduce the enormous gap between these and dopaminergic drugs, which have dominated the therapeutic market for the last sixty years.
Collapse
Affiliation(s)
- Tiziano Balzano
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, Madrid, Spain
| | - Noelia Esteban-García
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, Madrid, Spain
- PhD Program in Neuroscience Autónoma de Madrid University-Cajal Institute, Madrid, Spain
| | - Javier Blesa
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, Madrid, Spain
- Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III; Madrid, Madrid, Spain
| |
Collapse
|
20
|
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
|
21
|
Portugal CC, Almeida TO, Socodato R, Relvas JB. Src family kinases (SFKs): critical regulators of microglial homeostatic functions and neurodegeneration in Parkinson's and Alzheimer's diseases. FEBS J 2022; 289:7760-7775. [PMID: 34510775 DOI: 10.1111/febs.16197] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 08/03/2021] [Accepted: 09/10/2021] [Indexed: 01/14/2023]
Abstract
c-Src was the first protein kinase to be described as capable of phosphorylating tyrosine residues. Subsequent identification of other tyrosine-phosphorylating protein kinases with a similar structure to c-Src gave rise to the concept of Src family kinases (SFKs). Microglia are the resident innate immune cell population of the CNS. Under physiological conditions, microglia actively participate in brain tissue homeostasis, continuously patrolling the neuronal parenchyma and exerting neuroprotective actions. Activation of pathogen-associated molecular pattern (PAMP) and damage-associated molecular pattern (DAMP) receptors induces microglial proliferation, migration toward pathological foci, phagocytosis, and changes in gene expression, concurrent with the secretion of cytokines, chemokines, and growth factors. A significant body of literature shows that SFK stimulation positively associates with microglial activation and neuropathological conditions, including Alzheimer's and Parkinson's diseases. Here, we review essential microglial homeostatic functions regulated by SFKs, including phagocytosis, environmental sensing, and secretion of inflammatory mediators. In addition, we discuss the potential of SFK modulation for microglial homeostasis in Parkinson's and Alzheimer's diseases.
Collapse
Affiliation(s)
- Camila C Portugal
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Portugal
| | - Tiago O Almeida
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Portugal.,Doutoramento em Ciências Biomédicas, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Portugal
| | - Renato Socodato
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Portugal
| | - João B Relvas
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Portugal.,Department of Biomedicine, Faculty of Medicine, University of Porto, Portugal
| |
Collapse
|
22
|
Yi S, Wang L, Wang H, Ho MS, Zhang S. Pathogenesis of α-Synuclein in Parkinson's Disease: From a Neuron-Glia Crosstalk Perspective. Int J Mol Sci 2022; 23:14753. [PMID: 36499080 PMCID: PMC9739123 DOI: 10.3390/ijms232314753] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 11/29/2022] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder. The classical behavioral defects of PD patients involve motor symptoms such as bradykinesia, tremor, and rigidity, as well as non-motor symptoms such as anosmia, depression, and cognitive impairment. Pathologically, the progressive loss of dopaminergic (DA) neurons in the substantia nigra (SN) and the accumulation of α-synuclein (α-syn)-composed Lewy bodies (LBs) and Lewy neurites (LNs) are key hallmarks. Glia are more than mere bystanders that simply support neurons, they actively contribute to almost every aspect of neuronal development and function; glial dysregulation has been implicated in a series of neurodegenerative diseases including PD. Importantly, amounting evidence has added glial activation and neuroinflammation as new features of PD onset and progression. Thus, gaining a better understanding of glia, especially neuron-glia crosstalk, will not only provide insight into brain physiology events but also advance our knowledge of PD pathologies. This review addresses the current understanding of α-syn pathogenesis in PD, with a focus on neuron-glia crosstalk. Particularly, the transmission of α-syn between neurons and glia, α-syn-induced glial activation, and feedbacks of glial activation on DA neuron degeneration are thoroughly discussed. In addition, α-syn aggregation, iron deposition, and glial activation in regulating DA neuron ferroptosis in PD are covered. Lastly, we summarize the preclinical and clinical therapies, especially targeting glia, in PD treatments.
Collapse
Affiliation(s)
| | | | | | - Margaret S. Ho
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Shiping Zhang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| |
Collapse
|
23
|
Maguire E, Connor-Robson N, Shaw B, O’Donoghue R, Stöberl N, Hall-Roberts H. Assaying Microglia Functions In Vitro. Cells 2022; 11:3414. [PMID: 36359810 PMCID: PMC9654693 DOI: 10.3390/cells11213414] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 10/20/2022] [Accepted: 10/25/2022] [Indexed: 08/27/2023] Open
Abstract
Microglia, the main immune modulators of the central nervous system, have key roles in both the developing and adult brain. These functions include shaping healthy neuronal networks, carrying out immune surveillance, mediating inflammatory responses, and disposing of unwanted material. A wide variety of pathological conditions present with microglia dysregulation, highlighting the importance of these cells in both normal brain function and disease. Studies into microglial function in the context of both health and disease thus have the potential to provide tremendous insight across a broad range of research areas. In vitro culture of microglia, using primary cells, cell lines, or induced pluripotent stem cell derived microglia, allows researchers to generate reproducible, robust, and quantifiable data regarding microglia function. A broad range of assays have been successfully developed and optimised for characterizing microglial morphology, mediation of inflammation, endocytosis, phagocytosis, chemotaxis and random motility, and mediation of immunometabolism. This review describes the main functions of microglia, compares existing protocols for measuring these functions in vitro, and highlights common pitfalls and future areas for development. We aim to provide a comprehensive methodological guide for researchers planning to characterise microglial functions within a range of contexts and in vitro models.
Collapse
Affiliation(s)
- Emily Maguire
- UK Dementia Research Institute (UK DRI), School of Medicine, Cardiff University, Cardiff CF10 3AT, UK
| | | | | | | | | | | |
Collapse
|
24
|
Araújo B, Caridade-Silva R, Soares-Guedes C, Martins-Macedo J, Gomes ED, Monteiro S, Teixeira FG. Neuroinflammation and Parkinson's Disease-From Neurodegeneration to Therapeutic Opportunities. Cells 2022; 11:cells11182908. [PMID: 36139483 PMCID: PMC9497016 DOI: 10.3390/cells11182908] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/10/2022] [Accepted: 09/13/2022] [Indexed: 11/17/2022] Open
Abstract
Parkinson’s disease (PD) is the second most prevalent neurodegenerative disorder worldwide. Clinically, it is characterized by a progressive degeneration of dopaminergic neurons (DAn), resulting in severe motor complications. Preclinical and clinical studies have indicated that neuroinflammation can play a role in PD pathophysiology, being associated with its onset and progression. Nevertheless, several key points concerning the neuroinflammatory process in PD remain to be answered. Bearing this in mind, in the present review, we cover the impact of neuroinflammation on PD by exploring the role of inflammatory cells (i.e., microglia and astrocytes) and the interconnections between the brain and the peripheral system. Furthermore, we discuss both the innate and adaptive immune responses regarding PD pathology and explore the gut–brain axis communication and its influence on the progression of the disease.
Collapse
Affiliation(s)
- Bruna Araújo
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s-PT Government Associate Laboratory, 4710-057/4805-017 Braga/Guimarães, Portugal
- Medical and Industrial Biotechnology Laboratory (LABMI), Porto Research, Technology, and Innovation Center (PORTIC), Porto Polytechnic Institute, 4200-375 Porto, Portugal
- I3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - Rita Caridade-Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s-PT Government Associate Laboratory, 4710-057/4805-017 Braga/Guimarães, Portugal
- Medical and Industrial Biotechnology Laboratory (LABMI), Porto Research, Technology, and Innovation Center (PORTIC), Porto Polytechnic Institute, 4200-375 Porto, Portugal
- I3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - Carla Soares-Guedes
- Medical and Industrial Biotechnology Laboratory (LABMI), Porto Research, Technology, and Innovation Center (PORTIC), Porto Polytechnic Institute, 4200-375 Porto, Portugal
- I3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - Joana Martins-Macedo
- Medical and Industrial Biotechnology Laboratory (LABMI), Porto Research, Technology, and Innovation Center (PORTIC), Porto Polytechnic Institute, 4200-375 Porto, Portugal
- I3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - Eduardo D. Gomes
- Medical and Industrial Biotechnology Laboratory (LABMI), Porto Research, Technology, and Innovation Center (PORTIC), Porto Polytechnic Institute, 4200-375 Porto, Portugal
- I3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - Susana Monteiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s-PT Government Associate Laboratory, 4710-057/4805-017 Braga/Guimarães, Portugal
| | - Fábio G. Teixeira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s-PT Government Associate Laboratory, 4710-057/4805-017 Braga/Guimarães, Portugal
- Medical and Industrial Biotechnology Laboratory (LABMI), Porto Research, Technology, and Innovation Center (PORTIC), Porto Polytechnic Institute, 4200-375 Porto, Portugal
- I3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- Correspondence:
| |
Collapse
|
25
|
Chen SH, Han S, Hu CF, Zhou R, Gao Y, Tu D, Gao H, Feng J, Wang Y, Lu RB, Hong JS. Activation of the MAC1-ERK1/2-NOX2 Pathway Is Required for LPS-Induced Sustaining Reactive Microgliosis, Chronic Neuroinflammation and Neurodegeneration. Antioxidants (Basel) 2022; 11:1202. [PMID: 35740099 PMCID: PMC9220294 DOI: 10.3390/antiox11061202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/13/2022] [Accepted: 06/16/2022] [Indexed: 11/24/2022] Open
Abstract
Recent studies suggest that improper resolution of acute neuroinflammation may lead to long-lasting low-grade chronic neuroinflammation and drive progressive neurodegeneration. However, the molecular mechanism underlying the transition from acute to chronic neuroinflammation remains unclear. The main purpose of this study was to search for potential pathways mediating LPS-elicited chronic neuroinflammation and resultant neurodegeneration. Using microglia cultures prepared from C57BL/6J, MAC1-deficient, and MyD88-deficient mice, the initial study showed that activation of TLR-4 is not sufficient for maintaining chronic neuroinflammation despite its essential role in LPS-initiated acute neuroinflammation. Opposite to TLR-4, our studies showed significantly reduced intensity of chronic neuroinflammation, oxidative stress, and progressive loss of nigral dopaminergic neurons in MAC1-deficient neuron/glial cultures or mice stimulated with LPS. Mechanistic studies revealed the essential role ERK1/2 activation in chronic neuroinflammation-elicited neurodegeneration, which was demonstrated by using an ERK1/2 inhibitor in neuron-glial cultures. Taken together, we propose a key role of the MAC1-NOX2-ERK1/2 signaling pathway in the initiation and maintenance of low-grade chronic neuroinflammation. Continuing ERK1/2 phosphorylation and NOX2 activation form a vicious feedforward cycle in microglia to maintain the low-grade neuroinflammation and drive neurodegeneration.
