1
|
Yin S, Chi X, Wan F, Li Y, Zhou Q, Kou L, Sun Y, Wu J, Zou W, Wang Y, Jin Z, Huang J, Xiong N, Xia Y, Wang T. TREM2 signaling in Parkinson's disease: Regulation of microglial function and α-synuclein pathology. Int Immunopharmacol 2024; 143:113446. [PMID: 39490141 DOI: 10.1016/j.intimp.2024.113446] [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: 07/31/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Parkinson's disease (PD) is characterized by the loss of dopaminergic neurons, abnormal accumulation of α-synuclein (α-syn), and microglial activation. Triggering receptor expressed on myeloid cells 2 (TREM2) regulates multiple functions of microglia in the brain, and several studies have shown that TREM2 variant R47H is a risk factor for PD. However, the regulation of microglia by TREM2 in PD remains poorly understood. METHODS We constructed PD cell and animal models using α-syn preformed fibrils. siRNA knockdown and lentiviral overexpression were used to perturb TREM2 levels in cells, and TREM2 knockout mice and lentiviral overexpression was used in animal models to investigate the effects of TREM2 on microglial function, α-syn-related pathology, and dopaminergic neuron degeneration. RESULTS Microglia phagocytosed α-syn preformed fibrils in a concentration- and time-dependent manner, with some capacity to degrade α-syn aggregates. TREM2 expression increased in PD. In the context of PD, TREM2 knockout mice exhibited worsened pathological α-syn spread, decreased microglial reactivity, and increased loss of TH-positive neurons in the substantia nigra compared to wild-type mice. TREM2 overexpression enhanced reactive microglial aggregation towards the pathological site. Cellular experiments revealed that reduced TREM2 impaired microglial phagocytosis and proliferation, but enhanced autophagy via the PI3K/AKT/mTOR pathway. CONCLUSION TREM2 signaling in PD maintains microglial phagocytosis, proliferation, and reactivity, stabilizing autophagy and proliferation via the PI3K/AKT/mTOR pathway. Regulating TREM2 levels may be beneficial in PD treatment.
Collapse
Affiliation(s)
- Sijia Yin
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Xiaosa Chi
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Fang Wan
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Yunna Li
- Department of Neurology, The Central Hospital of Wuhan, 26 Shengli Street, Wuhan 430014, China
| | - Qiulu Zhou
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Liang Kou
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Yadi Sun
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Jiawei Wu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Wenkai Zou
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Yiming Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Zongjie Jin
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Jinsha Huang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Nian Xiong
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Yun Xia
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China.
| | - Tao Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China.
| |
Collapse
|
2
|
Trelle AN, Young CB, Vossler H, Ramos Benitez J, Cody KA, Mendiola JH, Swarovski MS, Guen YL, Feinstein I, Butler RR, Channappa D, Romero A, Park J, Shahid-Besanti M, Corso NK, Chau K, Smith AN, Skylar-Scott I, Yutsis MV, Fredericks CA, Tian L, Younes K, Kerchner GA, Deutsch GK, Davidzon GA, Sha SJ, Henderson VW, Longo FM, Greicius MD, Wyss-Coray T, Andreasson KI, Poston KL, Wagner AD, Mormino EC, Wilson EN. Plasma Aβ 42/Aβ 40 is sensitive to early cerebral amyloid accumulation and predicts risk of cognitive decline across the Alzheimer's disease spectrum. Alzheimers Dement 2024. [PMID: 39713875 DOI: 10.1002/alz.14442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 10/24/2024] [Accepted: 10/27/2024] [Indexed: 12/24/2024]
Abstract
INTRODUCTION The availability of amyloid beta (Aβ) targeting therapies for Alzheimer's disease (AD) is increasing the demand for scalable biomarkers that are sensitive to early cerebral Aβ accumulation. METHODS We evaluated fully-automated Lumipulse plasma Aβ42/Aβ40 immunoassays for detecting cerebral Aβ in 457 clinically unimpaired (CU) and clinically impaired (CI) Stanford Alzheimer's Disease Research Center (Stanford ADRC) participants and 186 CU in the Stanford Aging and Memory Study (SAMS). Longitudinal change in ADRC plasma Aβ42/Aβ40 and cognition and cross-sectional associations with SAMS memory and tau positron emission tomography (PET) were examined. RESULTS Plasma Aβ42/Aβ40 exhibited high performance in detecting amyloid positivity defined by PET (area under the curve [AUC]: 0.885, 95% confidence interval [CI]: 0.816-0.955). Once abnomal, plasma Aβ42/Aβ40 remained low and predicted cognitive decline in both CU and CI individuals. Among SAMS CU, plasma Aβ42/Aβ40 was associated with poorer hippocampal-dependent memory and elevated tau accumulation. DISCUSSION Lumipulse plasma Aβ42/Aβ40 is a scalable assay for detection of cerebral Aβ and prediction of risk for cognitive decline across the AD continuum. HIGHLIGHTS Lumipulse plasma amyloid beta (Aβ)42/Aβ40 exhibited high accuracy in detecting amyloid positivity. Plasma amyloid-positive (Aβ+) individuals exhibited stability of Aβ42/Aβ40 over time. Plasma Aβ42/Aβ40 predicted future cognitive decline across the Alzheimer's disease (AD) spectrum. Plasma Aβ42/Aβ40 was sensitive to memory and tau burden in clinically unimpaired older adults.
Collapse
Affiliation(s)
- Alexandra N Trelle
- Neurology & Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
| | - Christina B Young
- Neurology & Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
| | - Hillary Vossler
- Neurology & Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
| | - Javier Ramos Benitez
- Neurology & Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
| | - Karly A Cody
- Neurology & Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
| | - Justin H Mendiola
- Neurology & Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
| | - Michelle S Swarovski
- Neurology & Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
| | - Yann Le Guen
- Neurology & Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
- Quantitative Sciences Unit, Department of Medicine, Stanford University, Palo Alto, California, USA
| | - Igor Feinstein
- Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, California, USA
| | - Robert R Butler
- Neurology & Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
| | - Divya Channappa
- Neurology & Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
| | - America Romero
- Neurology & Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
| | - Jennifer Park
- Neurology & Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
| | - Marian Shahid-Besanti
- Neurology & Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
| | - Nicole K Corso
- Neurology & Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
| | - Kelly Chau
- Neurology & Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
| | - Amanda N Smith
- Neurology & Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
| | - Irina Skylar-Scott
- Neurology & Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
| | - Maya V Yutsis
- Neurology & Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
| | | | - Lu Tian
- Biomedical Data Science and Statistics, Stanford University, Stanford, California, USA
| | - Kyan Younes
- Neurology & Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
| | - Geoffrey A Kerchner
- Pharma Research and Early Development, F. Hoffmann-La Roche, Ltd., Basel, Switzerland
| | - Gayle K Deutsch
- Neurology & Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
| | | | - Sharon J Sha
- Neurology & Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, California, USA
| | - Victor W Henderson
- Neurology & Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, California, USA
- Department of Epidemiology & Population Health, Stanford University, Stanford, California, USA
| | - Frank M Longo
- Neurology & Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, California, USA
- The Phil and Penny Knight Initiative for Brain Resilience, Stanford University, Stanford, California, USA
| | - Michael D Greicius
- Neurology & Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, California, USA
- The Phil and Penny Knight Initiative for Brain Resilience, Stanford University, Stanford, California, USA
| | - Tony Wyss-Coray
- Neurology & Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, California, USA
- The Phil and Penny Knight Initiative for Brain Resilience, Stanford University, Stanford, California, USA
| | - Katrin I Andreasson
- Neurology & Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, California, USA
- The Phil and Penny Knight Initiative for Brain Resilience, Stanford University, Stanford, California, USA
| | - Kathleen L Poston
- Neurology & Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, California, USA
- The Phil and Penny Knight Initiative for Brain Resilience, Stanford University, Stanford, California, USA
- Neurosurgery, Stanford University, Stanford, California, USA
| | - Anthony D Wagner
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, California, USA
- Psychology, Stanford University, Stanford, California, USA
| | - Elizabeth C Mormino
- Neurology & Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, California, USA
| | - Edward N Wilson
- Neurology & Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, California, USA
- The Phil and Penny Knight Initiative for Brain Resilience, Stanford University, Stanford, California, USA
| |
Collapse
|
3
|
Winer JR, Vossler H, Young CB, Smith V, Romero A, Shahid-Besanti M, Abdelnour C, Wilson EN, Anders D, Pacheco Morales A, Andreasson KI, Yutsis MV, Henderson VW, Davidzon GA, Mormino EC, Poston KL. 18F-PI-2620 Tau PET is associated with cognitive and motor impairment in Lewy body disease. Brain Commun 2024; 7:fcae458. [PMID: 39741783 PMCID: PMC11686406 DOI: 10.1093/braincomms/fcae458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 11/22/2024] [Accepted: 12/17/2024] [Indexed: 01/03/2025] Open
Abstract
Co-pathology is frequent in Lewy body disease, which includes clinical diagnoses of both Parkinson's disease and dementia with Lewy bodies. Measuring concomitant pathology in vivo can improve clinical and research diagnoses and prediction of cognitive trajectories. Tau PET imaging may serve a dual role in Lewy body disease by measuring cortical tau aggregation as well as assessing dopaminergic loss attributed to binding to neuromelanin within substantia nigra. We sought to characterize 18F-PI-2620, a next generation PET tracer, in individuals with Lewy body disease. We recruited 141 participants for 18F-PI-2620 PET scans from the Stanford Alzheimer's Disease Research Center and the Stanford Aging and Memory Study, most of whom also had β-amyloid status available (139/141) from PET or cerebrospinal fluid. We compared 18F-PI-2620 uptake within entorhinal cortex, inferior temporal cortex, precuneus and lingual gyrus, as well as substantia nigra, across participants with Lewy body disease [Parkinson's disease (n = 29), dementia with Lewy bodies (n = 14)] and Alzheimer's disease (n = 28), in addition to cognitively unimpaired healthy older adults (n = 70). Mean bilateral signal was extracted from cortical regions of interest in 18F-PI-2620 standard uptake value ratio (inferior cerebellar grey reference) images normalized to template space. A subset of participants received cognitive testing and/or the Movement Disorders Society Unified Parkinson's Disease Rating Scale Part III motor exam (off medication). 18F-PI-2620 uptake was low overall in Lewy body disease and correlated with β-amyloid PET in temporal lobe regions and precuneus. Moreover, inferior temporal 18F-PI-2620 uptake was significantly elevated in β-amyloid positive relative to β-amyloid negative participants with Lewy body disease. Temporal lobe 18F-PI-2620 signal was not associated with memory in Lewy body disease, but uptake within precuneus and lingual gyrus was associated with worse executive function and attention/working memory performance. Finally, substantia nigra 18F-PI-2620 signal was significantly reduced in participants with Parkinson's disease, and lower substantia nigra signal was associated with greater motor impairment. These findings suggest that although levels are lower than in Alzheimer's disease, small elevations in cortical tau are associated with cognitive function in Lewy body disease relevant domains, and that reduced 18F-PI-2620 binding in substantia nigra may represent loss of dopaminergic neurons. Cortical tau and neuromelanin binding within substantia nigra represent two unique signals in the same PET image that may be informative in the context of cognitive and motor deficits, respectively, in Lewy body disease.
Collapse
Affiliation(s)
- Joseph R Winer
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Hillary Vossler
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Christina B Young
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Viktorija Smith
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - America Romero
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Marian Shahid-Besanti
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Carla Abdelnour
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Edward N Wilson
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - David Anders
- Department of Radiology, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Aimara Pacheco Morales
- Department of Radiology, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Katrin I Andreasson
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Maya V Yutsis
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Victor W Henderson
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94304, USA
- Department of Epidemiology and Population Health, Stanford University, Stanford, CA 94304, USA
| | - Guido A Davidzon
- Department of Radiology, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Elizabeth C Mormino
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94304, USA
- Wu Tsai Neuroscience Institute, Stanford University, Stanford, CA 94304, USA
| | - Kathleen L Poston
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94304, USA
- Wu Tsai Neuroscience Institute, Stanford University, Stanford, CA 94304, USA
| |
Collapse
|
4
|
Weiner HL. Immune mechanisms and shared immune targets in neurodegenerative diseases. Nat Rev Neurol 2024:10.1038/s41582-024-01046-7. [PMID: 39681722 DOI: 10.1038/s41582-024-01046-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/15/2024] [Indexed: 12/18/2024]
Abstract
The immune system plays a major part in neurodegenerative diseases. In some, such as multiple sclerosis, it is the primary driver of the disease. In others, such as Alzheimer disease, amyotrophic lateral sclerosis and Parkinson disease, it has an amplifying role. Immunotherapeutic approaches that target the adaptive and innate immune systems are being explored for the treatment of almost all neurological diseases, and the targets and approaches are often common across diseases. Microglia are the primary immune cells in the brain that contribute to disease pathogenesis, and are consequently a common immune target for therapy. Other therapeutic approaches target components of the peripheral immune system, such as regulatory T cells and monocytes, which in turn act within the CNS. This Review considers in detail how microglia, monocytes and T cells contribute to the pathogenesis of multiple sclerosis, Alzheimer disease, amyotrophic lateral sclerosis and Parkinson disease, and their potential as shared therapeutic targets across these diseases. The microbiome is also highlighted as an emerging therapeutic target that indirectly modulates the immune system. Therapeutic approaches being developed to target immune function in neurodegenerative diseases are discussed, highlighting how immune-based approaches developed to treat one disease could be applicable to multiple other neurological diseases.
Collapse
Affiliation(s)
- Howard L Weiner
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
5
|
Maass F, Canaslan S, van Riesen C, Hermann P, Schmitz M, Schulte C, Brockmann K, Synofzik M, Bähr M, Zerr I. Myelin basic protein and TREM2 quantification in the CSF of patients with Multiple System Atrophy and other Parkinsonian conditions. J Neurol 2024; 272:52. [PMID: 39666067 PMCID: PMC11638341 DOI: 10.1007/s00415-024-12747-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/05/2024] [Accepted: 11/06/2024] [Indexed: 12/13/2024]
Abstract
BACKGROUND It is well known that myelin disruption and neuroinflammation are early and distinct pathological hallmarks in multiple system atrophy (MSA) as well as in idiopathic Parkinson's disease and in other atypical Parkinsonian syndromes. The objective of this study was to assess the value of non-neuronal biomarker candidates that reflect myelin disruption and neuroinflammation. METHODS Myelin basic protein (MBP) and the soluble form of TREM2 were quantified in a comprehensive movement disorder cohort from two different neurological centers, comprising a total of 171 CSF samples. Commercially available ELISA systems were employed for quantification. RESULTS The results of the MBP analysis revealed a significant increase in cerebrospinal fluid (CSF) MBP levels in all atypical Parkinsonian conditions compared to PD. This differentiation was more pronounced in the MSA-c subtype compared to MSA-p. Receiver operating characteristic (ROC) analysis revealed a significant discrimination between PD and MSA (p = 0.032, AUC = 0.70), PD and DLB (p = 0.006, AUC = 0.79) and PD and tauopathies (p = 0.006, AUC = 0.74). The results of the TREM2 analysis demonstrated no significant differences between the PD and atypical Parkinsonian groups if not adjusted for confounders. After adjusting for age, sex, and disease duration, the PD group exhibited significantly higher TREM2 levels compared to the DLB group (p = 0.002). CONCLUSIONS In conclusion, MBP, but not TREM2, is elevated in the CSF of not only MSA but in all atypical Parkinsonian conditions compared to idiopathic Parkinson's disease. This highlights the value of the evaluation of myelin/oligodendrocyte-associated markers in neurodegenerative movement disorders.
Collapse
Affiliation(s)
- Fabian Maass
- Department of Neurology, University Medical Center Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany.
| | - Sezgi Canaslan
- Department of Neurology, University Medical Center Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany
| | - Christoph van Riesen
- Department of Neurology, University Medical Center Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Peter Hermann
- Department of Neurology, University Medical Center Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany
| | - Matthias Schmitz
- Department of Neurology, University Medical Center Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany
| | - Claudia Schulte
- Hertie Institute for Clinical Brain Research and Center of Neurology, Department of Neurodegenerative Diseases, University of Tübingen, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Kathrin Brockmann
- Hertie Institute for Clinical Brain Research and Center of Neurology, Department of Neurodegenerative Diseases, University of Tübingen, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Matthis Synofzik
- Hertie Institute for Clinical Brain Research and Center of Neurology, Department of Neurodegenerative Diseases, University of Tübingen, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Mathias Bähr
- Department of Neurology, University Medical Center Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany
| | - Inga Zerr
- Department of Neurology, University Medical Center Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| |
Collapse
|
6
|
Kodosaki E, Bell R, Sogorb-Esteve A, Wiltshire K, Zetterberg H, Heslegrave A. More than microglia: myeloid cells and biomarkers in neurodegeneration. Front Neurosci 2024; 18:1499458. [PMID: 39544911 PMCID: PMC11560917 DOI: 10.3389/fnins.2024.1499458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 10/16/2024] [Indexed: 11/17/2024] Open
Abstract
The role of myeloid cells (granulocytes and monocytes) in neurodegeneration and neurodegenerative disorders (NDD) is indisputable. Here we discuss the roles of myeloid cells in neurodegenerative diseases, and the recent advances in biofluid and imaging myeloid biomarker research with a focus on methods that can be used in the clinic. For this review, evidence from three neurodegenerative diseases will be included, Alzheimer's disease (AD), Parkinson's disease (PD), and multiple sclerosis (MS). We discuss the potential for these biomarkers to be used in humans with suspected NDD as prognostic, diagnostic, or monitoring tools, identify knowledge gaps in literature, and propose potential approaches to further elucidate the role of myeloid cells in neurodegeneration and better utilize myeloid biomarkers in the understanding and treatment of NDD.
