1
|
Yang B, Teymur A, Tang C, Wu T. V-set and immunoglobulin domain containing 4 as a potential predictor of Alzheimer's disease and advanced aging. J Alzheimers Dis 2025; 105:453-463. [PMID: 40116682 DOI: 10.1177/13872877251329463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2025]
Abstract
BackgroundV-set and immunoglobulin domain containing 4 (VSIG4) emerges as a significant player in the immune system pathways. It has been previously identified as a potential hub gene for Alzheimer's disease (AD) and aging, underscoring its importance in understanding these conditions.ObjectiveThis study aimed to evaluate the diagnostic potential of serum VSIG4 and identify trends in serum VSIG4 in relationship with other biomarkers and neurological tests.MethodsELISA was used to measure the serum concentration of VSIG4 in AD, compared to healthy subjects. The relationship between VSIG4 levels and the age of the subjects, as well as other AD-related serum proteins and various measures of cognition was examined.ResultsVSIG4 was significantly elevated in the serum of AD patients compared to healthy controls (p = 0.0074). Significant correlations were identified between serum VSIG4 and other notable proteins related to AD and inflammation, such as total tau, neurofilament light (NfL), YKL-40, CD14, FABP3, and TNF-α. Significant correlations were also identified between VSIG4 concentration and the results of neurological tests.ConclusionsSerum VSIG4 may reflect neuroinflammation and altered lipid processing, affecting the cognitive performance of AD and aging.
Collapse
Affiliation(s)
- Bowen Yang
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA
| | - Aygun Teymur
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA
| | - Chenling Tang
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA
| | - Tianfu Wu
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA
| |
Collapse
|
2
|
Cai H, Zhao T, Pang Y, Fu X, Ren Z, Quan S, Jia L. Systemic inflammatory markers in ageing, Alzheimer's disease and other dementias. Brain 2025; 148:480-492. [PMID: 39008616 DOI: 10.1093/brain/awae230] [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: 10/21/2023] [Revised: 05/28/2024] [Accepted: 06/23/2024] [Indexed: 07/17/2024] Open
Abstract
Systemic inflammation with alterations in inflammatory markers is involved in ageing and Alzheimer's disease. However, few studies have investigated the longitudinal trajectories of systemic inflammatory markers during ageing and Alzheimer's disease, and specific markers contributing to Alzheimer's disease remain undetermined. In this study, a longitudinal cohort (cohort 1: n = 290; controls, 136; preclinical Alzheimer's disease, 154) and a cross-sectional cohort (cohort 2: n = 351; controls, 62; Alzheimer's disease, 63; vascular dementia, 58; Parkinson's disease dementia, 56; behavioural variant frontotemporal dementia, 57; dementia with Lewy bodies, 55) were included. Plasma levels of inflammatory markers were measured every 2 years during a 10-year follow-up in the longitudinal cohort and once in the cross-sectional cohort. The study demonstrated that the inflammatory markers significantly altered during both ageing and the development of Alzheimer's disease. However, only complement C3, interleukin-1β and interleukin-6 exhibited significant changes in participants with preclinical Alzheimer's disease, and their longitudinal changes were significantly associated with the development of Alzheimer's disease compared to controls over the 10-year follow-up. In the cross-sectional cohort, complement C3 demonstrated specificity to Alzheimer's disease, while interleukin-1β and interleukin-6 were also altered in other dementias. The study provides a new perspective on the involvement of inflammatory markers in the ageing process and the development of Alzheimer's disease, implying that regulating inflammation may have a pivotal role in promoting successful ageing and in the prevention and treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Huimin Cai
- Innovation Centre for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Centre for Geriatric Diseases, Beijing 100053, China
| | - Tan Zhao
- Innovation Centre for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Centre for Geriatric Diseases, Beijing 100053, China
| | - Yana Pang
- Innovation Centre for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Centre for Geriatric Diseases, Beijing 100053, China
| | - Xiaofeng Fu
- Innovation Centre for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Centre for Geriatric Diseases, Beijing 100053, China
| | - Ziye Ren
- Innovation Centre for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Centre for Geriatric Diseases, Beijing 100053, China
| | - Shuiyue Quan
- Innovation Centre for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Centre for Geriatric Diseases, Beijing 100053, China
| | - Longfei Jia
- Innovation Centre for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Centre for Geriatric Diseases, Beijing 100053, China
| |
Collapse
|
3
|
Zhao B, Zang P, Quan M, Wang Q, Guo D, Jia J, Wang W. The Effect of APOE ε4 on Alzheimer's Disease Fluid Biomarkers: A Cross-Sectional Study Based on the COAST. CNS Neurosci Ther 2025; 31:e70202. [PMID: 39749650 PMCID: PMC11696244 DOI: 10.1111/cns.70202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 12/10/2024] [Accepted: 12/18/2024] [Indexed: 01/04/2025] Open
Abstract
AIMS To analyze the effect of APOE ε4 on fluid biomarkers and the correlations between blood molecules and CSF biomarkers in AD patients. METHODS This study enrolled 575 AD patients, 131 patients with non-AD dementia, and 112 cognitively normal (CN) participants, and AD patients were divided into APOE ε4 carriers and non-carriers. Cerebrospinal fluid (CSF) biomarkers and blood-derived biomolecules were compared between AD and CN groups, between non-AD dementia and CN groups, as well as within APOE ε4 subgroups of AD patients. Utilizing Spearman's correlation analysis and quantile regression analysis, the relationships between blood-derived biomolecules and CSF biomarkers were analyzed in APOE ε4 carriers and non-carriers. RESULTS The levels of CSF biomarkers and blood molecules exhibited significant differences between the AD and CN groups, including Aβ42, t-tau, p-tau 181, high-density lipoprotein, low-density lipoprotein (LDL), and uric acid. In AD patients, APOE ε4 carriers had increased levels of CSF t-tau, p-tau 181, and plasma LDL. In the correlation and regression analyses, the negative relationships between plasma TG and t-tau, between plasma TG and p-tau 181 levels, as well as the positive relationship between serum IgA and CSF Aβ42, were observed significantly in APOE ε4+ AD groups, but not in APOE ε4- AD group. CONCLUSION APOE ε4 is associated with accelerated progression of AD pathology. The blood-derived biomolecules correlated with CSF biomarkers in APOE ε4 carriers are related to neuroinflammation and lipid metabolism, which may indicate the role of APOE ε4 in AD pathophysiology and offer insights for diagnostic and therapeutic strategies for AD. TRIAL REGISTRATION ClinicalTrials.gov identifier: NCT03653156.