Collapse
Affiliation(s)
- Shih-Heng Chen
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA; (S.H.); (C.-F.H.); (R.Z.); (Y.G.); (D.T.); (Y.W.); (J.-S.H.)
| | - Shuangyu Han
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA; (S.H.); (C.-F.H.); (R.Z.); (Y.G.); (D.T.); (Y.W.); (J.-S.H.)
- Respiratory Department, Tianjin Medical University General Hospital, Tianjin 300052, China;
| | - Chih-Fen Hu
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA; (S.H.); (C.-F.H.); (R.Z.); (Y.G.); (D.T.); (Y.W.); (J.-S.H.)
- Department of Pediatrics, Tri-Service General Hospital, National Defense Medical Center, Taipei 10086, Taiwan
| | - Ran Zhou
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA; (S.H.); (C.-F.H.); (R.Z.); (Y.G.); (D.T.); (Y.W.); (J.-S.H.)
- Respiratory Department, Tianjin Medical University General Hospital, Tianjin 300052, China;
| | - Yun Gao
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA; (S.H.); (C.-F.H.); (R.Z.); (Y.G.); (D.T.); (Y.W.); (J.-S.H.)
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Institute for Brain Sciences, Nanjing University, Nanjing 210061, China;
| | - Dezhen Tu
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA; (S.H.); (C.-F.H.); (R.Z.); (Y.G.); (D.T.); (Y.W.); (J.-S.H.)
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Institute for Brain Sciences, Nanjing University, Nanjing 210061, China;
| | - Huiming Gao
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Institute for Brain Sciences, Nanjing University, Nanjing 210061, China;
| | - Jing Feng
- Respiratory Department, Tianjin Medical University General Hospital, Tianjin 300052, China;
| | - Yubao Wang
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA; (S.H.); (C.-F.H.); (R.Z.); (Y.G.); (D.T.); (Y.W.); (J.-S.H.)
- Institute of Infectious Diseases, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Ru-Band Lu
- Institute of Behavioral Medicine, National Cheng Kung University, Tainan 70101, Taiwan;
| | - Jau-Shyong Hong
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA; (S.H.); (C.-F.H.); (R.Z.); (Y.G.); (D.T.); (Y.W.); (J.-S.H.)
| |
Collapse
|
26
|
Miller SJ, Campbell CE, Jimenez-Corea HA, Wu GH, Logan R. Neuroglial Senescence, α-Synucleinopathy, and the Therapeutic Potential of Senolytics in Parkinson’s Disease. Front Neurosci 2022; 16:824191. [PMID: 35516803 PMCID: PMC9063319 DOI: 10.3389/fnins.2022.824191] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 03/22/2022] [Indexed: 12/02/2022] Open
Abstract
Parkinson’s disease (PD) is the most common movement disorder and the second most prevalent neurodegenerative disease after Alzheimer’s disease. Despite decades of research, there is still no cure for PD and the complicated intricacies of the pathology are still being worked out. Much of the research on PD has focused on neurons, since the disease is characterized by neurodegeneration. However, neuroglia has become recognized as key players in the health and disease of the central nervous system. This review provides a current perspective on the interactive roles that α-synuclein and neuroglial senescence have in PD. The self-amplifying and cyclical nature of oxidative stress, neuroinflammation, α-synucleinopathy, neuroglial senescence, neuroglial chronic activation and neurodegeneration will be discussed. Finally, the compelling role that senolytics could play as a therapeutic avenue for PD is explored and encouraged.
Collapse
Affiliation(s)
- Sean J. Miller
- Pluripotent Diagnostics Corp. (PDx), Molecular Medicine Research Institute, Sunnyvale, CA, United States
| | | | | | - Guan-Hui Wu
- Department of Neurology, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Robert Logan
- Pluripotent Diagnostics Corp. (PDx), Molecular Medicine Research Institute, Sunnyvale, CA, United States
- Department of Biology, Eastern Nazarene College, Quincy, MA, United States
- *Correspondence: Robert Logan,
| |
Collapse
|
27
|
MacLean M, López-Díez R, Vasquez C, Gugger PF, Schmidt AM. Neuronal-glial communication perturbations in murine SOD1 G93A spinal cord. Commun Biol 2022; 5:177. [PMID: 35228715 PMCID: PMC8885678 DOI: 10.1038/s42003-022-03128-y] [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: 06/14/2021] [Accepted: 02/08/2022] [Indexed: 12/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is an incurable disease characterized by proteinaceous aggregate accumulation and neuroinflammation culminating in rapidly progressive lower and upper motor neuron death. To interrogate cell-intrinsic and inter-cell type perturbations in ALS, single-nucleus RNA sequencing was performed on the lumbar spinal cord in the murine ALS model SOD1G93A transgenic and littermate control mice at peri-symptomatic onset stage of disease, age 90 days. This work uncovered perturbed tripartite synapse functions, complement activation and metabolic stress in the affected spinal cord; processes evidenced by cell death and proteolytic stress-associated gene sets. Concomitantly, these pro-damage events in the spinal cord co-existed with dysregulated reparative mechanisms. This work provides a resource of cell-specific niches in the ALS spinal cord and asserts that interwoven dysfunctional neuronal-glial communications mediating neurodegeneration are underway prior to overt disease manifestation and are recapitulated, in part, in the human post-mortem ALS spinal cord. In this paper, single-nucleus RNA sequencing was performed to provide a resource of cell-specific niches in the murine ALS model spinal cord at peri-symptomatic onset stage of disease. The data suggest that dysfunctional neuronal-glial communication occurs prior to disease onset, which is partially recapitulated in human post-mortem ALS spinal cord tissue.
Collapse
Affiliation(s)
- Michael MacLean
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Raquel López-Díez
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Carolina Vasquez
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Paul F Gugger
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Ann Marie Schmidt
- Diabetes Research Program, Department of Medicine, New York University Grossman School of Medicine, New York, NY, 10016, USA.