Collapse
Affiliation(s)
- Eleftheria Kodosaki
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
- UK Dementia Research Institute at UCL, London, United Kingdom
| | - Rosie Bell
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
- UK Dementia Research Institute at UCL, London, United Kingdom
| | - Aitana Sogorb-Esteve
- UK Dementia Research Institute at UCL, London, United Kingdom
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Katharine Wiltshire
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Henrik Zetterberg
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
- UK Dementia Research Institute at UCL, London, United Kingdom
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong SAR, China
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Amanda Heslegrave
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
- UK Dementia Research Institute at UCL, London, United Kingdom
| |
Collapse
|
7
|
Barba L, Abu-Rumeileh S, Barthel H, Massa F, Foschi M, Bellomo G, Gaetani L, Thal DR, Parnetti L, Otto M. Clinical and diagnostic implications of Alzheimer's disease copathology in Lewy body disease. Brain 2024; 147:3325-3343. [PMID: 38991041 DOI: 10.1093/brain/awae203] [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: 01/27/2024] [Revised: 05/03/2024] [Accepted: 06/02/2024] [Indexed: 07/13/2024] Open
Abstract
Concomitant Alzheimer's disease (AD) pathology is a frequent event in the context of Lewy body disease (LBD), occurring in approximately half of all cases. Evidence shows that LBD patients with AD copathology show an accelerated disease course, a greater risk of cognitive decline and an overall poorer prognosis. However, LBD-AD cases may show heterogeneous motor and non-motor phenotypes with a higher risk of dementia and, consequently, be not rarely misdiagnosed. In this review, we summarize the current understanding of LBD-AD by discussing the synergistic effects of AD neuropathological changes and Lewy pathology and their clinical relevance. Furthermore, we provide an extensive overview of neuroimaging and fluid biomarkers under assessment for use in LBD-AD and their possible diagnostic and prognostic values. AD pathology can be predicted in vivo by means of CSF, MRI and PET markers, whereas the most promising technique to date for identifying Lewy pathology in different biological tissues is the α-synuclein seed amplification assay. Pathological imaging and CSF AD biomarkers are associated with a higher likelihood of cognitive decline in LBD but do not always mirror the neuropathological severity as in pure AD. Implementing the use of blood-based AD biomarkers might allow faster screening of LBD patients for AD copathology, thus improving the overall diagnostic sensitivity for LBD-AD. Finally, we discuss the literature on novel candidate biomarkers being exploited in LBD-AD to investigate other aspects of neurodegeneration, such as neuroaxonal injury, glial activation and synaptic dysfunction. The thorough characterization of AD copathology in LBD should be taken into account when considering differential diagnoses of dementia syndromes, to allow prognostic evaluation on an individual level, and to guide symptomatic and disease-modifying therapies.
Collapse
Affiliation(s)
- Lorenzo Barba
- Department of Neurology, Martin-Luther-University of Halle-Wittenberg, Halle 06120, Germany
| | - Samir Abu-Rumeileh
- Department of Neurology, Martin-Luther-University of Halle-Wittenberg, Halle 06120, Germany
| | - Henryk Barthel
- Department of Nuclear Medicine, University Hospital of Leipzig, Leipzig 04103, Germany
| | - Federico Massa
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa 16132, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa 16132, Italy
| | - Matteo Foschi
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila 67100, Italy
- Department of Neuroscience, Neurology Unit, S. Maria delle Croci Hospital of Ravenna, AUSL Romagna, Ravenna 48121, Italy
| | - Giovanni Bellomo
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia 06129, Italy
| | - Lorenzo Gaetani
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia 06129, Italy
| | - Dietmar R Thal
- Department of Imaging and Pathology, Laboratory for Neuropathology, Leuven Brain Institute, KU Leuven, Leuven 3001, Belgium
- Department of Pathology, UZ Leuven, Leuven 3000, Belgium
| | - Lucilla Parnetti
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia 06129, Italy
| | - Markus Otto
- Department of Neurology, Martin-Luther-University of Halle-Wittenberg, Halle 06120, Germany
| |
Collapse
|
8
|
Wilson E, Umans J, Swarovski M, Minhas P, Midttun Ø, Ulvik AA, Shahid-Besanti M, Linortner P, Mhatre S, Wang Q, Channappa D, Corso N, Tian L, Fredericks C, Kerchner G, Plowey E, Cholerton B, Ueland P, Zabetian C, Gray N, Quinn J, Montine T, Sha S, Longo F, Wolk D, Chen-Plotkin A, Henderson V, Wyss-Coray T, Wagner A, Mormino E, Aghaeepour N, Poston K, Andreasson K. Parkinson's disease is characterized by vitamin B6-dependent inflammatory kynurenine pathway dysfunction. RESEARCH SQUARE 2024:rs.3.rs-4980210. [PMID: 39399688 PMCID: PMC11469709 DOI: 10.21203/rs.3.rs-4980210/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Parkinson's disease (PD) is a complex multisystem disorder clinically characterized by motor, non-motor, and premotor manifestations. Pathologically, PD involves neuronal loss in the substantia nigra, striatal dopamine deficiency, and accumulation of intracellular inclusions containing aggregates of α-synuclein. Recent studies demonstrate that PD is associated with dysregulated metabolic flux through the kynurenine pathway (KP), in which tryptophan is converted to kynurenine (KYN), and KYN is subsequently metabolized to neuroactive compounds quinolinic acid (QA) and kynurenic acid (KA). This multicenter study used highly sensitive liquid chromatography-tandem mass-spectrometry to compare blood and cerebral spinal fluid (CSF) KP metabolites between 158 unimpaired older adults and 177 participants with PD. Results indicate that increased neuroexcitatory QA/KA ratio in both plasma and CSF of PD participants associated with peripheral and cerebral inflammation and vitamin B6 deficiency. Furthermore, increased QA tracked with CSF tau and severity of both motor and non-motor PD clinical dysfunction. Importantly, plasma and CSF kynurenine metabolites classified PD participants with a high degree of accuracy (AUC = 0.897). Finally, analysis of metabolite data revealed subgroups with distinct KP profiles, and these were subsequently found to display distinct PD clinical features. Together, these data further support the hypothesis that the KP serves as a site of brain and periphery crosstalk, integrating B-vitamin status, inflammation and metabolism to ultimately influence PD clinical manifestation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Cyrus Zabetian
- VA Puget Sound Health Care System and University of Washington Seattle
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Gao Y, Sheng D, Chen W. Regulatory mechanism of miR-20a-5p in neuronal damage and inflammation in lipopolysaccharide-induced BV2 cells and MPTP-HCl-induced Parkinson's disease mice. Psychogeriatrics 2024; 24:752-764. [PMID: 38664198 DOI: 10.1111/psyg.13109] [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: 08/28/2023] [Revised: 02/05/2024] [Accepted: 02/25/2024] [Indexed: 07/05/2024]
Abstract
BACKGROUND Parkinson's disease (PD) is a prevailing neurodegenerative disorder increasingly affecting the elderly population. The involvement of microRNAs (miRNAs) in PD has been confirmed. We sought to explore the molecular mechanism of miR-20a-5p in PD. METHODS Lipopolysaccharide (LPS)-induced BV2 cell model and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine hydrochloride (MPTP-HCl)-induced PD mouse model were established. miR-20a-5p, inducible nitric oxide synthase (iNOS), interleukin (IL)-6, tumour necrosis factor (TNF)-α, transforming growth factor (TGF)-β1, and IL-10 expression in BV2 cells was examined by reverse transcription - quantitative polymerase chain reaction. Cell viability was assessed by MTT assay. The apoptotic rate and levels of Bcl-2, Bax, cleaved caspase-3, and signal transducer and activator of transmission (STAT)3 were examined by flow cytometry and Western blot. Bioinformatics software predicted the potential binding sites of miR-20a-5p and STAT3. Dual-luciferase experiment verified the binding relationship. Iba1-positive and tyrosine hydroxylase (TH)-positive cell numbers in substantia nigra pars compacta were detected by immunohistochemistry. The effect of miR-20a-5p on motor function in MPTP-induced PD mice was detected by Rota-rod test, Pole test, Traction test and Beam-crossing task. RESULTS miR-20a-5p was under-expressed in LPS-induced BV2 cells. Overexpression of miR-20a-5p increased the viability of LPS-induced BV2 cells and reduced apoptosis rates. Moreover, overexpression of miR-20a-5p reduced cleaved caspase-3, Bax, iNOS, IL-6, and TNF-α and increased Bcl-2 and TGF-β1, and IL-10. miR-20a-5p targeted STAT3. STAT3 overexpression partially reversed miR-20a-5p overexpression-mediated effects on LPS-induced BV2 cell viability, apoptosis, and inflammatory responses. miR-20a-5p overexpression inhibited MPTP-induced STAT3 and α-synuclein levels, microglia activation, and inflammatory response, and reduced the loss of TH-positive cells in mice. miR-20a-5p overexpression ameliorated MPTP-induced dyskinesia in PD model mice. CONCLUSION miR-20a-5p alleviates neuronal damage and suppresses inflammation by targeting STAT3 in PD.
Collapse
Affiliation(s)
- Yanlei Gao
- Emergency Department, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Dan Sheng
- Emergency Department, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Weiguang Chen
- Emergency Department, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| |
Collapse
|
10
|
Wang Y, Ye M, Ji Q, Liu Q, Xu X, Zhan Y. The longitudinal trajectory of CSF sTREM2: the alzheimer's disease neuroimaging initiative. Alzheimers Res Ther 2024; 16:138. [PMID: 38926894 PMCID: PMC11202383 DOI: 10.1186/s13195-024-01506-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND The soluble triggering receptor expressed on myeloid cells 2 (sTREM2) in cerebrospinal fluid (CSF) is considered a biomarker of microglia activity. The objective of this study was to investigate the trajectory of CSF sTREM2 levels over time and examine its association with sex. METHODS A total of 1,017 participants from the Alzheimer's Disease Neuroimaging Initiative Study (ADNI) with at least one CSF sTREM2 record were included. The trajectory of CSF sTREM2 was analyzed using a growth curve model. The association between CSF sTREM2 levels and sex was assessed using linear mixed-effect models. RESULTS CSF sTREM2 levels were increased with age over time (P < 0.0001). No significant sex difference was observed in sTREM2 levels across the entire sample; however, among the APOE ε4 allele carriers, women exhibited significantly higher sTREM2 levels than men (β = 0.146, P = 0.002). CONCLUSION Our findings highlight the association between CSF sTREM2 levels and age-related increments, underscoring the potential influence of aging on sTREM2 dynamics. Furthermore, our observations indicate a noteworthy association between sex and CSF sTREM2 levels, particularly in individuals carrying the APOE ε4 allele.
Collapse
Affiliation(s)
- Yu Wang
- Department of Epidemiology, School of Public Health (Shenzhen), Sun Yat-Sen University, Shenzhen, China
| | - Meijie Ye
- Department of Epidemiology, School of Public Health (Shenzhen), Sun Yat-Sen University, Shenzhen, China
| | - Qianqian Ji
- Department of Epidemiology, School of Public Health (Shenzhen), Sun Yat-Sen University, Shenzhen, China
| | - Qi Liu
- Department of Epidemiology, School of Public Health (Shenzhen), Sun Yat-Sen University, Shenzhen, China
| | - Xiaowei Xu
- Department of Neurology, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China.
| | - Yiqiang Zhan
- Department of Epidemiology, School of Public Health (Shenzhen), Sun Yat-Sen University, Shenzhen, China.
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
11
|
Lin C, Kong Y, Chen Q, Zeng J, Pan X, Miao J. Decoding sTREM2: its impact on Alzheimer's disease - a comprehensive review of mechanisms and implications. Front Aging Neurosci 2024; 16:1420731. [PMID: 38912524 PMCID: PMC11190086 DOI: 10.3389/fnagi.2024.1420731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 05/30/2024] [Indexed: 06/25/2024] Open
Abstract
Soluble Triggering Receptor Expressed on Myeloid Cells 2 (sTREM2) plays a crucial role in the pathogenesis of Alzheimer's disease (AD). This review comprehensively examines sTREM2's involvement in AD, focusing on its regulatory functions in microglial responses, neuroinflammation, and interactions with key pathological processes. We discuss the dynamic changes in sTREM2 levels in cerebrospinal fluid and plasma throughout AD progression, highlighting its potential as a therapeutic target. Furthermore, we explore the impact of genetic variants on sTREM2 expression and its interplay with other AD risk genes. The evidence presented in this review suggests that modulating sTREM2 activity could influence AD trajectory, making it a promising avenue for future research and drug development. By providing a holistic understanding of sTREM2's multifaceted role in AD, this review aims to guide future studies and inspire novel therapeutic strategies.
Collapse
Affiliation(s)
- Cui Lin
- Shenzhen Bao’an District Hospital of Traditional Chinese Medicine, Shenzhen, Guangdong, China
| | - Yu Kong
- Shenzhen Bao’an District Hospital of Traditional Chinese Medicine, Shenzhen, Guangdong, China
| | - Qian Chen
- Shenzhen Bao’an District Hospital of Traditional Chinese Medicine, Shenzhen, Guangdong, China
| | - Jixiang Zeng
- Shenzhen Bao’an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Xiaojin Pan
- Shenzhen Bao’an District Hospital of Traditional Chinese Medicine, Shenzhen, Guangdong, China
| | - Jifei Miao
- Shenzhen Bao’an District Hospital of Traditional Chinese Medicine, Shenzhen, Guangdong, China
| |
Collapse
|
12
|
Wilson EN, Wang C, Swarovski MS, Zera KA, Ennerfelt HE, Wang Q, Chaney A, Gauba E, Ramos Benitez JA, Le Guen Y, Minhas PS, Panchal M, Tan YJ, Blacher E, A Iweka C, Cropper H, Jain P, Liu Q, Mehta SS, Zuckerman AJ, Xin M, Umans J, Huang J, Durairaj AS, Serrano GE, Beach TG, Greicius MD, James ML, Buckwalter MS, McReynolds MR, Rabinowitz JD, Andreasson KI. TREM1 disrupts myeloid bioenergetics and cognitive function in aging and Alzheimer disease mouse models. Nat Neurosci 2024; 27:873-885. [PMID: 38539014 PMCID: PMC11102654 DOI: 10.1038/s41593-024-01610-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 02/22/2024] [Indexed: 04/21/2024]
Abstract
Human genetics implicate defective myeloid responses in the development of late-onset Alzheimer disease. A decline in peripheral and brain myeloid metabolism, triggering maladaptive immune responses, is a feature of aging. The role of TREM1, a pro-inflammatory factor, in neurodegenerative diseases is unclear. Here we show that Trem1 deficiency prevents age-dependent changes in myeloid metabolism, inflammation and hippocampal memory function in mice. Trem1 deficiency rescues age-associated declines in ribose 5-phosphate. In vitro, Trem1-deficient microglia are resistant to amyloid-β42 oligomer-induced bioenergetic changes, suggesting that amyloid-β42 oligomer stimulation disrupts homeostatic microglial metabolism and immune function via TREM1. In the 5XFAD mouse model, Trem1 haploinsufficiency prevents spatial memory loss, preserves homeostatic microglial morphology, and reduces neuritic dystrophy and changes in the disease-associated microglial transcriptomic signature. In aging APPSwe mice, Trem1 deficiency prevents hippocampal memory decline while restoring synaptic mitochondrial function and cerebral glucose uptake. In postmortem Alzheimer disease brain, TREM1 colocalizes with Iba1+ cells around amyloid plaques and its expression is associated with Alzheimer disease clinical and neuropathological severity. Our results suggest that TREM1 promotes cognitive decline in aging and in the context of amyloid pathology.