Collapse
Affiliation(s)
- Bote Zhao
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu HospitalCapital Medical UniversityBeijingChina
- National Center for Neurological Disorders and National Clinical Research Center for Geriatric DiseasesBeijingChina
- Center of Alzheimer's Disease, Beijing Institute of Brain DisordersCollaborative Innovation Center for Brain DisordersBeijingChina
| | - Peixi Zang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu HospitalCapital Medical UniversityBeijingChina
- Department of NeurologyGansu Provincial HospitalLanzhou CityGansu ProvinceChina
| | - Meina Quan
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu HospitalCapital Medical UniversityBeijingChina
- National Center for Neurological Disorders and National Clinical Research Center for Geriatric DiseasesBeijingChina
- Center of Alzheimer's Disease, Beijing Institute of Brain DisordersCollaborative Innovation Center for Brain DisordersBeijingChina
| | - Qianqian Wang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu HospitalCapital Medical UniversityBeijingChina
- National Center for Neurological Disorders and National Clinical Research Center for Geriatric DiseasesBeijingChina
- Center of Alzheimer's Disease, Beijing Institute of Brain DisordersCollaborative Innovation Center for Brain DisordersBeijingChina
| | - Dongmei Guo
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu HospitalCapital Medical UniversityBeijingChina
- National Center for Neurological Disorders and National Clinical Research Center for Geriatric DiseasesBeijingChina
- Center of Alzheimer's Disease, Beijing Institute of Brain DisordersCollaborative Innovation Center for Brain DisordersBeijingChina
| | - Jianping Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu HospitalCapital Medical UniversityBeijingChina
- National Center for Neurological Disorders and National Clinical Research Center for Geriatric DiseasesBeijingChina
- Center of Alzheimer's Disease, Beijing Institute of Brain DisordersCollaborative Innovation Center for Brain DisordersBeijingChina
| | - Wei Wang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu HospitalCapital Medical UniversityBeijingChina
- National Center for Neurological Disorders and National Clinical Research Center for Geriatric DiseasesBeijingChina
- Center of Alzheimer's Disease, Beijing Institute of Brain DisordersCollaborative Innovation Center for Brain DisordersBeijingChina
| |
Collapse
|
4
|
Daskoulidou N, Carpanini SM, Zelek WM, Morgan BP. Involvement of Complement in Alzheimer's Disease: From Genetics Through Pathology to Therapeutic Strategies. Curr Top Behav Neurosci 2025; 69:3-24. [PMID: 39455500 DOI: 10.1007/7854_2024_524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2024]
Abstract
Complement is a critical component of innate immunity, evolved to defend against pathogens and clear toxic debris ranging from dead and dying cells to immune complexes. These roles make complement a key player in homeostasis; however, complement has a dark side. When the rigid control mechanisms fail, complement becomes dysregulated, acting as a driver of inflammation and resultant pathology in numerous diseases. Roles of complement in Alzheimer's disease (AD) and other dementias have emerged in recent years, supported by genetic, biomarker and pathological evidence and animal model studies. Numerous questions remain regarding the precise roles of complement in the brain in health and disease, including where and when complement is expressed, how it contributes to immune defence and garbage disposal in the healthy brain, and exactly how complement contributes to pathology in dementias. In this brief review, we will summarise current knowledge on complement roles in brain, present the evidence implicating complement in AD and explore whether complement represents an attractive therapeutic target for AD.
Collapse
Affiliation(s)
| | - Sarah M Carpanini
- UK Dementia Research Institute Cardiff, Cardiff University, Cardiff, UK
| | - Wioleta M Zelek
- UK Dementia Research Institute Cardiff, Cardiff University, Cardiff, UK
| | - B Paul Morgan
- UK Dementia Research Institute Cardiff, Cardiff University, Cardiff, UK.
| |
Collapse
|
5
|
Krishnan A, Pathak A, Nicholas TB, Lee J, Waite L, Stanaway F. Racial and ethnic minority representation in dementia risk factor research: a scoping review of cohort studies. BMJ Open 2024; 14:e085592. [PMID: 39322589 PMCID: PMC11733783 DOI: 10.1136/bmjopen-2024-085592] [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: 02/20/2024] [Accepted: 08/29/2024] [Indexed: 09/27/2024] Open
Abstract
BACKGROUND Despite a potentially greater burden of dementia, racial and ethnic minority populations around the world may be more likely to be excluded from research examining risk factors for incident dementia. We aimed to systematically investigate and quantify racial and ethnic minority representation in dementia risk factor research. METHODS We performed a two-stage systematic search of databases-MEDLINE (Ovid SP), Embase (Ovid SP) and Scopus-from inception to March 2021 to identify population-based cohort studies looking at risk factors for dementia incidence. We included cohort studies which were population-based and incorporated a clinical dementia diagnosis. RESULTS Out of the 97 identified cohort studies, fewer than half (40 studies; 41%) reported the race or ethnicity of participants and just under one-third (29 studies; 30%) reported the inclusion of racial and ethnic minority groups. We found that inadequate reporting frequently prevented assessment of selection bias and only six studies that included racial and ethnic minority participants were at low risk for measurement bias in dementia diagnosis. In cohort studies including a multiethnic cohort, only 182 out of 337 publications incorporated race or ethnicity in data analysis-predominantly (90%) through adjustment for race or ethnicity as a confounder. Only 14 publications (4.2% of all publications reviewed) provided evidence about drivers of any observed inequalities. CONCLUSIONS Racial and ethnic minority representation in dementia risk factor research is inadequate. Comparisons of dementia risk between different racial and ethnic groups are likely hampered by significant selection and measurement bias. Moreover, the focus on 'adjusting out' the effect of race and ethnicity as a confounder prevents understanding of underlying drivers of observed inequalities. There is a pressing need to fundamentally change the way race, ethnicity and the inclusion of racial and ethnic minorities are considered in research if health inequalities are to be adequately addressed.