| |
Collapse
|
28
|
Linard M, Ravier A, Mougué L, Grgurina I, Boutillier AL, Foubert-Samier A, Blanc F, Helmer C. Infectious Agents as Potential Drivers of α-Synucleinopathies. Mov Disord 2022; 37:464-477. [PMID: 35040520 DOI: 10.1002/mds.28925] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 12/10/2021] [Accepted: 12/27/2021] [Indexed: 12/16/2022] Open
Abstract
α-synucleinopathies, encompassing Parkinson's disease, dementia with Lewy bodies, and multiple system atrophy, are devastating neurodegenerative diseases for which available therapeutic options are scarce, mostly because of our limited understanding of their pathophysiology. Although these pathologies are attributed to an intracellular accumulation of the α-synuclein protein in the nervous system with subsequent neuronal loss, the trigger(s) of this accumulation is/are not clearly identified. Among the existing hypotheses, interest in the hypothesis advocating the involvement of infectious agents in the onset of these diseases is renewed. In this article, we aimed to review the ongoing relevant factors favoring and opposing this hypothesis, focusing on (1) the potential antimicrobial role of α-synuclein, (2) potential entry points of pathogens in regard to early symptoms of diverse α-synucleinopathies, (3) pre-existing literature reviews assessing potential associations between infectious agents and Parkinson's disease, (4) original studies assessing these associations for dementia with Lewy bodies and multiple system atrophy (identified through a systematic literature review), and finally (5) potential susceptibility factors modulating the effects of infectious agents on the nervous system. © 2022 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Morgane Linard
- University of Bordeaux, INSERM, Bordeaux Population Health Research Center, UMR U1219, Bordeaux, France
| | - Alix Ravier
- CM2R (Memory Resource and Research Centre), Geriatrics Department, University Hospitals of Strasbourg, Strasbourg, France
| | - Louisa Mougué
- Cognitive-Behavioral Unit and Memory Consultations, Hospital of Sens, Sens, France
| | - Iris Grgurina
- University of Strasbourg, UMR7364 CNRS, LNCA, Strasbourg, France
| | | | - Alexandra Foubert-Samier
- University of Bordeaux, INSERM, Bordeaux Population Health Research Center, UMR U1219, Bordeaux, France.,French Reference Centre for MSA, University Hospital of Bordeaux, Bordeaux, France
| | - Frédéric Blanc
- CM2R (Memory Resource and Research Centre), Geriatrics Department, University Hospitals of Strasbourg, Strasbourg, France.,ICube Laboratory and FMTS (Fédération de Médecine Translationnelle de Strasbourg), Team IMIS, University of Strasbourg, Strasbourg, France
| | - Catherine Helmer
- University of Bordeaux, INSERM, Bordeaux Population Health Research Center, UMR U1219, Bordeaux, France
| |
Collapse
|
29
|
Immunisation with UB-312 in the Thy1SNCA mouse prevents motor performance deficits and oligomeric α-synuclein accumulation in the brain and gut. Acta Neuropathol 2022; 143:55-73. [PMID: 34741635 PMCID: PMC8732825 DOI: 10.1007/s00401-021-02381-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/20/2021] [Accepted: 10/29/2021] [Indexed: 01/13/2023]
Abstract
Alpha synuclein has a key role in the pathogenesis of Parkinson's disease (PD), Dementia with Lewy Bodies (LBD) and Multiple System Atrophy (MSA). Immunotherapies aiming at neutralising toxic αSyn species are being investigated in the clinic as potential disease modifying therapies for PD and other synucleinopathies. In this study, the effects of active immunisation against αSyn with the UB-312 vaccine were investigated in the Thy1SNCA/15 mouse model of PD. Young transgenic and wild-type mice received an immunisation regimen over a period of 6 weeks, then observed for an additional 9 weeks. Behavioural assessment was conducted before immunisation and at 15 weeks after the first dose. UB-312 immunisation prevented the development of motor impairment in the wire test and challenging beam test, which was associated with reduced levels of αSyn oligomers in the cerebral cortex, hippocampus and striatum of Thy1SNCA/15 mice. UB-312 immunotherapy resulted in a significant reduction of theαSyn load in the colon, accompanied by a reduction in enteric glial cell reactivity in the colonic ganglia. Our results demonstrate that immunisation with UB-312 prevents functional deficits and both central and peripheral pathology in Thy1SNCA/15 mice.
Collapse
|
30
|
Villegas L, Nørremølle A, Freude K, Vilhardt F. Nicotinamide Adenine Dinucleotide Phosphate Oxidases Are Everywhere in Brain Disease, but Not in Huntington's Disease? Front Aging Neurosci 2021; 13:736734. [PMID: 34803655 PMCID: PMC8602359 DOI: 10.3389/fnagi.2021.736734] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 10/08/2021] [Indexed: 11/13/2022] Open
Abstract
Huntington's disease (HD) is an inherited neurodegenerative disorder characterized by neuronal loss and tissue atrophy mainly in the striatum and cortex. In the early stages of the disease, impairment of neuronal function, synaptic dysfunction and white matter loss precedes neuronal death itself. Relative to other neurodegenerative diseases such as Alzheimer's and Parkinson's disease and Amyotrophic Lateral Sclerosis, where the effects of either microglia or NADPH oxidases (NOXs) are recognized as important contributors to disease pathogenesis and progression, there is a pronounced lack of information in HD. This information void contrasts with evidence from human HD patients where blood monocytes and microglia are activated well before HD clinical symptoms (PET scans), and the clear signs of oxidative stress and inflammation in post mortem HD brain. Habitually, NOX activity and oxidative stress in the central nervous system (CNS) are equated with microglia, but research of the last two decades has carved out important roles for NOX enzyme function in neurons. Here, we will convey recent information about the function of NOX enzymes in neurons, and contemplate on putative roles of neuronal NOX in HD. We will focus on NOX-produced reactive oxygen species (ROS) as redox signaling molecules in/among neurons, and the specific roles of NOXs in important processes such as neurogenesis and lineage specification, neurite outgrowth and growth cone dynamics, and synaptic plasticity where NMDAR-dependent signaling, and long-term depression/potentiation are redox-regulated phenomena. HD animal models and induced pluripotent stem cell (iPSC) studies have made it clear that the very same physiological processes are also affected in HD, and we will speculate on possible roles for NOX in the pathogenesis and development of disease. Finally, we also take into account the limited information on microglia in HD and relate this to any contribution of NOX enzymes.
Collapse
Affiliation(s)
- Luisana Villegas
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Anne Nørremølle
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Kristine Freude
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Frederik Vilhardt
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
31
|
Neuroinflammation in Parkinson's disease: a meta-analysis of PET imaging studies. J Neurol 2021; 269:2304-2314. [PMID: 34724571 DOI: 10.1007/s00415-021-10877-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/24/2021] [Accepted: 10/25/2021] [Indexed: 10/20/2022]
Abstract
Increasingly, evidence implicates an important role of neuroinflammation in neurodegeneration progression. Yet, brain imaging has not reached a consistent conclusion that neuroinflammation is involved in the pathogenesis of Parkinson's disease (PD). We aimed to review the evidence to quantitatively assess the existence and spatial distribution of neuroinflammation in the brain of PD patients. We systematically searched literature databases for case-control studies which used positron emission tomography to detect neuroinflammation represented by translocator protein (TSPO) levels in PD patients compared with healthy controls (HC). Standardized mean differences (SMD) were selected as effect sizes and random-effects models were used to combine effect sizes. Subgroup analyses for separate brain regions were conducted. Fifteen studies comprising 455 (HC = 198, PD = 238) participants and 19 brain regions were included. Compared to HC, PD patients had elevated TSPO levels in midbrain, putamen, anterior cingulate, posterior cingulate, thalamus, striatum, frontal, temporal, parietal, occipital, cortex, hippocampus, substantia nigra, pons, cerebellum, and caudate when using 1st-generation ligands. TSPO levels were elevated in the midbrain of PD patients when 2nd-generation ligands were used. We discussed the possible explanations of contrasting difference between these outcomes.
Collapse
|
32
|
Dauer Née Joppe K, Tatenhorst L, Caldi Gomes L, Zhang S, Parvaz M, Carboni E, Roser AE, El DeBakey H, Bähr M, Vogel-Mikuš K, Wang Ip C, Becker S, Zweckstetter M, Lingor P. Brain iron enrichment attenuates α-synuclein spreading after injection of preformed fibrils. J Neurochem 2021; 159:554-573. [PMID: 34176164 DOI: 10.1111/jnc.15461] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 06/22/2021] [Accepted: 06/23/2021] [Indexed: 12/12/2022]
Abstract
Regional iron accumulation and α-synuclein (α-syn) spreading pathology within the central nervous system are common pathological findings in Parkinson's disease (PD). Whereas iron is known to bind to α-syn, facilitating its aggregation and regulating α-syn expression, it remains unclear if and how iron also modulates α-syn spreading. To elucidate the influence of iron on the propagation of α-syn pathology, we investigated α-syn spreading after stereotactic injection of α-syn preformed fibrils (PFFs) into the striatum of mouse brains after neonatal brain iron enrichment. C57Bl/6J mouse pups received oral gavage with 60, 120, or 240 mg/kg carbonyl iron or vehicle between postnatal days 10 and 17. At 12 weeks of age, intrastriatal injections of 5-µg PFFs were performed to induce seeding of α-syn aggregates. At 90 days post-injection, PFFs-injected mice displayed long-term memory deficits, without affection of motor behavior. Interestingly, quantification of α-syn phosphorylated at S129 showed reduced α-syn pathology and attenuated spreading to connectome-specific brain regions after brain iron enrichment. Furthermore, PFFs injection caused intrastriatal microglia accumulation, which was alleviated by iron in a dose-dependent way. In primary cortical neurons in a microfluidic chamber model in vitro, iron application did not alter trans-synaptic α-syn propagation, possibly indicating an involvement of non-neuronal cells in this process. Our study suggests that α-syn PFFs may induce cognitive deficits in mice independent of iron. However, a redistribution of α-syn aggregate pathology and reduction of striatal microglia accumulation in the mouse brain may be mediated via iron-induced alterations of the brain connectome.