Collapse
Affiliation(s)
- Edward N Wilson
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Congcong Wang
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Michelle S Swarovski
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Kristy A Zera
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Hannah E Ennerfelt
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Qian Wang
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Aisling Chaney
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Esha Gauba
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Javier A Ramos Benitez
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Yann Le Guen
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Paras S Minhas
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Maharshi Panchal
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Yuting J Tan
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Eran Blacher
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Chinyere A Iweka
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Haley Cropper
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Poorva Jain
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Qingkun Liu
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Swapnil S Mehta
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Abigail J Zuckerman
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Matthew Xin
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Jacob Umans
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Jolie Huang
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Aarooran S Durairaj
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Geidy E Serrano
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ, USA
| | - Thomas G Beach
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ, USA
| | - Michael D Greicius
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Michelle L James
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Marion S Buckwalter
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Melanie R McReynolds
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
- Department of Chemistry, Princeton University, Princeton, NJ, USA
- Department of Biochemistry and Molecular Biology, Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, USA
| | - Joshua D Rabinowitz
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
- Department of Chemistry, Princeton University, Princeton, NJ, USA
| | - Katrin I Andreasson
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
| |
Collapse
|
13
|
Plastini MJ, Abdelnour C, Young CB, Wilson EN, Shahid-Besanti M, Lamoureux J, Andreasson KI, Kerchner GA, Montine TJ, Henderson VW, Poston KL. Multiple biomarkers improve diagnostic accuracy across Lewy body and Alzheimer's disease spectra. Ann Clin Transl Neurol 2024; 11:1197-1210. [PMID: 38436140 DOI: 10.1002/acn3.52034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 01/20/2024] [Accepted: 02/13/2024] [Indexed: 03/05/2024] Open
Abstract
OBJECTIVE More than half of neurodegenerative disease patients have multiple pathologies at autopsy; however, most receive one diagnosis during life. We used the α-synuclein seed amplification assay (αSyn-SAA) and CSF biomarkers for amyloidosis and Alzheimer's disease (AD) neuropathological change (ADNC) to determine the frequency of co-pathologies in participants clinically diagnosed with Lewy body (LB) disease or AD. METHODS Using receiver operating characteristic analyses on retrospective CSF samples from 150 participants determined αSyn-SAA accuracy, sensitivity, and specificity for identifying clinically defined LB disease and predicting future change in clinical diagnosis. CSF biomarkers helped determine the frequency of concomitant Lewy body pathology, ADNC, and/or amyloidosis in participants with LB disease and AD, across clinical spectra. RESULTS Following a decade-long follow-up, the clinically or autopsy-defined diagnosis changed for nine participants. αSyn-SAA demonstrated improved accuracy (91.3%), sensitivity (89.3%), and specificity (93.3%) for identifying LB disease compared to all non-LB disease, highlighting the limitations of clinical diagnosis alone. When examining biomarkers of co-pathology, amyloidosis was present in 18%, 48%, and 71% (χ2(2) = 13.56, p = 0.001) and AD biomarkers were present in 0%, 8.7%, and 42.9% (χ2(2) = 18.44, p < 0.001) of LB disease participants with different stages of cognitive impairment respectively. Co-occurring biomarkers for αSyn-SAA and amyloidosis were present in 12% and 14% of AD compared to 43% and 57% LB disease participants with different stages of cognitive impairment (χ2(3) = 13.87, p = 0.003). INTERPRETATION Our study shows that using a combination of αSyn-SAA and AD biomarkers can identify people with αSyn, ADNC, and co-pathology better and earlier than traditional clinical diagnostic criteria alone.
Collapse
Affiliation(s)
- Melanie J Plastini
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
- Knight Initiative for Brain Resilience, Stanford University, Stanford, California, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, California, USA
| | - Carla Abdelnour
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
- Knight Initiative for Brain Resilience, Stanford University, Stanford, California, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, California, USA
| | - Christina B Young
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
- Knight Initiative for Brain Resilience, Stanford University, Stanford, California, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, California, USA
| | - Edward N Wilson
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
- Knight Initiative for Brain Resilience, Stanford University, Stanford, California, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, California, USA
| | - Marian Shahid-Besanti
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
- Knight Initiative for Brain Resilience, Stanford University, Stanford, California, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, California, USA
| | | | - Katrin I Andreasson
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
- Knight Initiative for Brain Resilience, Stanford University, Stanford, California, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, California, USA
- Chan Zuckerberg Biohub, San Francisco, California, 94158, USA
| | - Geoffrey A Kerchner
- Pharma Research and Early Development, F. Hoffmann-La Roche, Ltd., Basel, Switzerland
| | - Thomas J Montine
- Knight Initiative for Brain Resilience, Stanford University, Stanford, California, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, California, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
| | - Victor W Henderson
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, California, USA
- Department of Epidemiology and Population Health, Stanford University, Stanford, California, USA
| | - Kathleen L Poston
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
- Knight Initiative for Brain Resilience, Stanford University, Stanford, California, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, California, USA
| |
Collapse
|
14
|
Rao Y, Peng B. Allogenic microglia replacement: A novel therapeutic strategy for neurological disorders. FUNDAMENTAL RESEARCH 2024; 4:237-245. [PMID: 38933508 PMCID: PMC11197774 DOI: 10.1016/j.fmre.2023.02.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 11/17/2022] [Accepted: 02/19/2023] [Indexed: 03/29/2023] Open
Abstract
Microglia are resident immune cells in the central nervous system (CNS) that play vital roles in CNS development, homeostasis and disease pathogenesis. Genetic defects in microglia lead to microglial dysfunction, which in turn leads to neurological disorders. The correction of the specific genetic defects in microglia in these disorders can lead to therapeutic effects. Traditional genetic defect correction approaches are dependent on viral vector-based genetic defect corrections. However, the viruses used in these approaches, including adeno-associated viruses, lentiviruses and retroviruses, do not primarily target microglia; therefore, viral vector-based genetic defect corrections are ineffective in microglia. Microglia replacement is a novel approach to correct microglial genetic defects via replacing microglia of genetic defects with allogenic healthy microglia. In this paper, we systematically review the history, rationale and therapeutic perspectives of microglia replacement, which would be a novel strategy for treating CNS disorders.
Collapse
Affiliation(s)
- Yanxia Rao
- Department of Laboratory Animal Science, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Bo Peng
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan University, Shanghai 200000, China
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China
| |
Collapse
|
15
|
Vicente M, Addo-Osafo K, Vossel K. Latest advances in mechanisms of epileptic activity in Alzheimer's disease and dementia with Lewy Bodies. Front Neurol 2024; 15:1277613. [PMID: 38390593 PMCID: PMC10882721 DOI: 10.3389/fneur.2024.1277613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 01/12/2024] [Indexed: 02/24/2024] Open
Abstract
Alzheimer's disease (AD) and dementia with Lewy bodies (DLB) stand as the prevailing sources of neurodegenerative dementia, impacting over 55 million individuals across the globe. Patients with AD and DLB exhibit a higher prevalence of epileptic activity compared to those with other forms of dementia. Seizures can accompany AD and DLB in early stages, and the associated epileptic activity can contribute to cognitive symptoms and exacerbate cognitive decline. Aberrant neuronal activity in AD and DLB may be caused by several mechanisms that are not yet understood. Hyperexcitability could be a biomarker for early detection of AD or DLB before the onset of dementia. In this review, we compare and contrast mechanisms of network hyperexcitability in AD and DLB. We examine the contributions of genetic risk factors, Ca2+ dysregulation, glutamate, AMPA and NMDA receptors, mTOR, pathological amyloid beta, tau and α-synuclein, altered microglial and astrocytic activity, and impaired inhibitory interneuron function. By gaining a deeper understanding of the molecular mechanisms that cause neuronal hyperexcitability, we might uncover therapeutic approaches to effectively ease symptoms and slow down the advancement of AD and DLB.
Collapse
Affiliation(s)
- Mariane Vicente
- Mary S. Easton Center for Alzheimer's Research and Care, Department of Neurology, David Geffen School of Medicine at University of California, Los Angeles, CA, United States
| | - Kwaku Addo-Osafo
- Mary S. Easton Center for Alzheimer's Research and Care, Department of Neurology, David Geffen School of Medicine at University of California, Los Angeles, CA, United States
| | - Keith Vossel
- Mary S. Easton Center for Alzheimer's Research and Care, Department of Neurology, David Geffen School of Medicine at University of California, Los Angeles, CA, United States
| |
Collapse
|
16
|
Qin C, Chen M, Dong MH, Yang S, Zhang H, You YF, Zhou LQ, Chu YH, Tang Y, Pang XW, Wu LJ, Tian DS, Wang W. Soluble TREM2 triggers microglial dysfunction in neuromyelitis optica spectrum disorders. Brain 2024; 147:163-176. [PMID: 37740498 DOI: 10.1093/brain/awad321] [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: 12/31/2022] [Revised: 06/21/2023] [Accepted: 09/14/2023] [Indexed: 09/24/2023] Open
Abstract
Microglia-mediated neuroinflammation contributes to acute demyelination in neuromyelitis optica spectrum disorders (NMOSD). Soluble triggering receptor expressed on myeloid cells 2 (sTREM2) in the CSF has been associated with microglial activation in several neurodegenerative diseases. However, the basis for this immune-mediated attack and the pathophysiological role of sTREM2 in NMOSD remain to be elucidated. Here, we performed Mendelian randomization analysis and identified a genetic association between increased CSF sTREM2 and NMOSD risk. CSF sTREM2 was elevated in patients with NMOSD and was positively correlated with neural injury and other neuroinflammation markers. Single-cell RNA sequencing of human macrophage/microglia-like cells in CSF, a proxy for microglia, showed that increased CSF sTREM2 was positively associated with microglial dysfunction in patients with NMOSD. Furthermore, we demonstrated that sTREM2 is a reliable biomarker of microglial activation in a mouse model of NMOSD. Using unbiased transcriptomic and lipidomic screens, we identified that excessive activation, overwhelmed phagocytosis of myelin debris, suppressed lipid metabolism and enhanced glycolysis underlie sTREM2-mediated microglial dysfunction, possibly through the nuclear factor kappa B (NF-κB) signalling pathway. These molecular and cellular findings provide a mechanistic explanation for the genetic association between CSF sTREM2 and NMOSD risk and indicate that sTREM2 could be a potential biomarker of NMOSD progression and a therapeutic target for microglia-mediated neuroinflammation.
Collapse
Affiliation(s)
- Chuan Qin
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Man Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ming-Hao Dong
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Sheng Yang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hang Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yun-Fan You
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Luo-Qi Zhou
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yun-Hui Chu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yue Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiao-Wei Pang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, NY 14600, USA
| | - Dai-Shi Tian
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
| |
Collapse
|
17
|
Liu C, Jiang Z, Liu S, Chu C, Wang J, Liu W, Sun Y, Dong M, Shi Q, Huang P, Zhu X. Frequency-Dependent Microstate Characteristics for Mild Cognitive Impairment in Parkinson's Disease. IEEE Trans Neural Syst Rehabil Eng 2023; 31:4115-4124. [PMID: 37831557 DOI: 10.1109/tnsre.2023.3324343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2023]
Abstract
Cognitive impairment is typically reflected in the time and frequency variations of electroencephalography (EEG). Integrating time-domain and frequency-domain analysis methods is essential to better understand and assess cognitive ability. Timely identification of cognitive levels in early Parkinson's disease (ePD) patients can help mitigate the risk of future dementia. For the investigation of the brain activity and states related to cognitive levels, this study recruited forty ePD patients for EEG microstate analysis, including 13 with mild cognitive impairment (MCI) and 27 without MCI (control group). To determine the specific frequency band on which the microstate analysis relies, a deep learning framework was employed to discern the frequency dependence of the cognitive level in ePD patients. The input to the convolutional neural network consisted of the power spectral density of multi-channel multi-point EEG signals. The visualization technique of gradient-weighted class activation mapping was utilized to extract the optimal frequency band for identifying MCI samples. Within this frequency band, microstate analysis was conducted and correlated with the Montreal Cognitive Assessment (MoCA) Scale. The deep neural network revealed significant differences in the 1-11.5Hz spectrum of the ePD-MCI group compared to the control group. In this characteristic frequency band, ePD-MCI patients exhibited a pattern of global microstate disorder. The coverage rate and occurrence frequency of microstate A and D increased significantly and were both negatively correlated with the MoCA scale. Meanwhile, the coverage, frequency and duration of microstate C decreased significantly and were positively correlated with the MoCA scale. Our work unveils abnormal microstate characteristics in ePD-MCI based on time-frequency fusion, enhancing our understanding of cognitively related brain dynamics and providing electrophysiological markers for ePD-MCI recognition.
Collapse
|
18
|
Gao H, Di J, Clausen BH, Wang N, Zhu X, Zhao T, Chang Y, Pang M, Yang Y, He R, Wang Y, Zhang L, Liu B, Qiu W, Lambertsen KL, Brambilla R, Rong L. Distinct myeloid population phenotypes dependent on TREM2 expression levels shape the pathology of traumatic versus demyelinating CNS disorders. Cell Rep 2023; 42:112629. [PMID: 37289590 DOI: 10.1016/j.celrep.2023.112629] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 04/23/2023] [Accepted: 05/24/2023] [Indexed: 06/10/2023] Open
Abstract
Triggering receptor expressed on myeloid cell 2 (TREM2) signaling often drives opposing effects in traumatic versus demyelinating CNS disorders. Here, we identify two distinct phenotypes of microglia and infiltrating myeloid populations dependent on TREM2 expression levels at the acute stage and elucidate how they mediate the opposing effects of TREM2 in spinal cord injury (SCI) versus multiple sclerosis animal models (experimental autoimmune encephalomyelitis [EAE]). High TREM2 levels sustain phagocytic microglia and infiltrating macrophages after SCI. In contrast, moderate TREM2 levels sustain immunomodulatory microglia and infiltrating monocytes in EAE. TREM2-ablated microglia (purine-sensing phenotype in SCI and reduced immunomodulatory phenotype in EAE) drive transient protection at the acute stage of both disorders, whereas reduced phagocytic macrophages and lysosome-activated monocytes lead to contrasting neuroprotective and demyelinating effects in SCI versus EAE, respectively. Our study provides comprehensive insights into the complex roles of TREM2 in myeloid populations across diverse CNS disorders, which has crucial implications in devising TREM2-targeting therapeutics.
Collapse
Affiliation(s)
- Han Gao
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China; Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou 510630, China.
| | - Jiawei Di
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China; Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou 510630, China
| | - Bettina Hjelm Clausen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark
| | - Nanxiang Wang
- Department of Orthopaedic Surgery, The 2nd Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Xizhong Zhu
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China; Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou 510630, China
| | - Tianlun Zhao
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China; Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou 510630, China
| | - Yanyu Chang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Mao Pang
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China; Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou 510630, China
| | - Yang Yang
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China; Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou 510630, China
| | - Ronghan He
- Department of Joint and Trauma Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Yuge Wang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Liangming Zhang
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China; Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou 510630, China
| | - Bin Liu
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China; Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou 510630, China
| | - Wei Qiu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Kate Lykke Lambertsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; Department of Neurology, Odense University Hospital, 5000 Odense, Denmark; BRIDGE - Brain Research - Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, 5000 Odense, Denmark
| | - Roberta Brambilla
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; BRIDGE - Brain Research - Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, 5000 Odense, Denmark; The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33161, USA.
| | - Limin Rong
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China; Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou 510630, China.
| |
Collapse
|
19
|
Zhang X, Zhong X, Wang L, Li H, Yang L, Li X, Yu X, Xie A. Effects of soluble TREM2 on motor progression in Parkinson's disease. Neurosci Lett 2023; 807:137277. [PMID: 37105353 DOI: 10.1016/j.neulet.2023.137277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/07/2023] [Accepted: 04/23/2023] [Indexed: 04/29/2023]
Abstract
OBJECTIVES To determine whether sTREM2 is changed during the pathogenesis of PD and reflect motor decline in PD individuals METHODS: The cerebrospinal fluid (CSF) from PD and healthy individuals were obtained to measure the expression of sTREM2 and further to evaluate the motor function at baseline and after four years of follow-up using the Parkinson's Progression Markers Initiative (PPMI) database. The relationship between motor disease progression at baseline and longitudinal CSF sTREM2 was evaluated by linear mixed-effects (LME) and multiple linear regression (MLR) models. The change rates of the motor symptoms and sTREM2 level in CSF were further rigorously analyzed using the LME model. Cox proportional-hazards regression models were used to evaluate the predictive values of sTREM2 in CSF for motor progression. The regulatory role of CSF α-syn and Tau between sTREM2 and motor assessments was evaluated by Mediating effect analysis. RESULTS We found no significant difference in CSF sTREM2 levels between the PD and HC groups (p = 0.155). However, late-onset PD patients had higher CSF sTREM2 levels than early-onset PD patients (p = 0.044). The basal levels of sTREM2 could predict motor progression over the four years of follow-up. The change rate of CSF sTREM2 was correlated with the progressive deterioration of motor function in PD individuals. Our observations also showed that CSF Tau was a significant mediator of the association between CSF sTREM2 and total UPDRS scores and UPDRS III and tremor at baseline with 26.5% and 33.5%, and 28.7% mediation, respectively. CONCLUSION Our results indicated that CSF sTREM2 was associated with the prognosis of PD motor symptoms. Besides, CSF Tau could effectively mediate the association of sTREM2 with motor progression.