Collapse
Affiliation(s)
- Arjun Krishnan
- School of Public Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Anupa Pathak
- School of Public Health, The University of Sydney, Sydney, New South Wales, Australia
| | | | - Jeffrey Lee
- School of Public Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Louise Waite
- Faculty of Medicine and Health, The University of Sydney Concord Clinical School, Sydney, New South Wales, Australia
- Centre for Education and Research on Ageing, The University of Sydney, Sydney, New South Wales, Australia
| | - Fiona Stanaway
- School of Public Health, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
6
|
Uchida K, Meno K, Korenaga T, Liu S, Suzuki H, Baba Y, Tagata C, Araki Y, Tsunemi S, Aso K, Inagaki S, Nakagawa S, Kobayashi M, Kakuma T, Asada T, Ota M, Takihara T, Arai T. Effect of matcha green tea on cognitive functions and sleep quality in older adults with cognitive decline: A randomized controlled study over 12 months. PLoS One 2024; 19:e0309287. [PMID: 39213264 PMCID: PMC11364242 DOI: 10.1371/journal.pone.0309287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024] Open
Abstract
OBJECTIVE Lifestyle habits after middle age significantly impact the maintenance of cognitive function in older adults. Nutritional intake is closely related to lifestyle habits; therefore, nutrition is a pivotal factor in the prevention of dementia in the preclinical stages. Matcha green tea powder (matcha), which contains epigallocatechin gallate, theanine, and caffeine, has beneficial effects on cognitive function and mood. We conducted a randomized, double-blind, placebo-controlled clinical study over 12 months to examine the effect of matcha on cognitive function and sleep quality. METHODS Ninety-nine participants, including 64 with subjective cognitive decline and 35 with mild cognitive impairment were randomized, with 49 receiving 2 g of matcha and 50 receiving a placebo daily. Participants were stratified based on two factors: age at baseline and APOE genotype. Changes in cognitive function and sleep quality were analyzed using a mixed-effects model. RESULTS Matcha consumption led to significant improvements in social acuity score (difference; -1.39, 95% confidence interval; -2.78, 0.002) (P = 0.028) as evaluated by the perception of facial emotions in cognitive function. The primary outcomes, that is, Montreal Cognitive Assessment and Alzheimer's Disease Cooperative Study Activity of Daily Living scores, showed no significant changes with matcha intervention. Meanwhile, Pittsburgh Sleep Quality Index scores indicated a trend toward improvement with a difference of 0.86 (95% confidence interval; -0.002, 1.71) (P = 0.088) between the groups in changes from baseline to 12 months. CONCLUSIONS The present study suggests regular consumption of matcha could improve emotional perception and sleep quality in older adults with mild cognitive decline. Given the widespread availability and cultural acceptance of matcha green tea, incorporating it into the daily routine may offer a simple yet effective strategy for cognitive enhancement and dementia prevention.
Collapse
Affiliation(s)
- Kazuhiko Uchida
- Institute of Biomedical Research, MCBI Inc., Tsukuba, Ibaraki, Japan
- Research Division, MCBI Inc., Tsukuba, Ibaraki, Japan
| | - Kohji Meno
- Research Division, MCBI Inc., Tsukuba, Ibaraki, Japan
| | | | - Shan Liu
- Research Division, MCBI Inc., Tsukuba, Ibaraki, Japan
| | | | - Yoshitake Baba
- Central Research Institute, ITO EN, LTD., Makinohara, Shizuoka, Japan
| | - Chika Tagata
- Central Research Institute, ITO EN, LTD., Makinohara, Shizuoka, Japan
| | - Yoshiharu Araki
- Central Research Institute, ITO EN, LTD., Makinohara, Shizuoka, Japan
| | - Shuto Tsunemi
- Central Research Institute, ITO EN, LTD., Makinohara, Shizuoka, Japan
| | - Kenta Aso
- Central Research Institute, ITO EN, LTD., Makinohara, Shizuoka, Japan
| | - Shun Inagaki
- Central Research Institute, ITO EN, LTD., Makinohara, Shizuoka, Japan
| | - Sae Nakagawa
- Central Research Institute, ITO EN, LTD., Makinohara, Shizuoka, Japan
| | - Makoto Kobayashi
- Central Research Institute, ITO EN, LTD., Makinohara, Shizuoka, Japan
| | - Tatsuyuki Kakuma
- Biostatistics Center, Kurume University Graduate School of Medicine, Kurume, Japan
| | - Takashi Asada
- Memory Clinic Toride, Toride, Ibaraki, Japan
- Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Miho Ota
- Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Takanobu Takihara
- Central Research Institute, ITO EN, LTD., Makinohara, Shizuoka, Japan
| | - Tetsuaki Arai
- Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| |
Collapse
|
7
|
Rezaee-Zavareh MS, Kim N, Yang JD. Starting the journey: Understanding the roles of complement proteins in liver diseases through mendelian randomization. Clin Mol Hepatol 2024; 30:150-153. [PMID: 38385196 PMCID: PMC11016470 DOI: 10.3350/cmh.2024.0116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 02/19/2024] [Accepted: 02/20/2024] [Indexed: 02/23/2024] Open
Affiliation(s)
| | - Naomy Kim
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ju Dong Yang
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
8
|
Nimmo J, Byrne R, Daskoulidou N, Watkins L, Carpanini S, Zelek W, Morgan B. The complement system in neurodegenerative diseases. Clin Sci (Lond) 2024; 138:387-412. [PMID: 38505993 PMCID: PMC10958133 DOI: 10.1042/cs20230513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 02/15/2024] [Accepted: 03/01/2024] [Indexed: 03/21/2024]
Abstract
Complement is an important component of innate immune defence against pathogens and crucial for efficient immune complex disposal. These core protective activities are dependent in large part on properly regulated complement-mediated inflammation. Dysregulated complement activation, often driven by persistence of activating triggers, is a cause of pathological inflammation in numerous diseases, including neurological diseases. Increasingly, this has become apparent not only in well-recognized neuroinflammatory diseases like multiple sclerosis but also in neurodegenerative and neuropsychiatric diseases where inflammation was previously either ignored or dismissed as a secondary event. There is now a large and rapidly growing body of evidence implicating complement in neurological diseases that cannot be comprehensively addressed in a brief review. Here, we will focus on neurodegenerative diseases, including not only the 'classical' neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease, but also two other neurological diseases where neurodegeneration is a neglected feature and complement is implicated, namely, schizophrenia, a neurodevelopmental disorder with many mechanistic features of neurodegeneration, and multiple sclerosis, a demyelinating disorder where neurodegeneration is a major cause of progressive decline. We will discuss the evidence implicating complement as a driver of pathology in these diverse diseases and address briefly the potential and pitfalls of anti-complement drug therapy for neurodegenerative diseases.