Collapse
Affiliation(s)
- Karina Dauer Née Joppe
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
- Center for Biostructural Imaging of Neurodegeneration, University Medical Center Goettingen, Goettingen, Germany
| | - Lars Tatenhorst
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
- Center for Biostructural Imaging of Neurodegeneration, University Medical Center Goettingen, Goettingen, Germany
| | - Lucas Caldi Gomes
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
- Center for Biostructural Imaging of Neurodegeneration, University Medical Center Goettingen, Goettingen, Germany
- Department of Neurology, School of Medicine, University Hospital rechts der Isar, Technical University of Munich, Munich, Germany
| | - Shuyu Zhang
- Department of Neurology, School of Medicine, University Hospital rechts der Isar, Technical University of Munich, Munich, Germany
| | - Mojan Parvaz
- Department of Neurology, School of Medicine, University Hospital rechts der Isar, Technical University of Munich, Munich, Germany
| | - Eleonora Carboni
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
| | - Anna-Elisa Roser
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
- Center for Biostructural Imaging of Neurodegeneration, University Medical Center Goettingen, Goettingen, Germany
| | - Hazem El DeBakey
- Department of Neurology, University Hospital of Wuerzburg, Wuerzburg, Germany
| | - Mathias Bähr
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
- Center for Biostructural Imaging of Neurodegeneration, University Medical Center Goettingen, Goettingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Goettingen, Goettingen, Germany
| | - Katarina Vogel-Mikuš
- Biotechnical faculty, University of Ljubljana, Ljubljana, Slovenia
- Jozef Stefan Institute, Ljubljana, Slovenia
| | - Chi Wang Ip
- Department of Neurology, University Hospital of Wuerzburg, Wuerzburg, Germany
| | - Stefan Becker
- Department of NMR Based Structural Biology, Max Planck Institute for Biophysical Chemistry, Goettingen, Germany
| | - Markus Zweckstetter
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Goettingen, Germany
- Research group Mass Spectrometry, Max Planck Institute for Biophysical Chemistry, Goettingen, Germany
| | - Paul Lingor
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
- Center for Biostructural Imaging of Neurodegeneration, University Medical Center Goettingen, Goettingen, Germany
- Department of Neurology, School of Medicine, University Hospital rechts der Isar, Technical University of Munich, Munich, Germany
| |
Collapse
|
33
|
Mechanistic Insight from Preclinical Models of Parkinson's Disease Could Help Redirect Clinical Trial Efforts in GDNF Therapy. Int J Mol Sci 2021; 22:ijms222111702. [PMID: 34769132 PMCID: PMC8583859 DOI: 10.3390/ijms222111702] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 10/23/2021] [Accepted: 10/25/2021] [Indexed: 11/16/2022] Open
Abstract
Parkinson’s disease (PD) is characterized by four pathognomonic hallmarks: (1) motor and non-motor deficits; (2) neuroinflammation and oxidative stress; (3) pathological aggregates of the α-synuclein (α-syn) protein; (4) neurodegeneration of the nigrostriatal system. Recent evidence sustains that the aggregation of pathological α-syn occurs in the early stages of the disease, becoming the first trigger of neuroinflammation and subsequent neurodegeneration. Thus, a therapeutic line aims at striking back α-synucleinopathy and neuroinflammation to impede neurodegeneration. Another therapeutic line is restoring the compromised dopaminergic system using neurotrophic factors, particularly the glial cell-derived neurotrophic factor (GDNF). Preclinical studies with GDNF have provided encouraging results but often lack evaluation of anti-α-syn and anti-inflammatory effects. In contrast, clinical trials have yielded imprecise results and have reported the emergence of severe side effects. Here, we analyze the discrepancy between preclinical and clinical outcomes, review the mechanisms of the aggregation of pathological α-syn, including neuroinflammation, and evaluate the neurorestorative properties of GDNF, emphasizing its anti-α-syn and anti-inflammatory effects in preclinical and clinical trials.
Collapse
|
34
|
Shan FY, Fung KM, Zieneldien T, Kim J, Cao C, Huang JH. Examining the Toxicity of α-Synuclein in Neurodegenerative Disorders. Life (Basel) 2021; 11:life11111126. [PMID: 34833002 PMCID: PMC8621244 DOI: 10.3390/life11111126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/14/2021] [Accepted: 10/19/2021] [Indexed: 12/20/2022] Open
Abstract
Simple Summary Neurodegenerative disorders are complex disorders that display a variety of clinical manifestations. The second-most common neurodegenerative disorder is Parkinson’s disease, and the leading pathological protein of the disorder is considered to be α-synuclein. Nonetheless, α-synuclein accumulation also seems to result in multiple system atrophy and dementia with Lewy bodies. In order to obtain a more proficient understanding in the pathological progression of these synucleinopathies, it is crucial to observe the post-translational modifications of α-synuclein and the conformations of α-synuclein, as well as its role in the dysfunction of cellular pathways. Abstract α-synuclein is considered the main pathological protein in a variety of neurodegenerative disorders, such as Parkinson’s disease, multiple system atrophy, and dementia with Lewy bodies. As of now, numerous studies have been aimed at examining the post-translational modifications of α-synuclein to determine their effects on α-synuclein aggregation, propagation, and oligomerization, as well as the potential cellular pathway dysfunctions caused by α-synuclein, to determine the role of the protein in disease progression. Furthermore, α-synuclein also appears to contribute to the fibrilization of tau and amyloid beta, which are crucial proteins in Alzheimer’s disease, advocating for α-synuclein’s preeminent role in neurodegeneration. Due to this, investigating the mechanisms of toxicity of α-synuclein in neurodegeneration may lead to a more proficient understanding of the timeline progression in neurodegenerative synucleinopathies and could thereby lead to the development of potent targeted therapies.
Collapse
Affiliation(s)
- Frank Y. Shan
- Department of Anatomic Pathology, Baylor Scott & White Medical Center, College of Medicine, Texas A&M University, Temple, TX 76508, USA
- Correspondence: (F.Y.S.); (T.Z.)
| | - Kar-Ming Fung
- Department of Pathology, University of Oklahoma Medical Center, University of Oklahoma, Norman, OK 73019, USA;
| | - Tarek Zieneldien
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33620, USA; (J.K.); (C.C.)
- Correspondence: (F.Y.S.); (T.Z.)
| | - Janice Kim
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33620, USA; (J.K.); (C.C.)
| | - Chuanhai Cao
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, Tampa, FL 33620, USA; (J.K.); (C.C.)
| | - Jason H. Huang
- Department of Neurosurgery, Baylor Scott & White Medical Center, College of Medicine, Texas A&M University, Temple, TX 76508, USA;
| |
Collapse
|
35
|
Pirooznia SK, Rosenthal LS, Dawson VL, Dawson TM. Parkinson Disease: Translating Insights from Molecular Mechanisms to Neuroprotection. Pharmacol Rev 2021; 73:33-97. [PMID: 34663684 DOI: 10.1124/pharmrev.120.000189] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Parkinson disease (PD) used to be considered a nongenetic condition. However, the identification of several autosomal dominant and recessive mutations linked to monogenic PD has changed this view. Clinically manifest PD is then thought to occur through a complex interplay between genetic mutations, many of which have incomplete penetrance, and environmental factors, both neuroprotective and increasing susceptibility, which variably interact to reach a threshold over which PD becomes clinically manifested. Functional studies of PD gene products have identified many cellular and molecular pathways, providing crucial insights into the nature and causes of PD. PD originates from multiple causes and a range of pathogenic processes at play, ultimately culminating in nigral dopaminergic loss and motor dysfunction. An in-depth understanding of these complex and possibly convergent pathways will pave the way for therapeutic approaches to alleviate the disease symptoms and neuroprotective strategies to prevent disease manifestations. This review is aimed at providing a comprehensive understanding of advances made in PD research based on leveraging genetic insights into the pathogenesis of PD. It further discusses novel perspectives to facilitate identification of critical molecular pathways that are central to neurodegeneration that hold the potential to develop neuroprotective and/or neurorestorative therapeutic strategies for PD. SIGNIFICANCE STATEMENT: A comprehensive review of PD pathophysiology is provided on the complex interplay of genetic and environmental factors and biologic processes that contribute to PD pathogenesis. This knowledge identifies new targets that could be leveraged into disease-modifying therapies to prevent or slow neurodegeneration in PD.
Collapse
Affiliation(s)
- Sheila K Pirooznia
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering (S.K.P., V.L.D., T.M.D.), Departments of Neurology (S.K.P., L.S.R., V.L.D., T.M.D.), Departments of Physiology (V.L.D.), Solomon H. Snyder Department of Neuroscience (V.L.D., T.M.D.), Department of Pharmacology and Molecular Sciences (T.M.D.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.); and Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.)
| | - Liana S Rosenthal
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering (S.K.P., V.L.D., T.M.D.), Departments of Neurology (S.K.P., L.S.R., V.L.D., T.M.D.), Departments of Physiology (V.L.D.), Solomon H. Snyder Department of Neuroscience (V.L.D., T.M.D.), Department of Pharmacology and Molecular Sciences (T.M.D.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.); and Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.)
| | - Valina L Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering (S.K.P., V.L.D., T.M.D.), Departments of Neurology (S.K.P., L.S.R., V.L.D., T.M.D.), Departments of Physiology (V.L.D.), Solomon H. Snyder Department of Neuroscience (V.L.D., T.M.D.), Department of Pharmacology and Molecular Sciences (T.M.D.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.); and Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.)