Collapse
Affiliation(s)
- Xue Zhang
- Department of Neurology, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao Cancer Hospital
| | - Xiaoling Zhong
- Department of Neurology, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao Cancer Hospital
| | - Ling Wang
- Department of Neurology, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao Cancer Hospital
| | - Haiyan Li
- Department of Neurology, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao Cancer Hospital
| | - Liying Yang
- Department of Neurology, Affiliated Hospital of Qingdao University
| | - Xiaoyuan Li
- Department of Neurology, Affiliated Hospital of Qingdao University
| | - Xiaolong Yu
- Department of Neurology, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao Cancer Hospital
| | - Anmu Xie
- Department of Neurology, Affiliated Hospital of Qingdao University.
| |
Collapse
|
20
|
Paolini Paoletti F, Gaetani L, Bellomo G, Chipi E, Salvadori N, Montanucci C, Mancini A, Filidei M, Nigro P, Simoni S, Tambasco N, Di Filippo M, Parnetti L. CSF neurochemical profile and cognitive changes in Parkinson's disease with mild cognitive impairment. NPJ Parkinsons Dis 2023; 9:68. [PMID: 37095141 PMCID: PMC10126070 DOI: 10.1038/s41531-023-00509-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 04/05/2023] [Indexed: 04/26/2023] Open
Abstract
Pathophysiological substrate(s) and progression of Parkinson's disease (PD) with mild cognitive impairment (PD-MCI) are still matter of debate. Baseline cerebrospinal fluid (CSF) neurochemical profile and cognitive changes after 2 years were investigated in a retrospective series of PD-MCI (n = 48), cognitively normal PD (PD-CN, n = 40), prodromal Alzheimer's disease (MCI-AD, n = 25) and cognitively healthy individuals with other neurological diseases (OND, n = 44). CSF biomarkers reflecting amyloidosis (Aβ42/40 ratio, sAPPα, sAPPβ), tauopathy (p-tau), neurodegeneration (t-tau, NfL, p-NfH), synaptic damage (α-syn, neurogranin) and glial activation (sTREM2, YKL-40) were measured. The great majority (88%) of PD-MCI patients was A-/T-/N-. Among all biomarkers considered, only NfL/p-NfH ratio was significantly higher in PD-MCI vs. PD-CN (p = 0.02). After 2 years, one-third of PD-MCI patients worsened; such worsening was associated with higher baseline levels of NfL, p-tau, and sTREM2. PD-MCI is a heterogeneous entity requiring further investigations on larger, longitudinal cohorts with neuropathological verification.
Collapse
Affiliation(s)
- Federico Paolini Paoletti
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
- Laboratory of Clinical Neurochemistry, Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Lorenzo Gaetani
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
- Laboratory of Clinical Neurochemistry, Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Giovanni Bellomo
- Laboratory of Clinical Neurochemistry, Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Elena Chipi
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Nicola Salvadori
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Chiara Montanucci
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Andrea Mancini
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Marta Filidei
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Pasquale Nigro
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Simone Simoni
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Nicola Tambasco
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Massimiliano Di Filippo
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Lucilla Parnetti
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy.
- Laboratory of Clinical Neurochemistry, Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy.
| |
Collapse
|
21
|
Soluble TREM2 in body fluid in Alzheimer's disease and Parkinson's disease. Neurol Sci 2023:10.1007/s10072-023-06729-5. [PMID: 36913148 DOI: 10.1007/s10072-023-06729-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 03/02/2023] [Indexed: 03/14/2023]
Abstract
BACKGROUND Previous studies showed conflicting results regarding soluble triggering receptor expressed on myeloid cells 2 (sTREM2) level alteration in body fluid in Alzheimer's disease (AD) and Parkinson's disease (PD). METHODS We applied the STATA 12.0 software to compute standard mean difference (SMD) and 95% confidence interval (CI). RESULTS The study showed elevated sTREM2 level in cerebrospinal fluid (CSF) in AD, mild cognitive impairment (MCI), and preclinical AD (pre-AD) patients, compared to healthy controls (HCs) with random effects models (AD: SMD 0.28, 95% CI 0.12 to 0.44, I2 = 77.6%, p < 0.001; MCI: SMD 0.29, 95% CI 0.09 to 0.48, I2 = 89.7%, p < 0.001; pre-AD: SMD 0.24, 95% CI 0.00 to 0.48, I2 = 80.8%, p < 0.001). The study showed no significant difference in sTREM2 level in plasma between AD patients and HCs with a random effects model (SMD 0.06, 95% CI - 0.16 to 0.28, I2 = 65.6%, p = 0.008). The study showed no significant difference in sTREM2 level in CSF or plasma between PD patients and HCs with random effects models (CSF: SMD 0.33, 95% CI - 0.02 to 0.67, I2 = 85.6%, p < 0.001; plasma: SMD 0.37, 95% CI - 0.17 to 0.92, I2 = 77.8%, p = 0.011). CONCLUSIONS In conclusion, the study highlighted the CSF sTREM2 as a promising biomarker in the different clinical stages of AD. More studies were essential to explore the CSF and plasmatic concentrations of sTREM2 alteration in PD.
Collapse
|
22
|
Bonomi CG, Assogna M, Di Donna MG, Bernocchi F, De Lucia V, Nuccetelli M, Fiorelli D, Loizzo S, Mercuri NB, Koch G, Martorana A, Motta C. Cerebrospinal Fluid sTREM-2, GFAP, and β-S100 in Symptomatic Sporadic Alzheimer's Disease: Microglial, Astrocytic, and APOE Contributions Along the Alzheimer's Disease Continuum. J Alzheimers Dis 2023; 92:1385-1397. [PMID: 36911936 DOI: 10.3233/jad-221010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
BACKGROUND Many transversal mechanisms act synergistically at different time-points in the cascade of Alzheimer's disease (AD), since amyloid-β (Aβ) deposition, tau pathology, and neuroinflammation influence each other. OBJECTIVE We explored the contributions of microglia and astrocytes in patients with symptomatic sporadic AD stratified according to AT(N) system and APOE genotype. METHODS We compared the cerebrospinal fluid (CSF) levels of sTREM-2 and markers of astrocytic activation (GFAP; β-S100) from 71 patients with AD (23 A+T-,48 A+T+; 38 APOEɛ3, 33 APOEɛ4) and 30 healthy controls (HC). With multivariate analyses we investigated associations between glial biomarkers, Aβ42, and p-tau in all subgroups. RESULTS CSF sTREM-2 was higher in A+T+ [1.437 (0.264)] and A+T- [1.355 (0.213)] than in HC [1.042 (0.198); both p < 0.001]; GFAP and β-S100 were comparable across groups. Considering all patients, sTREM-2 positively associated with Aβ 42 (p = 0.04) and p-tau (=0.016), with the first being present only in the A+T- subgroup (p = 0.023). GFAP positively associated with Aβ 42 in all patients (p = 0.020) and in the A+T+ subgroup (p = 0.04). Stratifying by APOE, a positive association of sTREM-2 and p-tau was confirmed selectively in carriers of ɛ4 (p = 0.018). Finally, sTREM-2 positively correlated with β-S100 in all subgroups, and with GFAP in A+T+ (p = 0.042). CONCLUSION Our results confirm the increase of CSF sTREM-2 in AD, which associates with reduced amyloidopathy in A+T- patients. Moreover, microglial activation seems to increase CSF tau levels in carriers of APOEɛ4, is associated with astrocytic reactivity (GFAP) in A+T+, and likely leads the acquisition of a more neurotoxic astrocytic phenotype (β-S100).
Collapse
Affiliation(s)
| | - Martina Assogna
- UOSD Centro Demenze, Policlinico Tor Vergata, University of Rome "Tor Vergata", Rome, Italy.,Non Invasive Brain Stimulation Unit, IRCCS Santa Lucia, Rome, Italy
| | - Martina Gaia Di Donna
- UOSD Centro Demenze, Policlinico Tor Vergata, University of Rome "Tor Vergata", Rome, Italy
| | - Francesca Bernocchi
- UOSD Centro Demenze, Policlinico Tor Vergata, University of Rome "Tor Vergata", Rome, Italy
| | - Vincenzo De Lucia
- UOSD Centro Demenze, Policlinico Tor Vergata, University of Rome "Tor Vergata", Rome, Italy
| | - Marzia Nuccetelli
- Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Denise Fiorelli
- Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Stefano Loizzo
- Department of Cardiovascular, Endocrine-Metabolic and Ageing-Associated Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Nicola Biagio Mercuri
- Neurology Unit, Policlinico Tor Vergata, University of Rome "Tor Vergata", Rome, Italy
| | - Giacomo Koch
- Non Invasive Brain Stimulation Unit, IRCCS Santa Lucia, Rome, Italy.,Human Physiology Unit, Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Alessandro Martorana
- UOSD Centro Demenze, Policlinico Tor Vergata, University of Rome "Tor Vergata", Rome, Italy
| | - Caterina Motta
- UOSD Centro Demenze, Policlinico Tor Vergata, University of Rome "Tor Vergata", Rome, Italy
| |
Collapse
|
23
|
Zhou S, Zhou X, Jiang Z, Ma J, Li Y, Qian Z, Li H. The Mechanism of SNHG8/Microrna-421-3p/Sorting Nexin 8 Axis on Dopaminergic Neurons in Substantia Nigra in a Mouse Model of Parkinson's Disease. Neurochem Res 2023; 48:942-955. [PMID: 36401052 DOI: 10.1007/s11064-022-03795-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 10/09/2022] [Accepted: 10/14/2022] [Indexed: 11/19/2022]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disease affecting the aging population. Particularly, long non-coding RNAs (lncRNAs) have been demonstrated to play vital roles in PD, while the role of lncRNA SNHG8 in PD remains to be further explored. C57BL/6 mice were induced by rotenone to establish a PD model in vivo, and then the dopaminergic (DA) neuronal damage and locomotor dysfunction in rotenone-treated mice were evaluated. Murine DA cell line MN9D was treated with rotenone to establish a cellular PD model in vitro. Then, the viability, apoptosis, mitochondrial dysfunction, endoplasmic reticulum stress, and autophagy in rotenone-treated MN9D cells were assessed. Expression levels of SNHG8, microRNA-421-3p (miR-421-3p), and sorting nexin 8 (SNX8) in the substantia nigra (SN) of PD mice and rotenone-treated MN9D cells were detected. The interaction between SNHG8 and miR-421-3p, and the targeting relationship between SNX8 and miR-421-3p were confirmed. SNHG8 and SNX8 expression levels were decreased while miR-421-3p expression level was increased in the SN of PD mice and rotenone-treated MN9D cells. Upregulated SNHG8 ameliorated dopaminergic neuron damage and locomotor dysfunction in PD mice. Meanwhile, upregulated SNHG8 enhanced viability, diminished apoptosis, and alleviated mitochondrial dysfunction, endoplasmic reticulum stress, and autophagy in rotenone-treated MN9D cells. Mechanistically, SNHG8 bound to miR-421-3p, and miR-421-3p targeted SNX8. Overexpressed SNHG8 downregulates miR-421-3p to alleviate rotenone-induced dopaminergic neuron injury in PD via upregulating SNX8.
Collapse
Affiliation(s)
- Siwei Zhou
- Geriatric Rehabilitation Center, Zhejiang Rehabilitation Medical Center, No.2828 Binsheng Road, Hangzhou, 310051, Zhejiang, China
| | - Xiaofang Zhou
- Geriatric Rehabilitation Center, Zhejiang Rehabilitation Medical Center, No.2828 Binsheng Road, Hangzhou, 310051, Zhejiang, China.
| | - Zewen Jiang
- Outpatient Department, Zhejiang Armed Police Corps Hospital, No.86 Jiangnan Road, Hangzhou, Zhejiang, China
| | - Jinrong Ma
- Geriatric Rehabilitation Center, Zhejiang Rehabilitation Medical Center, No.2828 Binsheng Road, Hangzhou, 310051, Zhejiang, China
| | - Yuanmei Li
- Geriatric Rehabilitation Center, Zhejiang Rehabilitation Medical Center, No.2828 Binsheng Road, Hangzhou, 310051, Zhejiang, China
| | - Zhiyong Qian
- Geriatric Rehabilitation Center, Zhejiang Rehabilitation Medical Center, No.2828 Binsheng Road, Hangzhou, 310051, Zhejiang, China
| | - Hua Li
- Geriatric Rehabilitation Center, Zhejiang Rehabilitation Medical Center, No.2828 Binsheng Road, Hangzhou, 310051, Zhejiang, China
| |
Collapse
|
24
|
Haider A, Elghazawy NH, Dawoud A, Gebhard C, Wichmann T, Sippl W, Hoener M, Arenas E, Liang SH. Translational molecular imaging and drug development in Parkinson's disease. Mol Neurodegener 2023; 18:11. [PMID: 36759912 PMCID: PMC9912681 DOI: 10.1186/s13024-023-00600-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 01/23/2023] [Indexed: 02/11/2023] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder that primarily affects elderly people and constitutes a major source of disability worldwide. Notably, the neuropathological hallmarks of PD include nigrostriatal loss and the formation of intracellular inclusion bodies containing misfolded α-synuclein protein aggregates. Cardinal motor symptoms, which include tremor, rigidity and bradykinesia, can effectively be managed with dopaminergic therapy for years following symptom onset. Nonetheless, patients ultimately develop symptoms that no longer fully respond to dopaminergic treatment. Attempts to discover disease-modifying agents have increasingly been supported by translational molecular imaging concepts, targeting the most prominent pathological hallmark of PD, α-synuclein accumulation, as well as other molecular pathways that contribute to the pathophysiology of PD. Indeed, molecular imaging modalities such as positron emission tomography (PET) and single-photon emission computed tomography (SPECT) can be leveraged to study parkinsonism not only in animal models but also in living patients. For instance, mitochondrial dysfunction can be assessed with probes that target the mitochondrial complex I (MC-I), while nigrostriatal degeneration is typically evaluated with probes designed to non-invasively quantify dopaminergic nerve loss. In addition to dopaminergic imaging, serotonin transporter and N-methyl-D-aspartate (NMDA) receptor probes are increasingly used as research tools to better understand the complexity of neurotransmitter dysregulation in PD. Non-invasive quantification of neuroinflammatory processes is mainly conducted by targeting the translocator protein 18 kDa (TSPO) on activated microglia using established imaging agents. Despite the overwhelming involvement of the brain and brainstem, the pathophysiology of PD is not restricted to the central nervous system (CNS). In fact, PD also affects various peripheral organs such as the heart and gastrointestinal tract - primarily via autonomic dysfunction. As such, research into peripheral biomarkers has taken advantage of cardiac autonomic denervation in PD, allowing the differential diagnosis between PD and multiple system atrophy with probes that visualize sympathetic nerve terminals in the myocardium. Further, α-synuclein has recently gained attention as a potential peripheral biomarker in PD. This review discusses breakthrough discoveries that have led to the contemporary molecular concepts of PD pathophysiology and how they can be harnessed to develop effective imaging probes and therapeutic agents. Further, we will shed light on potential future trends, thereby focusing on potential novel diagnostic tracers and disease-modifying therapeutic interventions.
Collapse
Affiliation(s)
- Achi Haider
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA.
- Department of Radiology and Imaging Sciences, Emory University, 101 Woodruff Circle, Atlanta, GA, 30322, USA.
| | - Nehal H Elghazawy
- Biochemistry Department, Faculty of Pharmacy and Biotechnology, German University in Cairo, Main Entrance of Al-Tagamoa Al-Khames, Cairo, 11835, Egypt
- Molecular Genetics Research Team (MGRT), Pharmaceutical Biology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo, Main Entrance of Al-Tagamoa Al-Khames, Cairo, 11835, Egypt
| | - Alyaa Dawoud
- Biochemistry Department, Faculty of Pharmacy and Biotechnology, German University in Cairo, Main Entrance of Al-Tagamoa Al-Khames, Cairo, 11835, Egypt
- Molecular Genetics Research Team (MGRT), Pharmaceutical Biology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo, Main Entrance of Al-Tagamoa Al-Khames, Cairo, 11835, Egypt
| | - Catherine Gebhard
- Department of Nuclear Medicine, University Hospital Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Thomas Wichmann
- Department of Neurology/School of Medicine, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Wolfgang Sippl
- Institute of Pharmacy, Department of Medicinal Chemistry, Martin-Luther-University Halle-Wittenberg, W.-Langenbeck-Str. 4, 06120, Halle, Germany
| | - Marius Hoener
- Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - Ernest Arenas
- Karolinska Institutet, MBB, Molecular Neurobiology, Stockholm, Sweden
| | - Steven H Liang
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA.