Collapse
Affiliation(s)
- Jacqui Nimmo
- UK Dementia Research Institute Cardiff, Cardiff University, Cardiff CF24 4HQ, U.K
| | - Robert A.J. Byrne
- UK Dementia Research Institute Cardiff, Cardiff University, Cardiff CF24 4HQ, U.K
| | - Nikoleta Daskoulidou
- UK Dementia Research Institute Cardiff, Cardiff University, Cardiff CF24 4HQ, U.K
| | - Lewis M. Watkins
- UK Dementia Research Institute Cardiff, Cardiff University, Cardiff CF24 4HQ, U.K
| | - Sarah M. Carpanini
- UK Dementia Research Institute Cardiff, Cardiff University, Cardiff CF24 4HQ, U.K
| | - Wioleta M. Zelek
- UK Dementia Research Institute Cardiff, Cardiff University, Cardiff CF24 4HQ, U.K
| | - B. Paul Morgan
- UK Dementia Research Institute Cardiff, Cardiff University, Cardiff CF24 4HQ, U.K
| |
Collapse
|
9
|
You Y, Chen Z, Hu WW. The role of microglia heterogeneity in synaptic plasticity and brain disorders: Will sequencing shed light on the discovery of new therapeutic targets? Pharmacol Ther 2024; 255:108606. [PMID: 38346477 DOI: 10.1016/j.pharmthera.2024.108606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/05/2024] [Accepted: 02/02/2024] [Indexed: 02/18/2024]
Abstract
Microglia play a crucial role in interacting with neuronal synapses and modulating synaptic plasticity. This function is particularly significant during postnatal development, as microglia are responsible for removing excessive synapses to prevent neurodevelopmental deficits. Dysregulation of microglial synaptic function has been well-documented in various pathological conditions, notably Alzheimer's disease and multiple sclerosis. The recent application of RNA sequencing has provided a powerful and unbiased means to decipher spatial and temporal microglial heterogeneity. By identifying microglia with varying gene expression profiles, researchers have defined multiple subgroups of microglia associated with specific pathological states, including disease-associated microglia, interferon-responsive microglia, proliferating microglia, and inflamed microglia in multiple sclerosis, among others. However, the functional roles of these distinct subgroups remain inadequately characterized. This review aims to refine our current understanding of the potential roles of heterogeneous microglia in regulating synaptic plasticity and their implications for various brain disorders, drawing from recent sequencing research and functional studies. This knowledge may aid in the identification of pathogenetic biomarkers and potential factors contributing to pathogenesis, shedding new light on the discovery of novel drug targets. The field of sequencing-based data mining is evolving toward a multi-omics approach. With advances in viral tools for precise microglial regulation and the development of brain organoid models, we are poised to elucidate the functional roles of microglial subgroups detected through sequencing analysis, ultimately identifying valuable therapeutic targets.
Collapse
Affiliation(s)
- Yi You
- Department of Pharmacology and Department of Pharmacy of the Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Zhong Chen
- Department of Pharmacology and Department of Pharmacy of the Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China; Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Wei-Wei Hu
- Department of Pharmacology and Department of Pharmacy of the Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China.
| |
Collapse
|
10
|
Li M, Ma YH, Guo Y, Liu JY, Tan L. Associations of cerebrospinal fluid complement proteins with Alzheimer's pathology, cognition, and brain structure in non-dementia elderly. Alzheimers Res Ther 2024; 16:12. [PMID: 38238858 PMCID: PMC10795368 DOI: 10.1186/s13195-023-01377-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 12/26/2023] [Indexed: 01/22/2024]
Abstract
BACKGROUND Cerebrospinal fluid (CSF) complement activation is a key part of neuroinflammation that occurs in the early stages of Alzheimer's disease (AD). However, the associations of CSF complement proteins with AD pathology, cognition, and structural neuroimaging biomarkers for AD have been rarely investigated. METHODS A total of 210 participants (125 mild cognitive impairment [MCI] patients and 85 normal controls) were included from Alzheimer's Disease Neuroimaging Initiative (ADNI) database who measured AD pathology, cognition, and neuroimaging at baseline and every 12 months. The mixed-effect linear models were utilized to investigate longitudinal associations of CSF complement proteins with AD pathology, cognition, and neuroimaging in cognitively normal (CN) and mild cognitive impairment (MCI) subjects. Causal mediation analyses were conducted to explore the potential mediators between CSF complement proteins and cognitive changes. RESULTS We found that the subjects with low CSF complement protein levels at baseline had worse outcomes in AD pathology, indicated by their lowest concentrations observed in A + and A + T + individuals. The reduced CSF complement proteins were associated with faster accumulation of tau among CN subjects and with cognitive decline and greater brain atrophy of specific regions among MCI subjects. Furthermore, mediation analyses showed that the effects of CSF complement proteins on cognitive performance were partially mediated by regional brain structures (mediation proportions range from 19.78 to 94.92%; p < 0.05). CONCLUSIONS This study demonstrated that CSF complement proteins were involved in the early progression of AD. Our results indicated that regional brain atrophy might be a plausible way to connect CSF complement protein levels and cognition.