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering (S.K.P., V.L.D., T.M.D.), Departments of Neurology (S.K.P., L.S.R., V.L.D., T.M.D.), Departments of Physiology (V.L.D.), Solomon H. Snyder Department of Neuroscience (V.L.D., T.M.D.), Department of Pharmacology and Molecular Sciences (T.M.D.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.); and Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.)
| |
Collapse
|
36
|
Monocyte markers correlate with immune and neuronal brain changes in REM sleep behavior disorder. Proc Natl Acad Sci U S A 2021; 118:2020858118. [PMID: 33658371 DOI: 10.1073/pnas.2020858118] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Synucleinopathies are neurodegenerative diseases with both central and peripheral immune responses. However, whether the peripheral immune changes occur early in disease and their relation to brain events is yet unclear. Isolated rapid-eye-movement (REM) sleep behavior disorder (iRBD) can precede synucleinopathy-related parkinsonism and provides a prodromal phenotype to study early Parkinson's disease events. In this prospective case-control study, we describe monocytic markers in a cohort of iRBD patients that were associated with the brain-imaging markers of inflammation and neuronal dysfunction. Using 11C-PK11195 positron emission tomography (PET), we previously showed increased immune activation in the substantia nigra of iRBD patients, while 18F-DOPA PET detected reduced putaminal dopaminergic function. Here we describe that patients' blood monocytic cells showed increased expression of CD11b, while HLA-DR expression was decreased compared to healthy controls. The iRBD patients had increased classical monocytes and mature natural killer cells. Remarkably, the levels of expression of Toll-like receptor 4 (TLR4) on blood monocytes in iRBD patients were positively correlated with nigral immune activation measured by 11C-PK11195 PET and negatively correlated with putaminal 18F-DOPA uptake; the opposite was seen for the percentage of CD163+ myeloid cells. This suggesting a deleterious role for TLR4 and, conversely, a protective one for the CD163 expression. We show an association between peripheral blood monocytes and brain immune and dopaminergic changes in a synucleinopathy-related disorder, thus suggesting a cross-talk among periphery and brain during the disease.
Collapse
|
37
|
Ma X, Wang Q, Yuan W, Wang Y, Zhou F, Kang K, Tong X, Liu Z. Electroacupuncture Alleviates Neuroinflammation and Motor Dysfunction by Regulating Intestinal Barrier Function in a Mouse Model of Parkinson Disease. J Neuropathol Exp Neurol 2021; 80:844-855. [PMID: 34343334 DOI: 10.1093/jnen/nlab046] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Gastrointestinal dysfunction is the main nonmotor characteristic of Parkinson disease (PD), manipulation of gastrointestinal function by altering gut-brain axis is a potentially novel entry point for the treatment of PD. Acupuncture has been reported to confer beneficial effects in the gastrointestinal diseases. Therefore, this study aimed to explore the effects and mechanism of acupuncture on the pathophysiology and gastrointestinal function of PD. A PD mouse model was established by rotenone, and electroacupuncture was used to regulate the gastrointestinal function. Rotenone was found to induce the types of brain pathologies and gastrointestinal dysfunction that are similar to those observed with PD. Electroacupuncture significantly increased the spontaneous activity of mice with PD and increased the expression of tyrosine hydroxylase, while reducing the expression of Iba-1 in substantia nigra (SN), suggesting that motor dysfunction and neurological damage was alleviated. In addition, electroacupuncture significantly reduced the deposition of α-synuclein in both colon and SN, reduced intestinal inflammation, and exerted protective effects on enteric nervous system and intestinal barrier. In conclusion, electroacupuncture confers beneficial effects on the gastrointestinal system of mice with PD and can alleviate neuroinflammation and neuropathic injury by inhibiting intestinal inflammation, promoting intestinal barrier repair and reducing α-synuclein deposition in the colon.
Collapse
Affiliation(s)
- Xue Ma
- From the Third College of Clinical Medicine, Zhejiang Chinese Medicine University, Zhejiang, China (XM); College of Acu-Moxibustion and Massage, Shaanxi University of Chinese Medicine, Shaanxi, China (QW, WY, YW, ZL); The Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China (FZ); Second College of Clinical Medicine, Shaanxi University of Chinese Medicine, Shaanxi, China (KK); and Xizang Minzu University, Shaanxi, China (XT)
| | - Qiang Wang
- From the Third College of Clinical Medicine, Zhejiang Chinese Medicine University, Zhejiang, China (XM); College of Acu-Moxibustion and Massage, Shaanxi University of Chinese Medicine, Shaanxi, China (QW, WY, YW, ZL); The Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China (FZ); Second College of Clinical Medicine, Shaanxi University of Chinese Medicine, Shaanxi, China (KK); and Xizang Minzu University, Shaanxi, China (XT)
| | - Wei Yuan
- From the Third College of Clinical Medicine, Zhejiang Chinese Medicine University, Zhejiang, China (XM); College of Acu-Moxibustion and Massage, Shaanxi University of Chinese Medicine, Shaanxi, China (QW, WY, YW, ZL); The Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China (FZ); Second College of Clinical Medicine, Shaanxi University of Chinese Medicine, Shaanxi, China (KK); and Xizang Minzu University, Shaanxi, China (XT)
| | - Yuan Wang
- From the Third College of Clinical Medicine, Zhejiang Chinese Medicine University, Zhejiang, China (XM); College of Acu-Moxibustion and Massage, Shaanxi University of Chinese Medicine, Shaanxi, China (QW, WY, YW, ZL); The Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China (FZ); Second College of Clinical Medicine, Shaanxi University of Chinese Medicine, Shaanxi, China (KK); and Xizang Minzu University, Shaanxi, China (XT)
| | - Feng Zhou
- From the Third College of Clinical Medicine, Zhejiang Chinese Medicine University, Zhejiang, China (XM); College of Acu-Moxibustion and Massage, Shaanxi University of Chinese Medicine, Shaanxi, China (QW, WY, YW, ZL); The Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China (FZ); Second College of Clinical Medicine, Shaanxi University of Chinese Medicine, Shaanxi, China (KK); and Xizang Minzu University, Shaanxi, China (XT)
| | - Kaiwen Kang
- From the Third College of Clinical Medicine, Zhejiang Chinese Medicine University, Zhejiang, China (XM); College of Acu-Moxibustion and Massage, Shaanxi University of Chinese Medicine, Shaanxi, China (QW, WY, YW, ZL); The Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China (FZ); Second College of Clinical Medicine, Shaanxi University of Chinese Medicine, Shaanxi, China (KK); and Xizang Minzu University, Shaanxi, China (XT)
| | - Xiaopeng Tong
- From the Third College of Clinical Medicine, Zhejiang Chinese Medicine University, Zhejiang, China (XM); College of Acu-Moxibustion and Massage, Shaanxi University of Chinese Medicine, Shaanxi, China (QW, WY, YW, ZL); The Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China (FZ); Second College of Clinical Medicine, Shaanxi University of Chinese Medicine, Shaanxi, China (KK); and Xizang Minzu University, Shaanxi, China (XT)
| | - Zhibin Liu
- From the Third College of Clinical Medicine, Zhejiang Chinese Medicine University, Zhejiang, China (XM); College of Acu-Moxibustion and Massage, Shaanxi University of Chinese Medicine, Shaanxi, China (QW, WY, YW, ZL); The Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China (FZ); Second College of Clinical Medicine, Shaanxi University of Chinese Medicine, Shaanxi, China (KK); and Xizang Minzu University, Shaanxi, China (XT)
| |
Collapse
|
38
|
McGregor BA, Schommer J, Guo K, Raihan MO, Ghribi O, Hur J, Porter JE. Alpha-Synuclein-induced DNA Methylation and Gene Expression in Microglia. Neuroscience 2021; 468:186-198. [PMID: 34082066 PMCID: PMC12037068 DOI: 10.1016/j.neuroscience.2021.05.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 05/21/2021] [Accepted: 05/24/2021] [Indexed: 12/18/2022]
Abstract
Synucleinopathy disorders are characterized by aggregates of α-synuclein (α-syn), which engage microglia to elicit a neuroinflammatory response. Here, we determined the gene expression and DNA methylation changes in microglia induced by aggregate α-syn. Transgenic murine Thy-1 promoter (mThy1)-Asyn mice overexpressing human α-syn are a model of synucleinopathy. Microglia from 3 and 13-month-old mice were used to isolate nucleic acids for methylated DNA and RNA-sequencing. α-Syn-regulated changes in gene expression and genomic methylation were determined and examined for functional enrichment followed by network analysis to further elucidate possible connections within the data. Microglial DNA isolated from our 3-month cohort had 5315 differentially methylated gene (DMG) changes, while RNA levels demonstrated a change in 119 differentially expressed genes (DEGs) between mThy1-Asyn mice and wild-type littermate controls. The 3-month DEGs and DMGs were highly associated with adhesion and migration signaling, suggesting a phenotypic transition from resting to active microglia. We observed 3742 DMGs and 3766 DEGs in 13-month mThy1-Asyn mice. These genes were often related to adhesion, migration, cell cycle, cellular metabolism, and immune response. Network analysis also showed increased cell mobility and inflammatory functions at 3 months, shifting to cell cycle, immune response, and metabolism changes at 13 months. We observed significant α-syn-induced methylation and gene expression changes in microglia. Our data suggest that α-syn overexpression initiates microglial activation leading to neuroinflammation and cellular metabolic stresses, which is associated with disease progression.