- Department of Radiology and Imaging Sciences, Emory University, 101 Woodruff Circle, Atlanta, GA, 30322, USA.
| |
Collapse
|
25
|
Giuliano C, Cerri S, Cesaroni V, Blandini F. Relevance of Biochemical Deep Phenotyping for a Personalised Approach to Parkinson's Disease. Neuroscience 2023; 511:100-109. [PMID: 36572171 DOI: 10.1016/j.neuroscience.2022.12.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 10/05/2022] [Accepted: 12/19/2022] [Indexed: 12/25/2022]
Abstract
Parkinson's disease (PD) is a multifactorial neurodegenerative disorder characterised by the progressive loss of dopaminergic neurons in the nigrostriatal tract. The identification of disease-modifying therapies is the Holy Grail of PD research, but to date no drug has been approved as such a therapy. A possible reason is the remarkable phenotypic heterogeneity of PD patients, which can generate confusion in the interpretation of results or even mask the efficacy of a therapeutic intervention. This heterogeneity should be taken into account in clinical trials, stratifying patients by their expected response to drugs designed to engage selected molecular targets. In this setting, stratification methods (clinical and genetic) should be supported by biochemical phenotyping of PD patients, in line with the deep phenotyping concept. Collection, from single patients, of a range of biological samples would streamline the generation of these profiles. Several studies have proposed biochemical characterisations of patient cohorts based on analysis of blood, cerebrospinal fluid, urine, stool, saliva and skin biopsy samples, with extracellular vesicles attracting increasing interest as a source of biomarkers. In this review we report and critically discuss major studies that used a biochemical approach to stratify their PD cohorts. The analyte most studied is α-synuclein, while other studies have focused on neurofilament light chain, lysosomal proteins, inflammasome-related proteins, LRRK2 and the urinary proteome. At present, stratification of PD patients, while promising, is still a nascent approach. Deep phenotyping of patients will allow clinical researchers to identify homogeneous subgroups for the investigation of tailored disease-modifying therapies, enhancing the chances of therapeutic success.
Collapse
Affiliation(s)
- Claudio Giuliano
- Unit of Cellular and Molecular Neurobiology, IRCCS Mondino Foundation, 27100 Pavia, Italy
| | - Silvia Cerri
- Unit of Cellular and Molecular Neurobiology, IRCCS Mondino Foundation, 27100 Pavia, Italy
| | - Valentina Cesaroni
- Unit of Cellular and Molecular Neurobiology, IRCCS Mondino Foundation, 27100 Pavia, Italy
| | - Fabio Blandini
- Unit of Cellular and Molecular Neurobiology, IRCCS Mondino Foundation, 27100 Pavia, Italy; Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy.
| |
Collapse
|
26
|
Tao QQ, Lin RR, Wu ZY. Early Diagnosis of Alzheimer's Disease: Moving Toward a Blood-Based Biomarkers Era. Clin Interv Aging 2023; 18:353-358. [PMID: 36911809 PMCID: PMC10001034 DOI: 10.2147/cia.s394821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 02/13/2023] [Indexed: 03/08/2023] Open
Affiliation(s)
- Qing-Qing Tao
- Department of Neurology and Research Center of Neurology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, People's Republic of China
| | - Rong-Rong Lin
- Department of Neurology and Research Center of Neurology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, People's Republic of China
| | - Zhi-Ying Wu
- Department of Neurology and Research Center of Neurology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, People's Republic of China.,MOE Frontier Science Center for Brain Science and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, People's Republic of China
| |
Collapse
|
27
|
Wang K, Cao X, Li Z, Liu S, Zhou Y, Guo L, Li P. Anesthesia and surgery-induced elevation of CSF sTREM2 is associated with early cognitive dysfunction after thoracoabdominal aortic dissection surgery. BMC Anesthesiol 2022; 22:413. [PMID: 36585610 PMCID: PMC9805002 DOI: 10.1186/s12871-022-01955-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 12/21/2022] [Indexed: 01/01/2023] Open
Abstract
PURPOSE Soluble triggering receptor expressed on myeloid cells 2 (sTREM2) concentration is increased in cerebrospinal fluid (CSF) in early symptomatic phase of Alzheimer's disease (AD). This study investigated whether CSF sTREM2 has a relationship with early cognitive dysfunction following surgery in cardiac surgery patients. METHODS A total of 82 patients undergoing thoracoabdominal aortic replacement were recruited in this study. Neuropsychological testing battery was conducted before and after surgery. Postoperative cognitive dysfunction (POCD) was defined as a Z-score > 1.96 on at least 2 different tests or Telephone Interviews for Cognitive Status-Modified (TICS-M) score < 27. The CSF and serum sTREM2, Aβ42, T-tau and P-tau were collected and measured by ELISA on day before surgery and postoperative day 3. RESULTS Patients were classified into POCD (n = 34) and non-POCD (n = 48) groups according to Z-score. Compared to non-POCD group, the levels of CSF sTREM2 (p < 0.001) and serum sTREM2 (p = 0.001) were significantly higher in POCD group on postoperative day 3. The levels of Aβ42 (p = 0.005) and Aβ42/T-tau ratio (p = 0.036) were significantly lower in POCD group on postoperative day 3. Multivariate logistic regression analysis revealed that higher value of postoperative CSF sTREM2 (odds ratio: 1.06, 95% confidence interval: 1.02-1.11, p = 0.009), age (OR: 1.15, 95%CI: 1.03-1.28, p = 0.014) and POD duration (OR: 2.47, 95%CI: 1.15-5.29, p = 0.02) were the risk factors of POCD. CONCLUSION This study indicates that anesthesia and surgery-induced elevation of CSF sTREM2 is associated with an increased risk of early cognitive dysfunction following surgery.
Collapse
Affiliation(s)
- Kexin Wang
- grid.412636.40000 0004 1757 9485Department of Anesthesiology, the First Hospital of China Medical University, Shenyang, Liaoning China
| | - Xuezhao Cao
- grid.412636.40000 0004 1757 9485Department of Anesthesiology, the First Hospital of China Medical University, Shenyang, Liaoning China
| | - Zhe Li
- grid.412636.40000 0004 1757 9485Department of Anesthesiology, the First Hospital of China Medical University, Shenyang, Liaoning China
| | - Sidan Liu
- grid.412636.40000 0004 1757 9485Department of Anesthesiology, the First Hospital of China Medical University, Shenyang, Liaoning China
| | - Yongjian Zhou
- grid.412636.40000 0004 1757 9485Department of Anesthesiology, the First Hospital of China Medical University, Shenyang, Liaoning China
| | - Lili Guo
- grid.412636.40000 0004 1757 9485Department of Anesthesiology, the First Hospital of China Medical University, Shenyang, Liaoning China
| | - Pengli Li
- grid.412636.40000 0004 1757 9485Department of Anesthesiology, the First Hospital of China Medical University, Shenyang, Liaoning China
| |
Collapse
|
28
|
Wilson EN, Young CB, Ramos Benitez J, Swarovski MS, Feinstein I, Vandijck M, Le Guen Y, Kasireddy NM, Shahid M, Corso NK, Wang Q, Kennedy G, Trelle AN, Lind B, Channappa D, Belnap M, Ramirez V, Skylar-Scott I, Younes K, Yutsis MV, Le Bastard N, Quinn JF, van Dyck CH, Nairn A, Fredericks CA, Tian L, Kerchner GA, Montine TJ, Sha SJ, Davidzon G, Henderson VW, Longo FM, Greicius MD, Wagner AD, Wyss-Coray T, Poston KL, Mormino EC, Andreasson KI. Performance of a fully-automated Lumipulse plasma phospho-tau181 assay for Alzheimer's disease. Alzheimers Res Ther 2022; 14:172. [PMID: 36371232 PMCID: PMC9652927 DOI: 10.1186/s13195-022-01116-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 10/31/2022] [Indexed: 11/13/2022]
Abstract
BACKGROUND The recent promise of disease-modifying therapies for Alzheimer's disease (AD) has reinforced the need for accurate biomarkers for early disease detection, diagnosis and treatment monitoring. Advances in the development of novel blood-based biomarkers for AD have revealed that plasma levels of tau phosphorylated at various residues are specific and sensitive to AD dementia. However, the currently available tests have shortcomings in access, throughput, and scalability that limit widespread implementation. METHODS We evaluated the diagnostic and prognostic performance of a high-throughput and fully-automated Lumipulse plasma p-tau181 assay for the detection of AD. Plasma from older clinically unimpaired individuals (CU, n = 463) and patients with mild cognitive impairment (MCI, n = 107) or AD dementia (n = 78) were obtained from the longitudinal Stanford University Alzheimer's Disease Research Center (ADRC) and the Stanford Aging and Memory Study (SAMS) cohorts. We evaluated the discriminative accuracy of plasma p-tau181 for clinical AD diagnosis, association with amyloid β peptides and p-tau181 concentrations in CSF, association with amyloid positron emission tomography (PET), and ability to predict longitudinal cognitive and functional change. RESULTS The assay showed robust performance in differentiating AD from control participants (AUC 0.959, CI: 0.912 to 0.990), and was strongly associated with CSF p-tau181, CSF Aβ42/Aβ40 ratio, and amyloid-PET global SUVRs. Associations between plasma p-tau181 with CSF biomarkers were significant when examined separately in Aβ+ and Aβ- groups. Plasma p-tau181 significantly increased over time in CU and AD diagnostic groups. After controlling for clinical diagnosis, age, sex, and education, baseline plasma p-tau181 predicted change in MoCA overall and change in CDR Sum of Boxes in the AD group over follow-up of up to 5 years. CONCLUSIONS This fully-automated and available blood-based biomarker assay therefore may be useful for early detection, diagnosis, prognosis, and treatment monitoring of AD.
Collapse
Affiliation(s)
- Edward N. Wilson
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA ,grid.168010.e0000000419368956Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA USA
| | - Christina B. Young
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA
| | - Javier Ramos Benitez
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA
| | - Michelle S. Swarovski
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA
| | - Igor Feinstein
- grid.168010.e0000000419368956Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA USA
| | | | - Yann Le Guen
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA
| | - Nandita M. Kasireddy
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA
| | - Marian Shahid
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA
| | - Nicole K. Corso
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA
| | - Qian Wang
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA
| | - Gabriel Kennedy
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA
| | - Alexandra N. Trelle
- grid.168010.e0000000419368956Psychology, Stanford University, Stanford, CA USA
| | - Betty Lind
- grid.410404.50000 0001 0165 2383Neurology, Portland VA Medical Center, Portland, OR USA ,grid.5288.70000 0000 9758 5690Neurology, Oregon Health & Science University, Portland, OR USA
| | - Divya Channappa
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA ,grid.168010.e0000000419368956Pathology, Stanford University, Stanford, CA USA
| | - Malia Belnap
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA
| | - Veronica Ramirez
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA
| | - Irina Skylar-Scott
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA
| | - Kyan Younes
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA
| | - Maya V. Yutsis
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA
| | | | - Joseph F. Quinn
- grid.410404.50000 0001 0165 2383Neurology, Portland VA Medical Center, Portland, OR USA ,grid.5288.70000 0000 9758 5690Neurology, Oregon Health & Science University, Portland, OR USA
| | | | - Angus Nairn
- grid.47100.320000000419368710Psychiatry, Yale University, New Haven, CT USA
| | - Carolyn A. Fredericks
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA
| | - Lu Tian
- grid.168010.e0000000419368956Biomedical Data Science, Stanford University, Stanford, CA USA
| | - Geoffrey A. Kerchner
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA
| | - Thomas J. Montine
- grid.168010.e0000000419368956Pathology, Stanford University, Stanford, CA USA
| | - Sharon J. Sha
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA
| | - Guido Davidzon
- grid.168010.e0000000419368956Radiology, Stanford University, Stanford, CA USA
| | - Victor W. Henderson
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA ,grid.168010.e0000000419368956Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA USA ,grid.168010.e0000000419368956Epidemiology & Population Health, Stanford University, Stanford, CA USA
| | - Frank M. Longo
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA ,grid.168010.e0000000419368956Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA USA
| | - Michael D. Greicius
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA ,grid.168010.e0000000419368956Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA USA
| | - Anthony D. Wagner
- grid.168010.e0000000419368956Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA USA ,grid.168010.e0000000419368956Psychology, Stanford University, Stanford, CA USA
| | - Tony Wyss-Coray
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA ,grid.168010.e0000000419368956Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA USA
| | - Kathleen L. Poston
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA ,grid.168010.e0000000419368956Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA USA
| | - Elizabeth C. Mormino
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA ,grid.168010.e0000000419368956Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA USA
| | - Katrin I. Andreasson
- grid.168010.e0000000419368956Neurology & Neurological Sciences, Stanford University, Stanford, CA USA ,grid.168010.e0000000419368956Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA USA ,grid.499295.a0000 0004 9234 0175Chan Zuckerberg Biohub, San Francisco, CA 94158 USA
| |
Collapse
|
29
|
Williams GP, Schonhoff AM, Sette A, Lindestam Arlehamn CS. Central and Peripheral Inflammation: Connecting the Immune Responses of Parkinson’s Disease. JOURNAL OF PARKINSON'S DISEASE 2022; 12:S129-S136. [PMID: 35754290 PMCID: PMC9535591 DOI: 10.3233/jpd-223241] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Inflammation has increasingly become a focus of study in regards to Parkinson’s disease (PD). Moreover, both central and peripheral sources of inflammation have been implicated in the pathogenesis of PD. Central inflammation consisting of activated microglia, astroglia, and T cell responses within the PD central nervous system; and peripheral inflammation referring to activated innate cells and T cell signaling in the enteric nervous system, gastrointestinal tract, and blood. This review will highlight important work that further implicates central and peripheral inflammation in playing a role in PD. We also discuss how these two distant inflammations appear related and how that may be mediated by autoantigenic responses to α-syn.
Collapse
Affiliation(s)
- Gregory P. Williams
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Aubrey M. Schonhoff
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Cecilia S. Lindestam Arlehamn
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| |
Collapse
|
30
|
Barragán-Álvarez CP, Flores-Fernandez JM, Hernández-Pérez OR, Ávila-Gónzalez D, Díaz NF, Padilla-Camberos E, Dublan-García O, Gómez-Oliván LM, Diaz-Martinez NE. Recent advances in the use of CRISPR/Cas for understanding the early development of molecular gaps in glial cells. Front Cell Dev Biol 2022; 10:947769. [PMID: 36120556 PMCID: PMC9479146 DOI: 10.3389/fcell.2022.947769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 08/01/2022] [Indexed: 12/03/2022] Open
Abstract
Glial cells are non-neuronal elements of the nervous system (NS) and play a central role in its development, maturation, and homeostasis. Glial cell interest has increased, leading to the discovery of novel study fields. The CRISPR/Cas system has been widely employed for NS understanding. Its use to study glial cells gives crucial information about their mechanisms and role in the central nervous system (CNS) and neurodegenerative disorders. Furthermore, the increasingly accelerated discovery of genes associated with the multiple implications of glial cells could be studied and complemented with the novel screening methods of high-content and single-cell screens at the genome-scale as Perturb-Seq, CRISP-seq, and CROPseq. Besides, the emerging methods, GESTALT, and LINNAEUS, employed to generate large-scale cell lineage maps have yielded invaluable information about processes involved in neurogenesis. These advances offer new therapeutic approaches to finding critical unanswered questions about glial cells and their fundamental role in the nervous system. Furthermore, they help to better understanding the significance of glial cells and their role in developmental biology.