Collapse
Affiliation(s)
- Meng Li
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Ya-Hui Ma
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, 266071, China
| | - Yun Guo
- School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Jia-Yao Liu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China.
| |
Collapse
|
11
|
Inoue M, Suzuki H, Meno K, Liu S, Korenaga T, Uchida K. Identification of Plasma Proteins as Biomarkers for Mild Cognitive Impairment and Alzheimer's Disease Using Liquid Chromatography-Tandem Mass Spectrometry. Int J Mol Sci 2023; 24:13064. [PMID: 37685872 PMCID: PMC10488247 DOI: 10.3390/ijms241713064] [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: 07/06/2023] [Revised: 08/11/2023] [Accepted: 08/19/2023] [Indexed: 09/10/2023] Open
Abstract
Blood proteins can be used for biomarkers to monitor the progression of cognitive decline, even in the early stages of disease. In this study, we developed a liquid chromatography-tandem mass spectrometry (LC-MS/MS)-based blood test to identify plasma proteins that can be used to detect mild cognitive impairment (MCI) and Alzheimer's disease (AD). Using this system, we quantified plasma proteins using isotope-labeled synthetic peptides. A total of 192 patients, including 63 with AD, 71 with MCI, and 58 non-demented controls (NDCs), were analyzed. Multinomial regression and receiver operating characteristic (ROC) analyses were performed to identify specific combinations of plasma protein panels that could differentiate among NDCs, those with MCI, and those with AD. We identified eight plasma protein biomarker candidates that can be used to distinguish between MCI and AD. These biomarkers were associated with coagulation pathways, innate immunity, lipid metabolism, and nutrition. The clinical potential to differentiate cognitive impairment from NDC was assessed using area under the curve values from ROC analysis, which yielded values of 0.83 for males and 0.71 for females. This LC-MS-based plasma protein panel allows the pathophysiology of AD to be followed through detection of cognitive decline and disease progression markers.
Collapse
Affiliation(s)
- Makoto Inoue
- Research Division, MCBI, 5-4-2 Toukoudai, Tsukuba 300-2635, Ibaraki, Japan (H.S.)
| | - Hideaki Suzuki
- Research Division, MCBI, 5-4-2 Toukoudai, Tsukuba 300-2635, Ibaraki, Japan (H.S.)
| | - Kohji Meno
- Research Division, MCBI, 5-4-2 Toukoudai, Tsukuba 300-2635, Ibaraki, Japan (H.S.)
| | - Shan Liu
- Research Division, MCBI, 5-4-2 Toukoudai, Tsukuba 300-2635, Ibaraki, Japan (H.S.)
| | - Tatsumi Korenaga
- Research Division, MCBI, 5-4-2 Toukoudai, Tsukuba 300-2635, Ibaraki, Japan (H.S.)
| | - Kazuhiko Uchida
- Clinical Bioinformatics Initiative, Institute for Biomedical Research, MCBI, 5-4-2 Toukoudai, Tsukuba 300-2635, Ibaraki, Japan
| |
Collapse
|
12
|
Veteleanu A, Pape S, Davies K, Kodosaki E, Hye A, Zelek WM, Strydom A, Morgan BP. Complement dysregulation and Alzheimer's disease in Down syndrome. Alzheimers Dement 2023; 19:1383-1392. [PMID: 36149090 PMCID: PMC10798358 DOI: 10.1002/alz.12799] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 08/03/2022] [Accepted: 08/24/2022] [Indexed: 11/12/2022]
Abstract
INTRODUCTION Down syndrome (DS) is associated with immune dysregulation and a high risk of early onset Alzheimer's disease (AD). Complement is a key part of innate immunity and driver of pathological inflammation, including neuroinflammation in AD. Complement dysregulation has been reported in DS; however, the pattern of dysregulation and its relationship to AD risk is unclear. METHODS Plasma levels of 14 complement biomarkers were measured in 71 adults with DS and 46 controls to identify DS-associated dysregulation; impact of apolipoprotein E (APOE) ε4 genotype, single nucleotide polymorphisms (SNPs) in CLU and CR1, and dementia on complement biomarkers was assessed. RESULTS Plasma levels of complement activation products (TCC, iC3b), proteins (C1q, C3, C9), and regulators (C1 inhibitor, factor H, FHR4, clusterin) were significantly elevated in DS versus controls while FI and sCR1 were significantly lower. In DS with AD (n = 13), C3 and FI were significantly decreased compared to non-AD DS (n = 58). Neither APOE genotype nor CLU SNPs impacted complement levels, while rs6656401 in CR1 significantly impacted plasma sCR1 levels. CONCLUSIONS Complement is dysregulated in DS, likely reflecting the generalized immune dysregulation state; measurement may help identify inflammatory events in individuals with DS. Complement biomarkers differed in DS with and without AD and may aid diagnosis and/or prediction. HIGHLIGHTS Complement is significantly dysregulated in plasma of people with DS who show changes in levels of multiple complement proteins compared to controls. People with DS and dementia show evidence of additional complement dysregulation with significantly lower levels of C3 and factor I compared to those without dementia. rs6656401 in CR1 was associated with significantly elevated sCR1 plasma levels in DS.
Collapse
Affiliation(s)
- Aurora Veteleanu
- School of MedicineUK Dementia Research InstituteCardiff UniversityCardiffUK
| | - Sarah Pape
- Department of Forensic and Neurodevelopmental ScienceInstitute of PsychiatryPsychology and NeuroscienceKing's CollegeLondonUK
| | - Kate Davies
- School of Medicine, Division of Infection and ImmunityCardiff UniversityCardiffUK
| | - Eleftheria Kodosaki
- School of Medicine, Division of Infection and ImmunityCardiff UniversityCardiffUK
| | - Abdul Hye
- Department of Forensic and Neurodevelopmental ScienceInstitute of PsychiatryPsychology and NeuroscienceKing's CollegeLondonUK
| | - Wioleta M. Zelek
- School of MedicineUK Dementia Research InstituteCardiff UniversityCardiffUK
- School of Medicine, Division of Infection and ImmunityCardiff UniversityCardiffUK
| | - Andre Strydom
- Department of Forensic and Neurodevelopmental ScienceInstitute of PsychiatryPsychology and NeuroscienceKing's CollegeLondonUK
| | - B. Paul Morgan
- School of MedicineUK Dementia Research InstituteCardiff UniversityCardiffUK
| |
Collapse
|
13
|
Burgelman M, Dujardin P, Vandendriessche C, Vandenbroucke RE. Free complement and complement containing extracellular vesicles as potential biomarkers for neuroinflammatory and neurodegenerative disorders. Front Immunol 2023; 13:1055050. [PMID: 36741417 PMCID: PMC9896008 DOI: 10.3389/fimmu.2022.1055050] [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: 09/27/2022] [Accepted: 12/07/2022] [Indexed: 01/21/2023] Open
Abstract
The complement system is implicated in a broad range of neuroinflammatory disorders such as Alzheimer's disease (AD) and multiple sclerosis (MS). Consequently, measuring complement levels in biofluids could serve as a potential biomarker for these diseases. Indeed, complement levels are shown to be altered in patients compared to controls, and some studies reported a correlation between the level of free complement in biofluids and disease progression, severity or the response to therapeutics. Overall, they are not (yet) suitable as a diagnostic tool due to heterogeneity of reported results. Moreover, measurement of free complement proteins has the disadvantage that information on their origin is lost, which might be of value in a multi-parameter approach for disease prediction and stratification. In light of this, extracellular vesicles (EVs) could provide a platform to improve the diagnostic power of complement proteins. EVs are nanosized double membrane particles that are secreted by essentially every cell type and resemble the (status of the) cell of origin. Interestingly, EVs can contain complement proteins, while the cellular origin can still be determined by the presence of EV surface markers. In this review, we summarize the current knowledge and future opportunities on the use of free and EV-associated complement proteins as biomarkers for neuroinflammatory and neurodegenerative disorders.