Collapse
Affiliation(s)
- Brett A McGregor
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202, USA
| | - Jared Schommer
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202, USA
| | - Kai Guo
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202, USA
| | - Md Obayed Raihan
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202, USA
| | - Othman Ghribi
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202, USA
| | - Junguk Hur
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202, USA.
| | - James E Porter
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202, USA.
| |
Collapse
|
39
|
Zheng T, Zhang Z. Activated microglia facilitate the transmission of α-synuclein in Parkinson's disease. Neurochem Int 2021; 148:105094. [PMID: 34097990 DOI: 10.1016/j.neuint.2021.105094] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 05/23/2021] [Accepted: 05/31/2021] [Indexed: 01/31/2023]
Abstract
Parkinson's disease (PD) is characterized by the loss of dopaminergic neurons in the substantia nigra pars compacta and abnormal aggregates of α-synuclein protein called Lewy bodies. To date, there is no drug that can definitely slow down or stop the progression of this disease. The discovery of the cell-to-cell transmission of pathologic α-synuclein seeds offers the possibility to explore novel treatment strategies to prevent the spread of α-synuclein, with the purpose of slowing down the progression of PD in its tracks. Although recent studies have made tremendous progress in understanding how α-synuclein spreads throughout the brain, neuroinflammation seems to play a crucial role in the development of α-synuclein pathology in PD. The activation of microglia, one of the hallmarks of the neuroinflammatory process, is suggested to influence the neuron-to-neuron transmission of α-synuclein. This review summarizes how activated microglia facilitate this process, and focuses on the following mechanisms including the activation of microglia in PD, the reduced ability of activated microglia to clear α-synuclein and increased migratory capacity of microglia in PD, as well as the cooperation between microglia and exosomes in mediating α-synuclein release and propagation. In conclusion, this article help collate information on microglia in-relation to PD.
Collapse
Affiliation(s)
- Tingting Zheng
- Department of Neurology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), 54 Youdian Road, Hangzhou 310006, China
| | - Zhengxiang Zhang
- Department of Neurology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), 54 Youdian Road, Hangzhou 310006, China.
| |
Collapse
|
40
|
Zaman V, Shields DC, Shams R, Drasites KP, Matzelle D, Haque A, Banik NL. Cellular and molecular pathophysiology in the progression of Parkinson's disease. Metab Brain Dis 2021; 36:815-827. [PMID: 33599945 PMCID: PMC8170715 DOI: 10.1007/s11011-021-00689-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 02/10/2021] [Indexed: 12/12/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder etiologically linked to the loss of substantia nigra (SN) dopaminergic neurons in the mid-brain. The etiopathology of sporadic PD is still unclear; however, the interaction of extrinsic and intrinsic factors may play a critical role in the onset and progression of the disease. Studies in animal models and human post-mortem tissue have identified distinct cellular and molecular changes in the diseased brain, suggesting complex interactions between different glial cell types and various molecular pathways. Small changes in the expression of specific genes in a single pathway or cell type possibly influence others at the cellular and system levels. These molecular and cellular signatures like neuroinflammation, oxidative stress, and autophagy have been observed in PD patients' brain tissue. While the etiopathology of PD is still poorly understood, the interplay between glial cells and molecular events may play a crucial role in disease onset and progression.
Collapse
Affiliation(s)
- Vandana Zaman
- Ralph H. Johnson Veterans Administration Medical Center, 109 Bee St, Charleston, SC, 29401, USA
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas St., Suite 301, Charleston, SC, 29425, USA
| | - Donald C Shields
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas St., Suite 301, Charleston, SC, 29425, USA
| | - Ramsha Shams
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas St., Suite 301, Charleston, SC, 29425, USA
- Department of Health and Human Performance, The Citadel, 171 Moultrie St, Charleston, SC, 29409, USA
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC, 29425, USA
| | - Kelsey P Drasites
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas St., Suite 301, Charleston, SC, 29425, USA
- Department of Health and Human Performance, The Citadel, 171 Moultrie St, Charleston, SC, 29409, USA
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC, 29425, USA
| | - Denise Matzelle
- Ralph H. Johnson Veterans Administration Medical Center, 109 Bee St, Charleston, SC, 29401, USA
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC, 29425, USA
| | - Azizul Haque
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC, 29425, USA.
| | - Narendra L Banik
- Ralph H. Johnson Veterans Administration Medical Center, 109 Bee St, Charleston, SC, 29401, USA.
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas St., Suite 301, Charleston, SC, 29425, USA.
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC, 29425, USA.
| |
Collapse
|
41
|
Mavroeidi P, Xilouri M. Neurons and Glia Interplay in α-Synucleinopathies. Int J Mol Sci 2021; 22:4994. [PMID: 34066733 PMCID: PMC8125822 DOI: 10.3390/ijms22094994] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/29/2021] [Accepted: 05/04/2021] [Indexed: 11/16/2022] Open
Abstract
Accumulation of the neuronal presynaptic protein alpha-synuclein within proteinaceous inclusions represents the key histophathological hallmark of a spectrum of neurodegenerative disorders, referred to by the umbrella term a-synucleinopathies. Even though alpha-synuclein is expressed predominantly in neurons, pathological aggregates of the protein are also found in the glial cells of the brain. In Parkinson's disease and dementia with Lewy bodies, alpha-synuclein accumulates mainly in neurons forming the Lewy bodies and Lewy neurites, whereas in multiple system atrophy, the protein aggregates mostly in the glial cytoplasmic inclusions within oligodendrocytes. In addition, astrogliosis and microgliosis are found in the synucleinopathy brains, whereas both astrocytes and microglia internalize alpha-synuclein and contribute to the spread of pathology. The mechanisms underlying the pathological accumulation of alpha-synuclein in glial cells that under physiological conditions express low to non-detectable levels of the protein are an area of intense research. Undoubtedly, the presence of aggregated alpha-synuclein can disrupt glial function in general and can contribute to neurodegeneration through numerous pathways. Herein, we summarize the current knowledge on the role of alpha-synuclein in both neurons and glia, highlighting the contribution of the neuron-glia connectome in the disease initiation and progression, which may represent potential therapeutic target for a-synucleinopathies.
Collapse
Affiliation(s)
| | - Maria Xilouri
- Center of Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece;
| |
Collapse
|
42
|
Greenwood EK, Brown DR. Senescent Microglia: The Key to the Ageing Brain? Int J Mol Sci 2021; 22:4402. [PMID: 33922383 PMCID: PMC8122783 DOI: 10.3390/ijms22094402] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 12/13/2022] Open
Abstract
Ageing represents the single biggest risk factor for development of neurodegenerative disease. Despite being such long-lived cells, microglia have been relatively understudied for their role in the ageing process. Reliably identifying aged microglia has proven challenging, not least due to the diversity of cell populations, and the limitations of available models, further complicated by differences between human and rodent cells. Consequently, the literature contains multiple descriptions and categorisations of microglia with neurotoxic phenotypes, including senescence, without any unifying markers. The role of microglia in brain homeostasis, particularly iron storage and metabolism, may provide a key to reliable identification.