Collapse
Affiliation(s)
- Carla Patricia Barragán-Álvarez
- Laboratorio de Reprogramación Celular y Bioingeniería de Tejidos, Biotecnología Médica y Farmacéutica, Centro de Investigación y Asistencia en Tecnología y Diseño Del Estado de Jalisco, Guadalajara, Mexico
| | - José Miguel Flores-Fernandez
- Departamento de Investigación e Innovación, Universidad Tecnológica de Oriental, Oriental, Mexico
- Department of Biochemistry & Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB, Canada
| | | | - Daniela Ávila-Gónzalez
- Laboratorio de Reprogramación Celular y Bioingeniería de Tejidos, Biotecnología Médica y Farmacéutica, Centro de Investigación y Asistencia en Tecnología y Diseño Del Estado de Jalisco, Guadalajara, Mexico
- Departamento de Fisiología y Desarrollo Celular, Instituto Nacional de Perinatología, México City, Mexico
| | - Nestor Fabian Díaz
- Departamento de Fisiología y Desarrollo Celular, Instituto Nacional de Perinatología, México City, Mexico
| | - Eduardo Padilla-Camberos
- Laboratorio de Reprogramación Celular y Bioingeniería de Tejidos, Biotecnología Médica y Farmacéutica, Centro de Investigación y Asistencia en Tecnología y Diseño Del Estado de Jalisco, Guadalajara, Mexico
| | - Octavio Dublan-García
- Laboratorio de Alimentos y Toxicología Ambiental, Facultad de Química, Universidad Autónoma Del Estado de México, Toluca, México
| | - Leobardo Manuel Gómez-Oliván
- Laboratorio de Alimentos y Toxicología Ambiental, Facultad de Química, Universidad Autónoma Del Estado de México, Toluca, México
| | - Nestor Emmanuel Diaz-Martinez
- Laboratorio de Reprogramación Celular y Bioingeniería de Tejidos, Biotecnología Médica y Farmacéutica, Centro de Investigación y Asistencia en Tecnología y Diseño Del Estado de Jalisco, Guadalajara, Mexico
| |
Collapse
|
31
|
Mehta NH, Suss RA, Dyke JP, Theise ND, Chiang GC, Strauss S, Saint-Louis L, Li Y, Pahlajani S, Babaria V, Glodzik L, Carare RO, de Leon MJ. Quantifying cerebrospinal fluid dynamics: A review of human neuroimaging contributions to CSF physiology and neurodegenerative disease. Neurobiol Dis 2022; 170:105776. [PMID: 35643187 PMCID: PMC9987579 DOI: 10.1016/j.nbd.2022.105776] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 05/21/2022] [Indexed: 01/13/2023] Open
Abstract
Cerebrospinal fluid (CSF), predominantly produced in the ventricles and circulating throughout the brain and spinal cord, is a key protective mechanism of the central nervous system (CNS). Physical cushioning, nutrient delivery, metabolic waste, including protein clearance, are key functions of the CSF in humans. CSF volume and flow dynamics regulate intracranial pressure and are fundamental to diagnosing disorders including normal pressure hydrocephalus, intracranial hypotension, CSF leaks, and possibly Alzheimer's disease (AD). The ability of CSF to clear normal and pathological proteins, such as amyloid-beta (Aβ), tau, alpha synuclein and others, implicates it production, circulation, and composition, in many neuropathologies. Several neuroimaging modalities have been developed to probe CSF fluid dynamics and better relate CSF volume and flow to anatomy and clinical conditions. Approaches include 2-photon microscopic techniques, MRI (tracer-based, gadolinium contrast, endogenous phase-contrast), and dynamic positron emission tomography (PET) using existing approved radiotracers. Here, we discuss CSF flow neuroimaging, from animal models to recent clinical-research advances, summarizing current endeavors to quantify and map CSF flow with implications towards pathophysiology, new biomarkers, and treatments of neurological diseases.
Collapse
Affiliation(s)
- Neel H Mehta
- Department of Biology, Cornell University, Ithaca, NY, USA
| | - Richard A Suss
- Division of Neuroradiology, Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jonathan P Dyke
- Citigroup Biomedical Imaging Center, Weill Cornell Medicine, New York, NY, USA
| | - Neil D Theise
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
| | - Gloria C Chiang
- Division of Neuroradiology, Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Sara Strauss
- Division of Neuroradiology, Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | | | - Yi Li
- Brain Health Imaging Institute, Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Silky Pahlajani
- Brain Health Imaging Institute, Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Vivek Babaria
- Orange County Spine and Sports, Interventional Physiatry, Newport Beach, CA, USA
| | - Lidia Glodzik
- Brain Health Imaging Institute, Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Roxana O Carare
- Department of Medicine, University of Southampton, Southampton, UK
| | - Mony J de Leon
- Brain Health Imaging Institute, Department of Radiology, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
32
|
Khezri MR, Ghasemnejad-Berenji M. Icariin: A Potential Neuroprotective Agent in Alzheimer's Disease and Parkinson's Disease. Neurochem Res 2022; 47:2954-2962. [PMID: 35802286 DOI: 10.1007/s11064-022-03667-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 06/16/2022] [Accepted: 06/18/2022] [Indexed: 11/28/2022]
Abstract
Alzheimer's disease (AD) and Parkinson's disease (PD) are the most common neurodegenerative diseases worldwide. They are characterized by the loss of neurons and synapses in special parts of the central nervous system (CNS). There is no definitive treatment for AD and PD, but extensive studies are underway to identify the effective drugs which can slow the progression of these diseases by affecting the factors involved in their pathophysiology (i.e., aggregated proteins, neuroinflammation, and oxidative stress). Icariin, a natural compound isolated from Epimedii herba, is known because of its anti-inflammatory and anti-oxidant properties. In this regard, there are numerous studies indicating its potential as a natural compound against the progression of CNS disorders, such as neurodegenerative diseases. Therefore, this review aims to re-examine findings on the pharmacologic effects of icariin on factors involved in the pathophysiology of AD and PD.
Collapse
Affiliation(s)
| | - Morteza Ghasemnejad-Berenji
- Department of Pharmacology and Toxicology, School of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran. .,Research Center for Experimental and Applied Pharmaceutical Sciences, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
33
|
The role of Triggering Receptor Expressed on Myeloid Cells 2 in Parkinson's disease and other neurodegenerative disorders. Behav Brain Res 2022; 433:113977. [PMID: 35752274 DOI: 10.1016/j.bbr.2022.113977] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 06/09/2022] [Accepted: 06/21/2022] [Indexed: 11/23/2022]
Abstract
Parkinson's disease (PD) is a progressive neurological disorder marked by cardinal clinical symptoms such as rigor, tremor, and akinesia. Albeit a loss of dopaminergic neurons from the substantia nigra pars compacta is causative for the movement impairments found in patients, molecular reasoning for this loss is still incomplete. In recent years, triggering factor expressed on myeloid cells (TREM2) gained attention in the field of neurodegeneration as it could be associated with different neurodegenerative disorders. Primarily identified as a risk factor in Alzheimer's disease, variants in TREM2 were linked to PD and multiple sclerosis, too. Expressed on phagocytic cells, such as macrophages and microglia, TREM2 puts the focus on inflammation associated conditions in PD and provides a molecular target that could at least partly explain the role of immune cells in PD. Here, we summarize expression patterns and molecular functions of TREM2, recapitulate on its role in inflammation, phagocytosis and cell survival, before turning to neurodegenerative disorders with an emphasis on PD.
Collapse
|
34
|
Wong YY, Wu CY, Yu D, Kim E, Wong M, Elez R, Zebarth J, Ouk M, Tan J, Liao J, Haydarian E, Li S, Fang Y, Li P, Pakosh M, Tartaglia MC, Masellis M, Swardfager W. Biofluid markers of blood-brain barrier disruption and neurodegeneration in Lewy body spectrum diseases: A systematic review and meta-analysis. Parkinsonism Relat Disord 2022; 101:119-128. [PMID: 35760718 DOI: 10.1016/j.parkreldis.2022.06.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 06/01/2022] [Accepted: 06/08/2022] [Indexed: 02/06/2023]
Abstract
BACKGROUND Mixed evidence supports blood-brain barrier (BBB) dysfunction in Lewy body spectrum diseases. METHODS We compare biofluid markers in people with idiopathic Parkinson's disease (PD) and people with PD dementia (PDD) and/or dementia with Lewy bodies (DLB), compared with healthy controls (HC). Seven databases were searched up to May 10, 2021. Outcomes included cerebrospinal fluid to blood albumin ratio (Qalb), and concentrations of 7 blood protein markers that also reflect BBB disruption and/or neurodegenerative co-pathology. We further explore differences between PD patients with and without evidence of dementia. Random-effects models were used to obtain standardized mean differences (SMD) with 95% confidence interval. RESULTS Of 13,949 unique records, 51 studies were meta-analyzed. Compared to HC, Qalb was higher in PD (NPD/NHC = 224/563; SMD = 0.960 [0.227-1.694], p = 0.010; I2 = 92.2%) and in PDD/DLB (NPDD/DLB/NHC = 265/670; SMD = 1.126 [0.358-1.893], p < 0.001; I2 = 78.2%). Blood neurofilament light chain (NfL) was higher in PD (NPD/NHC = 1848/1130; SMD = 0.747 [0.442-1.052], p < 0.001; I2 = 91.9%) and PDD/DLB (NPDD/DLB/NHC = 183/469; SMD = 1.051 [0.678-1.423], p = 0.004; I2 = 92.7%) than in HC. p-tau 181 (NPD/NHC = 276/164; SMD = 0.698 [0.149-1.247], p = 0.013; I2 = 82.7%) was also higher in PD compared to HC. In exploratory analyses, blood NfL was higher in PD without dementia (NPDND/NHC = 1005/740; SMD = 0.252 [0.042-0.462], p = 0.018; I2 = 71.8%) and higher in PDD (NPDD/NHC = 100/111; SMD = 0.780 [0.347-1.214], p < 0.001; I2 = 46.7%) compared to HC. Qalb (NPDD/NPDND = 63/191; SMD = 0.482 [0.189-0.774], p = 0.010; I2<0.001%) and NfL (NPDD/NPDND = 100/223; SMD = 0.595 [0.346-0.844], p < 0.001; I2 = 3.4%) were higher in PDD than in PD without dementia. CONCLUSIONS Biofluid markers suggest BBB disruption and neurodegenerative co-pathology involvement in common Lewy body diseases. Greater evidence of BBB breakdown was seen in Lewy body disease with cognitive impairment.
Collapse
Affiliation(s)
- Yuen Yan Wong
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada; Dr. Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Che-Yuan Wu
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada; Dr. Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Di Yu
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada; Dr. Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Esther Kim
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Melissa Wong
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Renata Elez
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Julia Zebarth
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Michael Ouk
- Dr. Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Jocelyn Tan
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Jiamin Liao
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Eileen Haydarian
- Dr. Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Siming Li
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Yaolu Fang
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Peihao Li
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Maureen Pakosh
- Library & Information Services, UHN Toronto Rehabilitation Institute, Toronto, Ontario, Canada
| | - Maria Carmela Tartaglia
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
| | - Mario Masellis
- Dr. Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada; Department of Medicine (Neurology), Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Walter Swardfager
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada; Dr. Sandra Black Centre for Brain Resilience and Recovery, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada; KITE UHN Toronto Rehabilitation Institute, Toronto, Ontario, Canada.
| |
Collapse
|
35
|
Qin Q, Wan H, Wang D, Li J, Qu Y, Zhao J, Li J, Xue Z. The Association of CSF sTREM2 With Cognitive Decline and Its Dynamic Change in Parkinson's Disease: Analysis of the PPMI Cohort. Front Aging Neurosci 2022; 14:892493. [PMID: 35783125 PMCID: PMC9245456 DOI: 10.3389/fnagi.2022.892493] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 05/24/2022] [Indexed: 01/20/2023] Open
Abstract
Background Soluble fragment of triggering receptor expressed on myeloid cells 2 (sTREM2) in cerebrospinal fluid (CSF) is a biomarker of microglial activation and increased in several neurodegenerative diseases. However, the role of sTREM2 in Parkinson's diseases (PDs) remains unclear. This study aims to investigate whether CSF sTREM2 is changed during the pathology of PD and its association with cognitive decline. Methods We recruited 219 de novo patients with PD and 100 healthy controls from Parkinson's Progression Markers Initiative (PPMI). Cross-sectional and longitudinal associations between cognition and CSF sTREM2 were evaluated using multivariable-adjusted models. To assess the changes in CSF sTREM2 during the pathology of PD, patients were classified through the A/T classification framework with addition of α-synuclein (α-syn), which we implemented based on the CSF amyloid β-peptide 1-42 (A) and phosphorylated tau (T) and α-syn (S). Results The CSF sTREM2 did not differ between healthy controls and patients with PD or between PD clinical subgroups (p > 0.05). However, higher baseline CSF sTREM2 predicted greater global cognitive decline in patients with PD (β = -0.585, p = 0.039). Moreover, after a mean follow-up of 5.51 ± 1.31 years, baseline CSF sTREM2 that elevated in the middle tertile (HR = 2.426, 95% CI: 1.023-5.754, p = 0.044) and highest tertile (HR = 2.833, 95% CI: 1.226-6.547, p = 0.015) were associated with a future high risk of cognitive decline. Additionally, CSF sTREM2 decreased in abnormal Aβ pathology (A+) and α-syn pathology (S+) but normal tau pathology, while increased in abnormal phosphorylated tau (T+) (p < 0.05). Conclusion CSF sTREM2 may be a promising predictor for the cognitive decline in PD rather than a diagnostic biomarker. The dynamic change in CSF sTREM2 in PD may help to the monitor of neuronal injury and microglial activity.
Collapse
Affiliation(s)
- Qixiong Qin
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hengming Wan
- Department of General Family Medicine, Liuzhou Worker's Hospital, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, China
| | - Danlei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingyi Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Qu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingwei Zhao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiangting Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zheng Xue
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
36
|
Shuken SR, Rutledge J, Iram T, Losada PM, Wilson EN, Andreasson KI, Leib RD, Wyss-Coray T. Limited Proteolysis-Mass Spectrometry Reveals Aging-Associated Changes in Cerebrospinal Fluid Protein Abundances and Structures. NATURE AGING 2022; 2:379-388. [PMID: 36741774 PMCID: PMC9893943 DOI: 10.1038/s43587-022-00196-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cerebrospinal fluid (CSF) proteins and their structures have been implicated repeatedly in aging and neurodegenerative diseases. Limited proteolysis-mass spectrometry (LiP-MS) is a method that enables proteome-wide screening for changes in both protein abundance and structure. To screen for novel aging-associated changes in the CSF proteome, we performed LiP-MS on CSF from young and old mice with a modified analysis pipeline. We found 38 protein groups change in abundance with aging, most dominantly immunoglobulins of the IgM subclass. We discovered six high-confidence candidates that appeared to change in structure with aging, of which Kng1, Itih2, Lp-PLA2, and 14-3-3 proteins have binding partners or proteoforms known previously to change in the brain with Alzheimer's disease. Intriguingly, using orthogonal validation by Western blot we found the LiP-MS hit Cd5l forms a covalent complex with IgM in mouse and human CSF whose abundance increases with aging. SOMAmer probe signals for all six LiP-MS hits in human CSF, especially 14-3-3 proteins, significantly associate with several clinical features relevant to cognitive function and neurodegeneration. Together, our findings show that LiP-MS can uncover age-related structural changes in CSF with relevance to neurodegeneration.
Collapse
Affiliation(s)
- Steven R. Shuken
- Department of Chemistry, Stanford University, Stanford, CA, USA.,Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Jarod Rutledge
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.,Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Tal Iram
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Patricia Moran Losada
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Edward N. Wilson
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Katrin I. Andreasson
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.,Program in Immunology, Stanford University, Stanford, CA, USA
| | - Ryan D. Leib
- Vincent Coates Foundation Mass Spectrometry Laboratory, Stanford University, Stanford, CA, USA
| | - Tony Wyss-Coray
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.,Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, California, USA.,To whom correspondence should be addressed:
| |
Collapse
|
37
|
Lu Y, Zhao Y, Zhang Q, Fang C, Bao A, Dong W, Peng Y, Peng H, Ju Z, He J, Zhang Y, Xu T, Zhong C. Soluble TREM2 is associated with death and cardiovascular events after acute ischemic stroke: an observational study from CATIS. J Neuroinflammation 2022; 19:88. [PMID: 35414082 PMCID: PMC9006629 DOI: 10.1186/s12974-022-02440-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 03/24/2022] [Indexed: 11/29/2022] Open
Abstract
Background Soluble triggering receptor expressed on myeloid cells 2 (sTREM2), which reflects microglia activation, has been reported closely associated with neuronal injury and neuroinflammation. We aimed to prospectively investigate the associations between plasma sTREM2 and clinical outcomes in acute ischemic stroke (AIS) patients. Methods Study participants were from the China Antihypertensive Trial in Acute Ischemic Stroke, plasma sTREM2 levels in the acute phase of AIS were measured in 3285 participants. The study outcomes were death, cardiovascular events and severe disability at 1 year after AIS. Cox proportional hazards models or logistic regression models were performed to examine the associations of plasma sTREM2 and clinical outcomes. Results After 1-year follow-up, 288 participants (8.8%) experienced cardiovascular events or died. Multivariable-adjusted hazard ratios or odds ratios (95% confidence intervals) for the highest quartile of sTREM2 were 1.57 (1.11–2.21) for the composite outcome of death and cardiovascular events, 1.68 (1.09–2.60) for death, and 1.53 (1.08–2.18) for death or severe disability compared to the lowest quartile. Moreover, incorporation sTREM2 into traditional risk factors model significantly improved risk prediction of the composite outcome of death and cardiovascular events as evidenced by net reclassification index and integrated discrimination improvement (all p values < 0.05). There were joint effects of sTREM2 and galectin-3 on death and cardiovascular events. Participants with simultaneous elevation of sTREM2 and galectin-3 levels had the highest risk of the composite outcome of death and cardiovascular events. Conclusions Elevated sTREM2 levels were independently associated with increased risks of death and cardiovascular events after AIS.