Collapse
Affiliation(s)
- Marlies Burgelman
- VIB Center for Inflammation Research, VIB, Ghent, Belgium,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Pieter Dujardin
- VIB Center for Inflammation Research, VIB, Ghent, Belgium,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Charysse Vandendriessche
- VIB Center for Inflammation Research, VIB, Ghent, Belgium,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Roosmarijn E. Vandenbroucke
- VIB Center for Inflammation Research, VIB, Ghent, Belgium,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium,*Correspondence: Roosmarijn E. Vandenbroucke,
| |
Collapse
|
14
|
Rasmussen KL, Tybjærg-Hansen A, Nordestgaard BG, Frikke-Schmidt R. Associations of Alzheimer Disease-Protective APOE Variants With Age-Related Macular Degeneration. JAMA Ophthalmol 2023; 141:13-21. [PMID: 36394841 PMCID: PMC9673029 DOI: 10.1001/jamaophthalmol.2022.4602] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 09/26/2022] [Indexed: 11/18/2022]
Abstract
Importance The association of major lipid genes with and their potential as drug targets for age-related macular degeneration (AMD) is unknown. These associations are important to study because AMD is the leading cause of irreversible late-onset blindness in high-income countries. Objective To determine whether the full range of structural genetic variation in apolipoprotein E (APOE), a master gene in peripheral and cerebral lipid metabolism, is associated with risk of AMD. Design, Setting, and Participants This cohort study used data from the Copenhagen City Heart Study (CCHS) and the Copenhagen General Population Study (CGPS) cohorts. Participants were followed from study inclusion at the time of blood sampling to occurrence of event, death, emigration, or December 7, 2018, whichever came first. For participants in CCHS, the APOE gene was sequenced, and 9 variants with a heterozygote frequency of at least 0.0002 were genotyped in the CGPS. Observers were masked to patient groupings. Data were analyzed from March to September 2021. Exposures The exposure was APOE status, and the direct gene product in plasma, apoE levels, was measured in all participants. Main Outcomes and Measures Cox regression was applied to estimate risk of AMD associated with APOE genotype. Results A total of 105 546 participants (mean [SD] age, 57.7 [13.4] years; 58 140 [55%] female participants) were included. Compared with participants with the common ɛ33 genotype, risk of AMD was lower in participants with ε44 (multifactorially adjusted hazard ratio [aHR], 0.66; 95% CI, 0.45-0.96) and ε43 (aHR, 0.80; 95% CI, 0.71-0.90) genotypes and higher in the ε32 (aHR, 1.15; 95% CI, 1.00-1.31) genotype. Compared with noncarriers, risk of AMD was higher for participants with Gly145Asp (aHR, 3.53; 95% CI, 1.14-10.96) and Arg154Cys (aHR, 4.52; 95% CI, 1-13-18.13) heterozygotes. Results were similar after further adjustment for lipid traits and after adjustment for the APOE ε2/ε3/ε4 variant. Combining all common and rare structural variants in a weighted allele score, risk of AMD per 1-mg/dL genetically higher plasma apoE was increased in the adjusted model (aHR, 1.12; 95% CI, 1.05-1.19), the adjusted model plus APOE ɛ2/ɛ3/ɛ4 status (aHR, 1.82; 95% CI, 1.20-2.76), and the adjusted model in individuals with the ε33 genotype only (aHR, 1.77; 95% CI, 1.14-2.75). Conclusions and Relevance These findings highlight that structural variation in APOE beyond the ε2/ε3/ε4 variants may be important for risk of AMD in a population of European ancestry. Rare functional ɛ2-like variants in APOE have previously been reported to have protective associations for Alzheimer disease but the present findings suggest a simultaneous high risk of AMD. This would limit the drug target potential of mechanisms resembling these variants.
Collapse
Affiliation(s)
- Katrine L. Rasmussen
- Department of Clinical Biochemistry, Copenhagen University Hospital–Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- The Copenhagen General Population Study, Herlev and Gentofte Hospital, Herlev, Denmark
| | - Anne Tybjærg-Hansen
- Department of Clinical Biochemistry, Copenhagen University Hospital–Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- The Copenhagen General Population Study, Herlev and Gentofte Hospital, Herlev, Denmark
- The Copenhagen City Heart Study, Frederiksberg Hospital, Frederiksberg, Denmark
| | - Børge G. Nordestgaard
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- The Copenhagen General Population Study, Herlev and Gentofte Hospital, Herlev, Denmark
- The Copenhagen City Heart Study, Frederiksberg Hospital, Frederiksberg, Denmark
- Department of Clinical Biochemistry, Copenhagen University Hospital–Herlev Gentofte, Herlev, Denmark
| | - Ruth Frikke-Schmidt
- Department of Clinical Biochemistry, Copenhagen University Hospital–Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- The Copenhagen General Population Study, Herlev and Gentofte Hospital, Herlev, Denmark
| |
Collapse
|
15
|
Chen X, Jiang S, Wang R, Bao X, Li Y. Neural Stem Cells in the Treatment of Alzheimer's Disease: Current Status, Challenges, and Future Prospects. J Alzheimers Dis 2023; 94:S173-S186. [PMID: 36336934 PMCID: PMC10473082 DOI: 10.3233/jad-220721] [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] [Accepted: 09/27/2022] [Indexed: 11/06/2022]
Abstract
Alzheimer's disease (AD), a progressive dementia, is one of the world's most dangerous and debilitating diseases. Clinical trial results of amyloid-β (Aβ) and tau regulators based on the pretext of straightforward amyloid and tau immunotherapy were disappointing. There are currently no effective strategies for slowing the progression of AD. Further understanding of the mechanisms underlying AD and the development of novel therapeutic options are critical. Neurogenesis is impaired in AD, which contributes to memory deficits. Transplanted neural stem cells (NSCs) can regenerate degraded cholinergic neurons, and new neurons derived from NSCs can form synaptic connections with neighboring neurons. In theory, employing NSCs to replace and restore damaged cholinergic neurons and brain connections may offer new treatment options for AD. However there remain barriers to surmount before NSC-based therapy can be used clinically. The objective of this article is to describe recent advances in the treatment of AD models and clinical trials involving NSCs. In addition, we discuss the challenges and prospects associated with cell transplant therapy for AD.