Collapse
Affiliation(s)
| | - David R. Brown
- Department of Biology and Biochemistry, University of Bath, Bath BA2 7AY, UK;
| |
Collapse
|
43
|
Maurya SK, Bhattacharya N, Mishra S, Bhattacharya A, Banerjee P, Senapati S, Mishra R. Microglia Specific Drug Targeting Using Natural Products for the Regulation of Redox Imbalance in Neurodegeneration. Front Pharmacol 2021; 12:654489. [PMID: 33927630 PMCID: PMC8076853 DOI: 10.3389/fphar.2021.654489] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 03/08/2021] [Indexed: 12/14/2022] Open
Abstract
Microglia, a type of innate immune cell of the brain, regulates neurogenesis, immunological surveillance, redox imbalance, cognitive and behavioral changes under normal and pathological conditions like Alzheimer's, Parkinson's, Multiple sclerosis and traumatic brain injury. Microglia produces a wide variety of cytokines to maintain homeostasis. It also participates in synaptic pruning and regulation of neurons overproduction by phagocytosis of neural precursor cells. The phenotypes of microglia are regulated by the local microenvironment of neurons and astrocytes via interaction with both soluble and membrane-bound mediators. In case of neuron degeneration as observed in acute or chronic neurodegenerative diseases, microglia gets released from the inhibitory effect of neurons and astrocytes, showing activated phenotype either of its dual function. Microglia shows neuroprotective effect by secreting growths factors to heal neurons and clears cell debris through phagocytosis in case of a moderate stimulus. But the same microglia starts releasing pro-inflammatory cytokines like TNF-α, IFN-γ, reactive oxygen species (ROS), and nitric oxide (NO), increasing neuroinflammation and redox imbalance in the brain under chronic signals. Therefore, pharmacological targeting of microglia would be a promising strategy in the regulation of neuroinflammation, redox imbalance and oxidative stress in neurodegenerative diseases. Some studies present potentials of natural products like curcumin, resveratrol, cannabidiol, ginsenosides, flavonoids and sulforaphane to suppress activation of microglia. These natural products have also been proposed as effective therapeutics to regulate the progression of neurodegenerative diseases. The present review article intends to explain the molecular mechanisms and functions of microglia and molecular dynamics of microglia specific genes and proteins like Iba1 and Tmem119 in neurodegeneration. The possible interventions by curcumin, resveratrol, cannabidiol, ginsenosides, flavonoids and sulforaphane on microglia specific protein Iba1 suggest possibility of natural products mediated regulation of microglia phenotypes and its functions to control redox imbalance and neuroinflammation in management of Alzheimer's, Parkinson's and Multiple Sclerosis for microglia-mediated therapeutics.
Collapse
Affiliation(s)
| | - Neetu Bhattacharya
- Department of Zoology, Dyal Singh College, University of Delhi, Delhi, India
| | - Suman Mishra
- Department of Molecular Medicine and Biotechnology, SGPGI, Lucknow, India
| | - Amit Bhattacharya
- Department of Zoology, Ramjas College, University of Delhi, Delhi, India
| | - Pratibha Banerjee
- Immunogenomics Laboratory, Department of Human Genetics & Molecular Medicine, Central University of Punjab, Bathinda, India
| | - Sabyasachi Senapati
- Immunogenomics Laboratory, Department of Human Genetics & Molecular Medicine, Central University of Punjab, Bathinda, India
| | - Rajnikant Mishra
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Banaras Hindu University, Varanasi, India
| |
Collapse
|
44
|
Hu CF, Wu SP, Lin GJ, Shieh CC, Hsu CS, Chen JW, Chen SH, Hong JS, Chen SJ. Microglial Nox2 Plays a Key Role in the Pathogenesis of Experimental Autoimmune Encephalomyelitis. Front Immunol 2021; 12:638381. [PMID: 33868265 PMCID: PMC8050344 DOI: 10.3389/fimmu.2021.638381] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 03/12/2021] [Indexed: 12/12/2022] Open
Abstract
While oxidative stress has been linked to multiple sclerosis (MS), the role of superoxide-producing phagocyte NADPH oxidase (Nox2) in central nervous system (CNS) pathogenesis remains unclear. This study investigates the impact of Nox2 gene ablation on pro- and anti-inflammatory cytokine and chemokine production in a mouse experimental autoimmune encephalomyelitis (EAE) model. Nox2 deficiency attenuates EAE-induced neural damage and reduces disease severity, pathogenic immune cells infiltration, demyelination, and oxidative stress in the CNS. The number of autoreactive T cells, myeloid cells, and activated microglia, as well as the production of cytokines and chemokines, including GM-CSF, IFNγ, TNFα, IL-6, IL-10, IL-17A, CCL2, CCL5, and CXCL10, were much lower in the Nox2-/- CNS tissues but remained unaltered in the peripheral lymphoid organs. RNA-seq profiling of microglial transcriptome identified a panel of Nox2 dependent proinflammatory genes: Pf4, Tnfrsf9, Tnfsf12, Tnfsf13, Ccl7, Cxcl3, and Cxcl9. Furthermore, gene ontology and pathway enrichment analyses revealed that microglial Nox2 plays a regulatory role in multiple pathways known to be important for MS/EAE pathogenesis, including STAT3, glutathione, leukotriene biosynthesis, IL-8, HMGB1, NRF2, systemic lupus erythematosus in B cells, and T cell exhaustion signaling. Taken together, our results provide new insights into the critical functions performed by microglial Nox2 during the EAE pathogenesis, suggesting that Nox2 inhibition may represent an important therapeutic target for MS.
Collapse
Affiliation(s)
- Chih-Fen Hu
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan.,Department of Pediatrics, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - San-Pin Wu
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| | - Gu-Jiun Lin
- Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
| | - Chi-Chang Shieh
- Institute of Clinical Medicine, National Cheng Kung University College of Medicine, Tainan, Taiwan
| | - Chih-Sin Hsu
- Genomics Center for Clinical and Biotechnological Applications of Cancer Progression Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Jing-Wun Chen
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Shih-Heng Chen
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| | - Jau-Shyong Hong
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, United States
| | - Shyi-Jou Chen
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan.,Department of Pediatrics, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.,Department of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
45
|
Kakoty V, K C S, Dubey SK, Yang CH, Kesharwani P, Taliyan R. The gut-brain connection in the pathogenicity of Parkinson disease: Putative role of autophagy. Neurosci Lett 2021; 753:135865. [PMID: 33812929 DOI: 10.1016/j.neulet.2021.135865] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 03/13/2021] [Accepted: 03/25/2021] [Indexed: 01/09/2023]
Abstract
Parkinson disease (PD) is a progressive movement functionality disorder resulting in tremor and inability to execute voluntary functions combined with the preponderant non-motor disturbances encompassing constipation and gastrointestinal irritation. Despite continued research, the pathogenesis of PD is not yet clear. The available class of drugs for effective symptomatic management of PD includes a combination of levodopa and carbidopa. In recent past, the link between gut with PD has been explored. According to recent preclinical evidence, pathogens such as virus or bacterium may initiate entry into the gut via the nasal cavity that may aggravate lewy pathology in the gut that eventually propagates and progresses towards the brain via the vagus nerve resulting in the prodromal non-motor symptoms. Additionally, experimental evidence also suggests that alpha-synuclein misfolding commences at a very early stage in the gut and is transported via the vagus nerve prior to seeding PD pathology in the brain. However, this progression and resultant deterioration of the neurones can effectively be altered by an autophagy inducer, Trehalose, although the mechanism behind it is still enigmatic. Hence, this review will mainly focus on analysing the basic components of the gut that might be responsible for aggravating lewy pathology, the mediator(s) responsible for transmission of PD pathology from gut to brain and the important role of trehalose in ameliorating gut dysbiosis related PD complications that would eventually pave the way for therapeutic management of PD.
Collapse
Affiliation(s)
- Violina Kakoty
- Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science, Pilani, India
| | - Sarathlal K C
- Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science, Pilani, India
| | - Sunil Kumar Dubey
- R&D Healthcare Division, Emami Ltd, Kolkatta, India; Department of Pharmacy, Birla Institute of Technology and Science, Pilani, Pilani Campus, Rajasthan, 333031, India
| | - Chih Hao Yang
- Department of Pharmacology, Taipei Medical University, Taiwan
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India.
| | - Rajeev Taliyan
- Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science, Pilani, India.
| |
Collapse
|
46
|
Xia Y, Zhang G, Kou L, Yin S, Han C, Hu J, Wan F, Sun Y, Wu J, Li Y, Huang J, Xiong N, Zhang Z, Wang T. Reactive microglia enhance the transmission of exosomal alpha-synuclein via toll-like receptor 2. Brain 2021; 144:2024-2037. [PMID: 33792662 DOI: 10.1093/brain/awab122] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 02/14/2021] [Accepted: 03/08/2021] [Indexed: 01/16/2023] Open
Abstract
Increasing evidence suggests that microglial activation is strongly linked to the initiation and progression of Parkinson's disease (PD). Cell-to-cell propagation of α-synuclein (α-syn) pathology is a highlighted feature of PD, and the focus of such research has been primarily on neurons. However, recent studies as well as the data contained herein suggest that microglia, the primary phagocytes in the brain, play a direct role in the spread of α-syn pathology. Recent data revealed that plasma exosomes derived from PD patients (PD-EXO) carry pathological α-syn and target microglia preferentially. Hence, PD-EXO is likely a key tool for investigating the role of microglia in α-syn transmission. We showed that intrastriatal injection of PD-EXO resulted in the propagation of exosomal α-syn from microglia to neurons following microglia activation. Toll-like receptor 2 (TLR2) in microglia was activated by exosomal α-syn and acted as a crucial mediator of PD-EXO-induced microglial activation. Additionally, partial microglia depletion resulted in a significant decrease of exogenous α-syn in the substantia nigra (SN). Furthermore, exosomal α-syn internalization was initiated by binding to TLR2 of microglia. Excessive α-syn phagocytosis may induce the inflammatory responses of microglia and provide the seed for microglia-to-neuron transmission. Consistently, TLR2 silencing in microglia mitigated α-syn pathology in vivo. Overall, the present data support the idea that the interaction of exosomal α-syn and microglial TLR2 contribute to excessive α-syn phagocytosis and microglial activation, which lead to the further propagation and spread of α-syn pathology, thereby highlighting the pivotal roles of reactive microglia in α-syn transmission.