Collapse
Affiliation(s)
- Yaling Lu
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, 199 Renai Road, Industrial Park District, Suzhou, 215123, Jiangsu, China
| | - Yu Zhao
- Department of Orthopaedics, Suzhou Science & Technology Town Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Qi Zhang
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, 199 Renai Road, Industrial Park District, Suzhou, 215123, Jiangsu, China
| | - Chongquan Fang
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, 199 Renai Road, Industrial Park District, Suzhou, 215123, Jiangsu, China
| | - Anran Bao
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, 199 Renai Road, Industrial Park District, Suzhou, 215123, Jiangsu, China
| | - Wenjing Dong
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, 199 Renai Road, Industrial Park District, Suzhou, 215123, Jiangsu, China
| | - Yanbo Peng
- Department of Neurology, Affiliated Hospital of North China University of Science and Technology, Hebei, China
| | - Hao Peng
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, 199 Renai Road, Industrial Park District, Suzhou, 215123, Jiangsu, China
| | - Zhong Ju
- Department of Neurology, Kerqin District First People's Hospital of Tongliao City, Tongliao, Inner Mongolia, China
| | - Jiang He
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA.,Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Yonghong Zhang
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, 199 Renai Road, Industrial Park District, Suzhou, 215123, Jiangsu, China
| | - Tan Xu
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, 199 Renai Road, Industrial Park District, Suzhou, 215123, Jiangsu, China.
| | - Chongke Zhong
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, 199 Renai Road, Industrial Park District, Suzhou, 215123, Jiangsu, China.
| |
Collapse
|
38
|
Dong MH, Zhou LQ, Tang Y, Chen M, Xiao J, Shang K, Deng G, Qin C, Tian DS. CSF sTREM2 in neurological diseases: a two-sample Mendelian randomization study. J Neuroinflammation 2022; 19:79. [PMID: 35382840 PMCID: PMC8985278 DOI: 10.1186/s12974-022-02443-9] [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: 11/09/2021] [Accepted: 03/24/2022] [Indexed: 12/12/2022] Open
Abstract
Background Soluble triggering receptor expressed on myeloid cells 2 (sTREM2) in cerebrospinal fluid (CSF) has been described as a biomarker for microglial activation, which were observed increased in a variety of neurological disorders. Objective Our objective was to explore whether genetically determined CSF sTREM2 levels are causally associated with different neurological diseases by conducting a two-sample Mendelian randomization (MR) study. Methods Single nucleotide polymorphisms significantly associated with CSF sTREM2 levels were selected as instrumental variables to estimate the causal effects on clinically common neurological diseases, including stroke, Alzheimer’s diseases, Parkinson’s diseases, amyotrophic lateral sclerosis, multiple sclerosis, and epilepsy and their subtypes. Summary-level statistics of both exposure and outcomes were applied in an MR framework. Results Genetically predicted per 1 pg/dL increase of CSF sTREM2 levels was associated with higher risk of multiple sclerosis (OR = 1.038, 95%CI = 1.014–1.064, p = 0.002). Null association was found in risk of other included neurological disorders. Conclusions These findings provide support for a potential causal relationship between elevated CSF sTREM2 levels and higher risk of multiple sclerosis. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02443-9.
Collapse
Affiliation(s)
- Ming-Hao Dong
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Luo-Qi Zhou
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yue Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Man Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jun Xiao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ke Shang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Gang Deng
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Chuan Qin
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Dai-Shi Tian
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
39
|
Li TR, Yang Q, Hu X, Han Y. Biomarkers and Tools for Predicting Alzheimer's Disease in the Preclinical Stage. Curr Neuropharmacol 2022; 20:713-737. [PMID: 34030620 PMCID: PMC9878962 DOI: 10.2174/1570159x19666210524153901] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 04/27/2021] [Accepted: 05/08/2021] [Indexed: 11/22/2022] Open
Abstract
Alzheimer's disease (AD) is the only leading cause of death for which no disease-modifying therapy is currently available. Over the past decade, a string of disappointing clinical trial results has forced us to shift our focus to the preclinical stage of AD, which represents the most promising therapeutic window. However, the accurate diagnosis of preclinical AD requires the presence of brain β- amyloid deposition determined by cerebrospinal fluid or amyloid-positron emission tomography, significantly limiting routine screening and diagnosis in non-tertiary hospital settings. Thus, an easily accessible marker or tool with high sensitivity and specificity is highly needed. Recently, it has been discovered that individuals in the late stage of preclinical AD may not be truly "asymptomatic" in that they may have already developed subtle or subjective cognitive decline. In addition, advances in bloodderived biomarker studies have also allowed the detection of pathologic changes in preclinical AD. Exosomes, as cell-to-cell communication messengers, can reflect the functional changes of their source cell. Methodological advances have made it possible to extract brain-derived exosomes from peripheral blood, making exosomes an emerging biomarker carrier and liquid biopsy tool for preclinical AD. The eye and its associated structures have rich sensory-motor innervation. In this regard, studies have indicated that they may also provide reliable markers. Here, our report covers the current state of knowledge of neuropsychological and eye tests as screening tools for preclinical AD and assesses the value of blood and brain-derived exosomes as carriers of biomarkers in conjunction with the current diagnostic paradigm.
Collapse
Affiliation(s)
- Tao-Ran Li
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China
| | - Qin Yang
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China
| | - Xiaochen Hu
- Department of Psychiatry, University of Cologne, Medical Faculty, Cologne, 50924, Germany
| | - Ying Han
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China;,Center of Alzheimer’s Disease, Beijing Institute for Brain Disorders, Beijing, 100053, China;,National Clinical Research Center for Geriatric Disorders, Beijing, 100053, China;,School of Biomedical Engineering, Hainan University, Haikou, 570228, China;,Address correspondence to this author at the Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China; Tel: +86 13621011941; E-mail:
| |
Collapse
|
40
|
Soluble Trem2 is a negative regulator of erythrophagocytosis after intracerebral hemorrhage in a CD36 receptor recycling manner. J Adv Res 2022; 44:185-199. [PMID: 36725189 PMCID: PMC9936424 DOI: 10.1016/j.jare.2022.03.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 02/23/2022] [Accepted: 03/15/2022] [Indexed: 02/04/2023] Open
Abstract
INTRODUCTION Microglia and macrophages participate in hematoma clearance after intracerebral hemorrhage (ICH), thereby facilitating tissue restoration and neurological recovery. Triggering receptor expressed on myeloid cells 2 (Trem2) has been indicated as a major pathology-induced immune signaling hub on the microglial/macrophage surface. Soluble Trem2 (sTrem2), the proteolytic form of Trem2, is abundant in the body fluid and is positively correlated with the pathological process. OBJECTIVES In the present study, we aimed to investigate the potential role of sTrem2 in hematoma resolution after ICH and to elucidate its underlying mechanisms. METHODS We explored the biological functions of sTrem2 in the murine ICH brain by stereotaxic injection of recombinant sTrem2 protein or by adeno-associated virus-mediated expression. Erythrocyte phagocytosis was assessed using flow cytometry and immunofluorescence. Western blotting was performed to evaluate protein expression. Changes in behavior, sTrem2-induced down-stream pathway, and microglia were examined. RESULTS sTrem2 impedes hematoma resolution and impairs functional motor and sensory recovery. Interestingly, sTrem2 bypasses full-length Trem2, negatively regulating microglial/macrophage erythrophagocytosis, and promotes an inflammatory phenotype, which is associated with reduced retromer levels and impaired recycling of the pro-erythrophagocytic receptor CD36. Rescue of retromer Vps35 abolishes the phagocytosis-inhibiting effects and lysosome-dependent CD36 degradation caused by sTrem2. CONCLUSION These findings indicate sTrem2 as a negative factor against microglia/macrophage-mediated hematoma and related neuronal damage clearance, provide insight into the mechanisms by which erythrophagocytosis is regulated and how it may be impaired after ICH, and suggest that the anti-proteolytic activity of Trem2 can be explored for ICH therapy.
Collapse
|
41
|
Zhang J, Sun B, Yang J, Chen Z, Li Z, Zhang N, Li H, Shen L. Comparison of the effect of rotenone and 1‑methyl‑4‑phenyl‑1,2,3,6‑tetrahydropyridine on inducing chronic Parkinson's disease in mouse models. Mol Med Rep 2022; 25:91. [PMID: 35039876 PMCID: PMC8809117 DOI: 10.3892/mmr.2022.12607] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 12/16/2021] [Indexed: 11/23/2022] Open
Abstract
Animal models for Parkinson's disease (PD) are very useful in understanding the pathogenesis of PD and screening for new therapeutic approaches. The present study compared two commonly used neurotoxin‑induced mouse models of chronic PD to guide model selection, explore the pathogenesis and mechanisms underlying PD and develop effective treatments. The chronic PD mouse models were established via treatment with rotenone or 1‑methyl‑4‑phenyl‑1,2,3,6-tetrahydropyridine (MPTP) for 6 weeks. The effects of rotenone and MPTP in the mice were compared by assessing neurobehavior, neuropathology and mitochondrial function through the use of the pole, rotarod and open field tests, immunohistochemistry for tyrosine hydroxylase (TH), glial fibrillary acidic protein (GFAP), ionized calcium‑binding adapter molecule 1 (Iba‑1), neuronal nuclear antigen (NeuN) and (p)S129 α‑synuclein, immunofluorescence for GFAP, Iba‑1 and NeuN, western blotting for TH, oxygen consumption, complex I enzyme activity. The locomotor activity, motor coordination and exploratory behavior in both rotenone and MPTP groups were significantly lower compared with the control group. However, behavioral tests were no significant differences between the two groups. In the MPTP group, the loss of dopaminergic (DA) neurons in the substantia nigra (SN) pars compacta, the reduction of the tyrosine hydroxylase content in the SN and striatum and the astrocyte proliferation and microglial activation in the SN were more significant compared with the rotenone group. Notably, mitochondrial‑dependent oxygen consumption and complex I enzyme activity in the SN were significantly reduced in the rotenone group compared with the MPTP group. In addition, Lewy bodies were present only in SN neurons in the rotenone group. Although no significant differences in neurobehavior were observed between the two mouse models, the MPTP model reproduced the pathological features of PD more precisely in terms of the loss of DA neurons, decreased dopamine levels and neuroinflammation in the SN. On the other hand, the rotenone model was more suitable for studying the role of mitochondrial dysfunction (deficient complex I activity) and Lewy body formation in the SN, which is a characteristic pathological feature of PD. The results indicated that MPTP and rotenone PD models have advantages and disadvantages, therefore one or both should be selected based on the purpose of the study.
Collapse
Affiliation(s)
- Jing Zhang
- Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Bohao Sun
- Department of Pathology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Jifeng Yang
- Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Zhuo Chen
- Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Zhengzheng Li
- Department of Internal Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Nan Zhang
- Department of Internal Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Hongzhi Li
- Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Luxi Shen
- Department of Internal Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| |
Collapse
|
42
|
Filipello F, Goldsbury C, Feng YS, Locca A, Karch CM, Piccio L. Soluble TREM2: Innocent bystander or active player in neurological diseases? Neurobiol Dis 2022; 165:105630. [PMID: 35041990 PMCID: PMC10108835 DOI: 10.1016/j.nbd.2022.105630] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 01/10/2022] [Accepted: 01/11/2022] [Indexed: 12/14/2022] Open
Abstract
Triggering receptor expressed on myeloid cells 2 (TREM2) is an innate immune receptor expressed by macrophages and microglia in the central nervous system (CNS). TREM2 has attracted a lot of interest in the past decade for its critical role in modulating microglia functions under homeostatic conditions and in neurodegenerative diseases. Genetic variation in TREM2 is sufficient to cause Nasu-Hakola disease, a rare pre-senile dementia with bone cysts, and to increase risk for Alzheimer's disease, frontotemporal dementia, and other neurodegenerative disorders. Beyond the role played by TREM2 genetic variants in these diseases, TREM2 engagement is a key step in microglia activation in response to different types of tissue injury (e.g. β-Amyloid deposition, demyelination, apoptotic cell death) leading to enhanced microglia metabolism, phagocytosis, proliferation and survival. TREM2 also exists as a soluble form (sTREM2), generated from receptor shedding or alternative splicing, which is detectable in plasma and cerebrospinal fluid (CSF). Genetic variation, physiological conditions and disease status impact CSF sTREM2 levels. Clinical and preclinical studies suggest that targeting and/or monitoring sTREM2 could have clinical and therapeutic implications. Despite the critical role of sTREM2 in neurologic disease, its function remains poorly understood. Here, we review the current literature on sTREM2 regarding its origin, genetic variation, and possible functions as a biomarker in neurological disorders and as a potential active player in CNS diseases and target for therapies.
Collapse
Affiliation(s)
- Fabia Filipello
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA; Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Claire Goldsbury
- Brain and Mind Centre and Charles Perkins Centre, School of Medical Sciences, University of Sydney, Sydney, NSW 2050, Australia
| | - You Shih Feng
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Alberto Locca
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Celeste M Karch
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Laura Piccio
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA; Brain and Mind Centre and Charles Perkins Centre, School of Medical Sciences, University of Sydney, Sydney, NSW 2050, Australia; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
43
|
Sheng X, Yao Y, Huang R, Xu Y, Zhu Y, Chen L, Zhang L, Wang W, Zhuo R, Can D, Chang CF, Zhang YW, Xu H, Bu G, Zhong L, Chen XF. Identification of the minimal active soluble TREM2 sequence for modulating microglial phenotypes and amyloid pathology. J Neuroinflammation 2021; 18:286. [PMID: 34893068 PMCID: PMC8665564 DOI: 10.1186/s12974-021-02340-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 12/02/2021] [Indexed: 11/24/2022] Open
Abstract
Background TREM2 is a microglial receptor genetically linked to the risk for Alzheimer’s disease (AD). The cerebrospinal fluid (CSF) levels of soluble TREM2 (sTREM2) have emerged as a valuable biomarker for the disease progression in AD and higher CSF levels of sTREM2 are linked to slower cognitive decline. Increasing sTREM2 in mouse models of amyloidosis reduces amyloid-related pathology through modulating microglial functions, suggesting a beneficial role of sTREM2 in microglia biology and AD pathology. Methods In the current study, we performed serial C- and N-terminal truncations of sTREM2 protein to define the minimal sequence requirement for sTREM2 function. We initially assessed the impacts of sTREM2 mutants on microglial functions by measuring cell viability and inflammatory responses. The binding of the sTREM2 mutants to oligomeric Aβ was determined by solid-phase protein binding assay and dot blot assay. We further evaluated the impacts of sTREM2 mutants on amyloid-related pathology by direct stereotaxic injection of sTREM2 proteins into the brain of 5xFAD mice. Results We found that both sTREM2 fragments 41–81 and 51–81 enhance cell viability and inflammatory responses in primary microglia. However, the fragment 51–81 exhibited impaired affinity to oligomeric Aβ. When administrated to the 5xFAD mice brain, the sTREM2 fragment 41–81, but not 51–81, increased the number of plaque-associated microglia and reduced the plaque deposition. Interestingly, the fragment 41–81 was more efficient than the physiological form of sTREM2 in ameliorating Aβ-related pathology. Conclusions Our results indicate that the interaction of sTREM2 truncated variants with Aβ is essential for enhancing microglial recruitment to the vicinity of an amyloid plaque and reducing the plaque load. Importantly, we identified a 41-amino acid sequence of sTREM2 that is sufficient for modulating microglial functions and more potent than the full-length sTREM2 in reducing the plaque load and the plaque-associated neurotoxicity. Taken together, our data provide more insights into the mechanisms underlying sTREM2 function and the minimal active sTREM2 sequence represents a promising candidate for AD therapy.