Collapse
Affiliation(s)
- Xiaokun Chen
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Shenzhong Jiang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Renzhi Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Xinjie Bao
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yongning Li
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| |
Collapse
|
16
|
Sierra DP, Tripathi A, Pillai A. Dysregulation of complement system in neuropsychiatric disorders: A mini review. Biomark Neuropsychiatry 2022; 7. [PMID: 37123465 PMCID: PMC10136364 DOI: 10.1016/j.bionps.2022.100056] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Complement system is one of the most important defense mechanisms of the innate immune system. In addition to their roles in immune regulation, complement proteins are also involved in neurodevelopment and adult brain plasticity. Complement dysregulation has been shown in neurodevelopmental disorders including schizophrenia and autism spectrum disorder as well as in mood disorders. A number of clinical as well as genetic studies suggest the role of complement proteins in the cortical thinning and excessive synaptic pruning frequently associated with schizophrenia. The changes in complement proteins are also associated with the pathophysiology of autism spectrum disorder, major depressive disorder and bipolar disorder, but warrant further research. In addition, rodent models suggest a strong case for complement system in anxiety-like behavior. In this article, we review the recent findings on the role of complement system in neuropsychiatric disorders. The possible uses for future complement targeted therapies are also discussed.
Collapse
Affiliation(s)
- Danny Perez Sierra
- Pathophysiology of Neuropsychiatric Disorders Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Ashutosh Tripathi
- Pathophysiology of Neuropsychiatric Disorders Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Anilkumar Pillai
- Pathophysiology of Neuropsychiatric Disorders Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
- Research and Development, Charlie Norwood VA Medical Center, Augusta, GA, USA
- Department of Psychiatry and Health Behavior, Medical College of Georgia, Augusta University, Augusta, GA, USA
- Correspondence to: Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA. (A. Pillai)
| |
Collapse
|
17
|
Hegazy SH, Thomassen JQ, Rasmussen IJ, Nordestgaard BG, Tybjærg‐Hansen A, Frikke‐Schmidt R. C-reactive protein levels and risk of dementia-Observational and genetic studies of 111,242 individuals from the general population. Alzheimers Dement 2022; 18:2262-2271. [PMID: 35112776 PMCID: PMC9790296 DOI: 10.1002/alz.12568] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 10/22/2021] [Accepted: 12/10/2021] [Indexed: 01/31/2023]
Abstract
INTRODUCTION Increased plasma levels of C-reactive protein (CRP) in midlife are associated with increased risk of Alzheimer's disease (AD), whereas in older age the opposite association is observed. Whether genetically determined CRP is associated with AD remains unclear. METHODS A total of 111,242 White individuals from the Copenhagen General Population Study and the Copenhagen City Heart Study were included. Plasma levels of CRP and four regulatory genetic variants in the CRP gene were determined. RESULTS For CRP percentile group 1 to 5 (lowest plasma CRP) versus the 50 to 75 group (reference), the hazard ratio for AD was 1.69 (95% confidence interval 1.29-2.16). Genetically low CRP was associated with increased risk of AD in individuals with body mass index ≤25 kg/m2 (P = 4 × 10-6 ). DISCUSSION Low plasma levels of CRP at baseline were associated with high risk of AD in individuals from the general population. These observational findings were supported by genetic studies.
Collapse
Affiliation(s)
- Sharif H. Hegazy
- Department of Clinical BiochemistryCopenhagen University Hospital–RigshospitaletCopenhagenDenmark
| | - Jesper Qvist Thomassen
- Department of Clinical BiochemistryCopenhagen University Hospital–RigshospitaletCopenhagenDenmark
| | - Ida Juul Rasmussen
- Department of Clinical BiochemistryCopenhagen University Hospital–RigshospitaletCopenhagenDenmark
| | - Børge G. Nordestgaard
- The Copenhagen General Population StudyCopenhagen University Hospital–Herlev and GentofteHerlevDenmark,Department of Clinical BiochemistryCopenhagen University Hospital–Herlev and GentofteHerlevDenmark,The Copenhagen City Heart StudyCopenhagen University Hospital–Bispebjerg and FrederiksbergFrederiksbergDenmark,Department of Clinical MedicineUniversity of CopenhagenCopenhagenDenmark
| | - Anne Tybjærg‐Hansen
- Department of Clinical BiochemistryCopenhagen University Hospital–RigshospitaletCopenhagenDenmark,The Copenhagen General Population StudyCopenhagen University Hospital–Herlev and GentofteHerlevDenmark,The Copenhagen City Heart StudyCopenhagen University Hospital–Bispebjerg and FrederiksbergFrederiksbergDenmark,Department of Clinical MedicineUniversity of CopenhagenCopenhagenDenmark
| | - Ruth Frikke‐Schmidt
- Department of Clinical BiochemistryCopenhagen University Hospital–RigshospitaletCopenhagenDenmark,The Copenhagen General Population StudyCopenhagen University Hospital–Herlev and GentofteHerlevDenmark,Department of Clinical MedicineUniversity of CopenhagenCopenhagenDenmark
| |
Collapse
|
18
|
Carpanini SM, Torvell M, Bevan RJ, Byrne RAJ, Daskoulidou N, Saito T, Saido TC, Taylor PR, Hughes TR, Zelek WM, Morgan BP. Terminal complement pathway activation drives synaptic loss in Alzheimer's disease models. Acta Neuropathol Commun 2022; 10:99. [PMID: 35794654 PMCID: PMC9258209 DOI: 10.1186/s40478-022-01404-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 06/29/2022] [Indexed: 11/23/2022] Open
Abstract
Complement is involved in developmental synaptic pruning and pathological synapse loss in Alzheimer's disease. It is posited that C1 binding initiates complement activation on synapses; C3 fragments then tag them for microglial phagocytosis. However, the precise mechanisms of complement-mediated synaptic loss remain unclear, and the role of the lytic membrane attack complex (MAC) is unexplored. We here address several knowledge gaps: (i) is complement activated through to MAC at the synapse? (ii) does MAC contribute to synaptic loss? (iii) can MAC inhibition prevent synaptic loss? Novel methods were developed and optimised to quantify C1q, C3 fragments and MAC in total and regional brain homogenates and synaptoneurosomes from WT and AppNL-G-F Alzheimer's disease model mouse brains at 3, 6, 9 and 12 months of age. The impact on synapse loss of systemic treatment with a MAC blocking antibody and gene knockout of a MAC component was assessed in Alzheimer's disease model mice. A significant increase in C1q, C3 fragments and MAC was observed in AppNL-G-F mice compared to controls, increasing with age and severity. Administration of anti-C7 antibody to AppNL-G-F mice modulated synapse loss, reflected by the density of dendritic spines in the vicinity of plaques. Constitutive knockout of C6 significantly reduced synapse loss in 3xTg-AD mice. We demonstrate that complement dysregulation occurs in Alzheimer's disease mice involving the activation (C1q; C3b/iC3b) and terminal (MAC) pathways in brain areas associated with pathology. Inhibition or ablation of MAC formation reduced synapse loss in two Alzheimer's disease mouse models, demonstrating that MAC formation is a driver of synapse loss. We suggest that MAC directly damages synapses, analogous to neuromuscular junction destruction in myasthenia gravis.