Collapse
Affiliation(s)
- Yun Xia
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guoxin Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Liang Kou
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sijia Yin
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chao Han
- Department of Neurology, Anhui Provincial Hospital, The First Affiliated Hospital of Science and Technology of China, Hefei, China
| | - Junjie Hu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fang Wan
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yadi Sun
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiawei Wu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yunna Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinsha Huang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Nian Xiong
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Tao Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
47
|
Harms AS, Ferreira SA, Romero-Ramos M. Periphery and brain, innate and adaptive immunity in Parkinson's disease. Acta Neuropathol 2021; 141:527-545. [PMID: 33555429 PMCID: PMC7952334 DOI: 10.1007/s00401-021-02268-5] [Citation(s) in RCA: 173] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 12/29/2020] [Accepted: 01/18/2021] [Indexed: 12/21/2022]
Abstract
Parkinson’s disease (PD) is a neurodegenerative disorder where alpha-synuclein plays a central role in the death and dysfunction of neurons, both, in central, as well as in the peripheral nervous system. Besides the neuronal events observed in patients, PD also includes a significant immune component. It is suggested that the PD-associated immune response will have consequences on neuronal health, thus opening immunomodulation as a potential therapeutic strategy in PD. The immune changes during the disease occur in the brain, involving microglia, but also in the periphery with changes in cells of the innate immune system, particularly monocytes, as well as those of adaptive immunity, such as T-cells. This realization arises from multiple patient studies, but also from data in animal models of the disease, providing strong evidence for innate and adaptive immune system crosstalk in the central nervous system and periphery in PD. Here we review the data showing that alpha-synuclein plays a crucial role in the activation of the innate and adaptive immune system. We will also describe the studies suggesting that inflammation in PD includes early changes in innate and adaptive immune cells that develop dynamically through time during disease, contributing to neuronal degeneration and symptomatology in patients. This novel finding has contributed to the definition of PD as a multisystem disease that should be approached in a more integratory manner rather than a brain-focused classical approach.
Collapse
Affiliation(s)
- Ashley S Harms
- Department of Neurology and Center for Neurodegeneration and Experimental Therapeutics, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sara A Ferreira
- Department of Biomedicine and CNS Disease Modelling Group, Aarhus University, Høegh-Guldbergsgade 10, 8000C, Aarhus, Denmark
| | - Marina Romero-Ramos
- Department of Biomedicine and CNS Disease Modelling Group, Aarhus University, Høegh-Guldbergsgade 10, 8000C, Aarhus, Denmark.
| |
Collapse
|
48
|
Ernest James Phillips T, Maguire E. Phosphoinositides: Roles in the Development of Microglial-Mediated Neuroinflammation and Neurodegeneration. Front Cell Neurosci 2021; 15:652593. [PMID: 33841102 PMCID: PMC8032904 DOI: 10.3389/fncel.2021.652593] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 03/08/2021] [Indexed: 12/11/2022] Open
Abstract
Microglia are increasingly recognized as vital players in the pathology of a variety of neurodegenerative conditions including Alzheimer’s (AD) and Parkinson’s (PD) disease. While microglia have a protective role in the brain, their dysfunction can lead to neuroinflammation and contributes to disease progression. Also, a growing body of literature highlights the seven phosphoinositides, or PIPs, as key players in the regulation of microglial-mediated neuroinflammation. These small signaling lipids are phosphorylated derivates of phosphatidylinositol, are enriched in the brain, and have well-established roles in both homeostasis and disease.Disrupted PIP levels and signaling has been detected in a variety of dementias. Moreover, many known AD disease modifiers identified via genetic studies are expressed in microglia and are involved in phospholipid metabolism. One of these, the enzyme PLCγ2 that hydrolyzes the PIP species PI(4,5)P2, displays altered expression in AD and PD and is currently being investigated as a potential therapeutic target.Perhaps unsurprisingly, neurodegenerative conditions exhibiting PIP dyshomeostasis also tend to show alterations in aspects of microglial function regulated by these lipids. In particular, phosphoinositides regulate the activities of proteins and enzymes required for endocytosis, toll-like receptor signaling, purinergic signaling, chemotaxis, and migration, all of which are affected in a variety of neurodegenerative conditions. These functions are crucial to allow microglia to adequately survey the brain and respond appropriately to invading pathogens and other abnormalities, including misfolded proteins. AD and PD therapies are being developed to target many of the above pathways, and although not yet investigated, simultaneous PIP manipulation might enhance the beneficial effects observed. Currently, only limited therapeutics are available for dementia, and although these show some benefits for symptom severity and progression, they are far from curative. Given the importance of microglia and PIPs in dementia development, this review summarizes current research and asks whether we can exploit this information to design more targeted, or perhaps combined, dementia therapeutics. More work is needed to fully characterize the pathways discussed in this review, but given the strength of the current literature, insights in this area could be invaluable for the future of neurodegenerative disease research.
Collapse
Affiliation(s)
| | - Emily Maguire
- UK Dementia Research Institute at Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
49
|
Kakoty V, K C S, Dubey SK, Yang CH, Kesharwani P, Taliyan R. Lentiviral mediated gene delivery as an effective therapeutic approach for Parkinson disease. Neurosci Lett 2021; 750:135769. [PMID: 33636285 DOI: 10.1016/j.neulet.2021.135769] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/07/2021] [Accepted: 02/19/2021] [Indexed: 11/28/2022]
Abstract
Continual strategies to devise a complete therapeutic cure for neurodegenerative conditions has been a challenge, majorly due to the presence of blood brain barrier. Lack of targeted delivery in order to minimize loss of dopamine (DA) neurones has been a major challenge to overcome anomalies in Parkinson Disease (PD). PD is a neuromotor degenerative disorder deteriorating motor coordination in affected individuals. Recent research has highlighted the use of lentiviral vectors (LVs) for selective delivery of neuroprotective substance for complete halt of disease progression in PD. LVs have the ability to infect both dividing and non-dividing cells along with non-encoding capability of viral protein that might elicit an immune response. This review will mainly focus on understanding the basic mechanism of action of LVs and its therapeutic aid in PD.
Collapse
Affiliation(s)
- Violina Kakoty
- Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science, Pilani, India
| | - Sarathlal K C
- Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science, Pilani, India
| | - Sunil Kumar Dubey
- Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science, Pilani, India; R&D Healthcare Division, Emami Ltd, Kolkatta, India
| | - Chih Hao Yang
- Department of Pharmacology, Taipei Medical University, Taiwan
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India.
| | - Rajeev Taliyan
- Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science, Pilani, India.
| |
Collapse
|
50
|
Bogale TA, Faustini G, Longhena F, Mitola S, Pizzi M, Bellucci A. Alpha-Synuclein in the Regulation of Brain Endothelial and Perivascular Cells: Gaps and Future Perspectives. Front Immunol 2021; 12:611761. [PMID: 33679750 PMCID: PMC7933041 DOI: 10.3389/fimmu.2021.611761] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 01/27/2021] [Indexed: 12/27/2022] Open
Abstract
Misfolded proteins, inflammation, and vascular alterations are common pathological hallmarks of neurodegenerative diseases. Alpha-synuclein is a small synaptic protein that was identified as a major component of Lewy bodies and Lewy neurites in the brain of patients affected by Parkinson's disease (PD), Lewy body dementia (LBD), and other synucleinopathies. It is mainly involved in the regulation of synaptic vesicle trafficking but can also control mitochondrial/endoplasmic reticulum (ER) homeostasis, lysosome/phagosome function, and cytoskeleton organization. Recent evidence supports that the pathological forms of α-synuclein can also reduce the release of vasoactive and inflammatory mediators from endothelial cells (ECs) and modulates the expression of tight junction (TJ) proteins important for maintaining the blood-brain barrier (BBB). This hints that α-synuclein deposition can affect BBB integrity. Border associated macrophages (BAMs) are brain resident macrophages found in association with the vasculature (PVMs), meninges (MAMs), and choroid plexus (CPMs). Recent findings indicate that these cells play distinct roles in stroke and neurodegenerative disorders. Although many studies have addressed how α-synuclein may modulate microglia, its effect on BAMs has been scarcely investigated. This review aims at summarizing the main findings supporting how α-synuclein can affect ECs and/or BAMs function as well as their interplay and effect on other cells in the brain perivascular environment in physiological and pathological conditions. Gaps of knowledge and new perspectives on how this protein can contribute to neurodegeneration by inducing BBB homeostatic changes in different neurological conditions are highlighted.
Collapse
Affiliation(s)
- Tizibt Ashine Bogale
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Gaia Faustini
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Francesca Longhena
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Stefania Mitola
- Biotechnology Division, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- Laboratory for Preventive and Personalized Medicine, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Marina Pizzi
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Arianna Bellucci
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- Laboratory for Preventive and Personalized Medicine, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| |
Collapse
|