Collapse
Affiliation(s)
- Xuan Sheng
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Yunling Yao
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Ruizhi Huang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Ying Xu
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Yifei Zhu
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Linting Chen
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Lianshuai Zhang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Wanbing Wang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Rengong Zhuo
- Xiamen Key Laboratory of Chiral Drugs, School of Medicine, Xiamen University, Xiamen, 361102, China.,Shenzhen Research Institute of Xiamen University, Shenzhen, 518063, China
| | - Dan Can
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Che-Feng Chang
- Graduate Institute of Physiology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Yun-Wu Zhang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Huaxi Xu
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Li Zhong
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, China.
| | - Xiao-Fen Chen
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, China. .,Shenzhen Research Institute of Xiamen University, Shenzhen, 518063, China.
| |
Collapse
|
44
|
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: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [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
|
45
|
Mo M, Tang Y, Wei L, Qiu J, Peng G, Lin Y, Zhou M, Dai W, Zhang Z, Chen X, Liu H, Ding L, Ye P, Wu Y, Zhu X, Wu Z, Guo W, Xu P. Soluble Triggering Receptor Expressed on Myeloid Cells 2 From Cerebrospinal Fluid in Sleep Disorders Related to Parkinson's Disease. Front Aging Neurosci 2021; 13:753210. [PMID: 34658845 PMCID: PMC8511683 DOI: 10.3389/fnagi.2021.753210] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 09/07/2021] [Indexed: 01/04/2023] Open
Abstract
Background: Triggering receptor expressed on myeloid cells 2 (TREM2) is a microglial receptor exclusively expressed in the central nervous system (CNS). It contributes to abnormal protein aggregation in neurodegenerative disorders, but its role in Parkinson’s disease (PD) is still unclear. Methods: In this case-control study, we measured the concentration of the soluble fragment of TREM2 (sTREM2) in PD patients, evaluated their sleep conditions by the PD sleep scale (PDSS), and analyzed the relationship between sTREM2 and PD symptoms. Results: We recruited 80 sporadic PD patients and 65 healthy controls without disease-related variants in TREM2. The concentration of sTREM2 in the CSF was significantly higher in PD patients than in healthy controls (p < 0.01). In the PD group, the concentration of sTREM2 had a positive correlation with α-syn in the CSF (Pearson r = 0.248, p = 0.027). Receiver operating characteristic curve (ROC) analyses showed that sTREM2 in the CSF had a significant diagnostic value for PD (AUC, 0.791; 95% CI, 0.711–0.871, p < 0.05). The subgroup analysis showed that PD patients with sleep disorders had a significantly higher concentration of sTREM2 in their CSF (p < 0.01). The concentration of sTREM2 in the CSF had a negative correlation with the PDSS score in PD patients (Pearson r = −0.555, p < 0.01). The ROC analyses showed that sTREM2 in the CSF had a significant diagnostic value for sleep disorders in PD (AUC, 0.733; 95% CI, 0.619–0.846, p < 0.05). Conclusion: Our findings suggest that CSF sTREM2 may be a potential biomarker for PD and it could help predict sleep disorders in PD patients, but multicenter prospective studies with more participants are still needed to confirm its diagnostic value in future.
Collapse
Affiliation(s)
- Mingshu Mo
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yuting Tang
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Lijian Wei
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jiewen Qiu
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Guoyou Peng
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yuwan Lin
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Miaomiao Zhou
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wei Dai
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhiling Zhang
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiang Chen
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hanqun Liu
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Liuyan Ding
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Panghai Ye
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yijuan Wu
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaoqin Zhu
- Department of Physiology, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Zhuohua Wu
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wenyuan Guo
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Pingyi Xu
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
46
|
Bartl M, Dakna M, Galasko D, Hutten SJ, Foroud T, Quan M, Marek K, Siderowf A, Franz J, Trenkwalder C, Mollenhauer B. Biomarkers of neurodegeneration and glial activation validated in Alzheimer's disease assessed in longitudinal cerebrospinal fluid samples of Parkinson's disease. PLoS One 2021; 16:e0257372. [PMID: 34618817 PMCID: PMC8496858 DOI: 10.1371/journal.pone.0257372] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 08/29/2021] [Indexed: 12/25/2022] Open
Abstract
Aim Several pathophysiological processes are involved in Parkinson’s disease (PD) and could inform in vivo biomarkers. We assessed an established biomarker panel, validated in Alzheimer’s Disease, in a PD cohort. Methods Longitudinal cerebrospinal fluid (CSF) samples from PPMI (252 PD, 115 healthy controls, HC) were analyzed at six timepoints (baseline, 6, 12, 24, 36, and 48 months follow-up) using Elecsys® electrochemiluminescence immunoassays to quantify neurofilament light chain (NfL), soluble TREM2 receptor (sTREM2), chitinase-3-like protein 1 (YKL40), glial fibrillary acidic protein (GFAP), interleukin-6 (IL-6), S100, and total α-synuclein (αSyn). Results αSyn was significantly lower in PD (mean 103 pg/ml vs. HC: 127 pg/ml, p<0.01; area under the curve [AUC]: 0.64), while all other biomarkers were not significantly different (AUC NfL: 0.49, sTREM2: 0.54, YKL40: 0.57, GFAP: 0.55, IL-6: 0.53, S100: 0.54, p>0.05) and none showed a significant difference longitudinally. We found significantly higher levels of all these markers between PD patients who developed cognitive decline during follow-up, except for αSyn and IL-6. Conclusion Except for αSyn, the additional biomarkers did not differentiate PD and HC, and none showed longitudinal differences, but most markers predict cognitive decline in PD during follow-up.
Collapse
Affiliation(s)
- Michael Bartl
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
| | - Mohammed Dakna
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
| | - Douglas Galasko
- Department of Neurosciences, University of California, San Diego, San Diego, CA, United States of America
| | - Samantha J. Hutten
- The Michael J. Fox Foundation for Parkinson’s Research, New York, NY, United States of America
| | - Tatiana Foroud
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United States of America
| | - Marian Quan
- Roche Diagnostics, Indianapolis, IN, United States of America
| | - Kenneth Marek
- Institute for Neurodegenerative Disorders, New Haven, CT, United States of America
| | - Andrew Siderowf
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Jonas Franz
- Institute of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
- Campus Institute for Dynamics of Biological Networks, University of Göttingen, Göttingen, Germany
- Max Planck Institute for Experimental Medicine, Göttingen, Germany
| | - Claudia Trenkwalder
- Paracelsus-Elena-Klinik, Kassel, Germany
- Department of Neurosurgery, University Medical Center Goettingen, Goettingen, Germany
| | - Brit Mollenhauer
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
- Paracelsus-Elena-Klinik, Kassel, Germany
- * E-mail:
| | | |
Collapse
|
47
|
Tsai HH, Chen YF, Yen RF, Lo YL, Yang KC, Jeng JS, Tsai LK, Chang CF. Plasma soluble TREM2 is associated with white matter lesions independent of amyloid and tau. Brain 2021; 144:3371-3380. [PMID: 34515756 DOI: 10.1093/brain/awab332] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 07/21/2021] [Accepted: 08/08/2021] [Indexed: 12/20/2022] Open
Abstract
Cerebral small vessel disease is one of the most common causes of cognitive decline and stroke. While several lines of evidence have established a relationship between inflammation and cerebrovascular pathology, the mechanistic link has not yet been elucidated. Recent studies suggest activation of immune mediators, including the soluble form of triggering receptor expressed on myeloid cells 2 (TREM2), may be critical regulators. In this study, we compared the plasma levels of soluble TREM2 and its correlations with neuroimaging markers and cerebral amyloid load in ten patients with Alzheimer's disease and 66 survivors of spontaneous intracerebral haemorrhage with cerebral amyloid angiopathy or hypertensive small vessel disease, two of the most common types of sporadic small vessel disease. We performed brain MRI and 11C-Pittsburgh compound B PET for all participants to evaluate radiological small vessel disease markers and cerebral amyloid burden, and 18F-T807 PET in a subgroup of patients to evaluate cortical tau pathology. Plasma soluble TREM2 levels were comparable between patients with Alzheimer's disease and small vessel disease (P=0.690). In patients with small vessel disease, plasma soluble TREM2 was significantly associated with white matter hyperintensity volume (P<0.001), but not with cerebral amyloid load. Among patients with Alzheimer's disease and cerebral amyloid angiopathy, plasma soluble TREM2 was independently associated with a tau-positive scan (P=0.001) and white matter hyperintensity volume (P=0.013), but not amyloid load (P=0.221). Our results indicate plasma soluble TREM2 is associated with white matter hyperintensity independent of amyloid and tau pathology. These findings highlight the potential utility of plasma soluble TREM2 as a strong predictive marker for small vessel disease-related white matter injury and hold clinical implications for targeting the innate immune response when treating this disease.
Collapse
Affiliation(s)
- Hsin-Hsi Tsai
- Department of Neurology, National Taiwan University Hospital Beihu Branch, Taipei, Taiwan.,Department of Neurology, 3Medical Imaging, and 4Nuclear Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Ya-Fang Chen
- Department and Graduate Institute of Pharmacology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Ruoh-Fang Yen
- Department and Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yen-Ling Lo
- Department of Neurology, National Taiwan University Hospital Beihu Branch, Taipei, Taiwan
| | - Kai-Chien Yang
- Department and Graduate Institute of Pharmacology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Jiann-Shing Jeng
- Department of Neurology, 3Medical Imaging, and 4Nuclear Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Li-Kai Tsai
- Department of Neurology, 3Medical Imaging, and 4Nuclear Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Che-Feng Chang
- Department and Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan
| |
Collapse
|
48
|
Schulz I, Kruse N, Gera RG, Kremer T, Cedarbaum J, Barbour R, Zago W, Schade S, Otte B, Bartl M, Hutten SJ, Trenkwalder C, Mollenhauer B. Systematic Assessment of 10 Biomarker Candidates Focusing on α-Synuclein-Related Disorders. Mov Disord 2021; 36:2874-2887. [PMID: 34363416 DOI: 10.1002/mds.28738] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/30/2021] [Accepted: 07/14/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Objective diagnostic biomarkers are needed to support a clinical diagnosis. OBJECTIVES To analyze markers in various neurodegenerative disorders to identify diagnostic biomarker candidates for mainly α-synuclein (aSyn)-related disorders (ASRD) in serum and/or cerebrospinal fluid (CSF). METHODS Upon initial testing of commercially available kits or published protocols for the quantification of the candidate markers, assays for the following were selected: total and phosphorylated aSyn (pS129aSyn), neurofilament light chain (NfL), phosphorylated neurofilament heavy chain (pNfH), tau protein (tau), ubiquitin C-terminal hydrolase L1 (UCHL-1), glial fibrillary acidic protein (GFAP), calcium-binding protein B (S100B), soluble triggering receptor expressed on myeloid cells 2 (sTREM-2), and chitinase-3-like protein 1 (YKL-40). The cohort comprised participants with Parkinson's disease (PD, n = 151), multiple system atrophy (MSA, n = 17), dementia with Lewy bodies (DLB, n = 45), tau protein-related neurodegenerative disorders (n = 80, comprising patients with progressive supranuclear palsy (PSP, n = 38), corticobasal syndrome (CBS, n = 16), Alzheimer's disease (AD, n = 11), and frontotemporal degeneration/amyotrophic lateral sclerosis (FTD/ALS, n = 15), as well as healthy controls (HC, n = 20). Receiver operating curves (ROC) with area under the curves (AUC) are given for each marker. RESULTS CSF total aSyn was decreased. NfL, pNfH, UCHL-1, GFAP, S100B, and sTREM-2 were increased in patients with neurodegenerative disease versus HC (P < 0.05). As expected, some of the markers were highest in AD (i.e., UCHL-1, GFAP, S100B, sTREM-2, YKL-40). Within ASRD, CSF NfL levels were higher in MSA than PD and DLB (P < 0.05). Comparing PD to HC, interesting serum markers were S100B (AUC: 0.86), sTREM2 (AUC: 0.87), and NfL (AUC: 0.78). CSF S100B and serum GFAP were highest in DLB. CONCLUSIONS Levels of most marker candidates tested in serum and CSF significantly differed between disease groups and HC. In the stratification of PD versus other tau- or aSyn-related conditions, CSF NfL levels best discriminated PD and MSA. CSF S100B and serum GFAP best discriminated PD and DLB. © 2021 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson Movement Disorder Society.
Collapse
Affiliation(s)
| | - Niels Kruse
- Department of Neuropathology, University Medical Centre Goettingen, Goettingen, Germany
| | - Roland G Gera
- Department of Medical Statistics, University Medical Centre Goettingen, Goettingen, Germany
| | - Thomas Kremer
- Roche Pharmaceutical Research and Early Development, NRD Neuroscience and Rare Disease, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Jesse Cedarbaum
- Coeruleus Clinical Sciences LLC, Woodbidge, Connecticut, USA.,Yale University School of Medicine, New Haven, Connecticut, USA
| | - Robin Barbour
- Prothena Biosciences Inc., San Francisco, California, USA
| | - Wagner Zago
- Prothena Biosciences Inc., San Francisco, California, USA
| | - Sebastian Schade
- Department of Neurology, University Medical Centre Goettingen, Goettingen, Germany
| | - Birgit Otte
- Department of Neurology, University Medical Centre Goettingen, Goettingen, Germany
| | - Michael Bartl
- Department of Neurology, University Medical Centre Goettingen, Goettingen, Germany
| | - Samantha J Hutten
- The Michael J. Fox Foundation for Parkinson's Research, New York, New York, USA
| | - Claudia Trenkwalder
- Paracelsus-Elena-Klinik, Kassel, Germany.,Department of Neurosurgery, University Medical Centre Goettingen, Goettingen, Germany
| | - Brit Mollenhauer
- Paracelsus-Elena-Klinik, Kassel, Germany.,Department of Neurology, University Medical Centre Goettingen, Goettingen, Germany
| |
Collapse
|
49
|
Li D, Odessey R, Li D, Pacifici D. Plasmonic Interferometers as TREM2 Sensors for Alzheimer's Disease. BIOSENSORS 2021; 11:217. [PMID: 34356688 PMCID: PMC8301914 DOI: 10.3390/bios11070217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/25/2021] [Accepted: 06/26/2021] [Indexed: 11/17/2022]
Abstract
We report an effective surface immobilization protocol for capture of Triggering Receptor Expressed on Myeloid Cells 2 (TREM2), a receptor whose elevated concentration in cerebrospinal fluid has recently been associated with Alzheimer's disease (AD). We employ the proposed surface functionalization scheme to design, fabricate, and assess a biochemical sensing platform based on plasmonic interferometry that is able to detect physiological concentrations of TREM2 in solution. These findings open up opportunities for label-free biosensing of TREM2 in its soluble form in various bodily fluids as an early indicator of the onset of clinical dementia in AD. We also show that plasmonic interferometry can be a powerful tool to monitor and optimize surface immobilization schemes, which could be applied to develop other relevant antibody tests.
Collapse
Affiliation(s)
- Dingdong Li
- School of Engineering, Brown University, 184 Hope St, Providence, RI 02912, USA; (D.L.); (R.O.); (D.L.)
| | - Rachel Odessey
- School of Engineering, Brown University, 184 Hope St, Providence, RI 02912, USA; (D.L.); (R.O.); (D.L.)
| | - Dongfang Li
- School of Engineering, Brown University, 184 Hope St, Providence, RI 02912, USA; (D.L.); (R.O.); (D.L.)
| | - Domenico Pacifici
- School of Engineering, Brown University, 184 Hope St, Providence, RI 02912, USA; (D.L.); (R.O.); (D.L.)
- Department of Physics, Brown University, 182 Hope St, Providence, RI 02912, USA
| |
Collapse
|
50
|
Shafi S, Singh A, Ibrahim AM, Alhajri N, Abu Izneid T, Pottoo FH. Role of triggering receptor expressed on myeloid cells 2 (TREM2) in neurodegenerative dementias. Eur J Neurosci 2021; 53:3294-3310. [PMID: 33786894 DOI: 10.1111/ejn.15215] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 03/22/2021] [Indexed: 01/04/2023]
Abstract
Neurodegeneration is a debilitating condition that causes nerve cell degeneration or death. Neurodegenerative diseases (NDDs) such as Alzheimer's disease (AD), Parkinson's disease (PD), frontotemporal dementia (FTD), and Lewy body dementia (LBD) are posing a larger population burden of dementia worldwide. Neurodegenerative dementia is one of the main challenges in public health with its main characteristics being permanent loss of memory, impairment in cognition, and impaired daily functions. The published literature about genetic studies of these disorders suggests genetic underpinning in the pathogenesis of neurodegenerative dementia. In the process of underlining the pathogenesis of NDD, growing evidence has related genetic variations in the triggering receptor expressed on myeloid cells 2 (TREM2). This review paper aims to provide a detailed information regarding the association of TREM2 and NDDs leading to dementia. A central consideration is AD that accounts for almost 50%-70% of all late-life dementias alone or in combination with other neurological disorders. Other prevalent neurodegenerative conditions that lead to dementia are also discussed. Such studies are important as they can give a comprehensive knowledge of TREM2's role in various NDDs, in order to maximize the potential for developing new therapeutic approaches.
Collapse
Affiliation(s)
- Sadat Shafi
- Pharmaceutical Medicine, Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Archu Singh
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Abdallah Mohammad Ibrahim
- Fundamentals of Nursing Department, College of Nursing, Imam Abdul Rahman Bin Faisal University, Dammam, Saudi Arabia
| | - Noora Alhajri
- Department of Epidemiology and Population Health, College of Medicine and Health Science, Khalifa University, Abu Dhabi, UAE
| | | | - Faheem Hyder Pottoo
- Department of Pharmacology, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, Damman, Saudi Arabia
| |
Collapse
|