Collapse
Affiliation(s)
- Sarah M Carpanini
- UK Dementia Research Institute Cardiff, and Systems Immunity Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK
| | - Megan Torvell
- UK Dementia Research Institute Cardiff, and Systems Immunity Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK
| | - Ryan J Bevan
- UK Dementia Research Institute Cardiff, and Systems Immunity Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK
| | - Robert A J Byrne
- UK Dementia Research Institute Cardiff, and Systems Immunity Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK
| | - Nikoleta Daskoulidou
- UK Dementia Research Institute Cardiff, and Systems Immunity Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Japan
| | - Philip R Taylor
- UK Dementia Research Institute Cardiff, and Systems Immunity Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK
| | - Timothy R Hughes
- UK Dementia Research Institute Cardiff, and Systems Immunity Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK
| | - Wioleta M Zelek
- UK Dementia Research Institute Cardiff, and Systems Immunity Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK
| | - B Paul Morgan
- UK Dementia Research Institute Cardiff, and Systems Immunity Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, CF24 4HQ, UK.
| |
Collapse
|
19
|
Systemic inflammatory markers in relation to cognitive function and measures of brain atrophy: a Mendelian randomization study. GeroScience 2022; 44:2259-2270. [PMID: 35689786 PMCID: PMC9616983 DOI: 10.1007/s11357-022-00602-7] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 06/03/2022] [Indexed: 11/07/2022] Open
Abstract
Observational studies have implied associations between multiple cytokines and cognitive decline, anti-inflammatory drugs however did not yield any protective effects on cognitive decline. We aimed to assess the associations of systemic inflammation, as measured by multiple cytokine and growth factor, with cognitive performance and brain atrophy using two-sample Mendelian randomization (MR). Independent genetic instruments (p < 5e − 8 and p < 5e − 6) for 41 systemic inflammatory markers were retrieved from a genome-wide association study conducted in 8293 Finnish participants. Summary statistics for gene-outcome associations were obtained for cognitive performance (N = 257,841) and for brain atrophy measures of cerebral cortical surface area and thickness (N = 51,665) and hippocampal volume (N = 33,536). To rule out the heterogeneity in the cognitive performance, we additionally included three domains: the fluid intelligence score (N = 108,818), prospective memory result (N = 111,099), and reaction time (N = 330,069). Main results were computed by inverse-variance weighting; sensitivity analyses taking pleiotropy and invalid instruments into account were performed by using weighted-median estimator, MR-Egger, and MR PRESSO. After correcting for multiple testing using false discovery rate, only genetically predicted (with p < 5e − 6 threshold) per-SD (standard deviation) higher IL-8 was associated with − 0.103 (− 0.155, − 0.051, padjusted = 0.004) mm3 smaller hippocampal volume and higher intelligence fluid score [β: 0.103 SD (95% CI: 0.042, 0.165), padjusted = 0.041]. Sensitivity analyses generally showed similar results, and no pleiotropic effect, heterogeneity, or possible reverse causation was detected. Our results suggested a possible causal association of high IL-8 levels with better cognitive performance but smaller hippocampal volume among the general healthy population, highlighting the complex role of inflammation in dementia-related phenotypes. Further research is needed to elucidate mechanisms underlying these associations.
Collapse
|
20
|
Hao J, Guo Y, Guo K, Yang Q. Peripheral Inflammatory Biomarkers of Alzheimer’s Disease. J Alzheimers Dis 2022; 88:389-398. [PMID: 35599478 DOI: 10.3233/jad-215422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disease of unknown pathological origin. The clinical diagnosis of AD is time-consuming and needs to a combination of clinical evaluation, psychological testing, and imaging assessments. Biomarkers may be good indicators for the clinical diagnosis of AD; hence, it is important to identify suitable biomarkers for the diagnosis and treatment of AD. Peripheral inflammatory biomarkers have been the focus of research in recent years. This review summarizes the role of inflammatory biomarkers in the disease course of AD.
Collapse
Affiliation(s)
- Jing Hao
- Department of Neurology, Anyang People’s Hospital, Xinxiang Medical University, Anyang, P.R. China
| | - Yanping Guo
- Department of Neurology, Anyang People’s Hospital, Xinxiang Medical University, Anyang, P.R. China
| | - Keke Guo
- Department of Neurology, Anyang People’s Hospital, Xinxiang Medical University, Anyang, P.R. China
| | - Qingcheng Yang
- Department of Neurology, Anyang People’s Hospital, Xinxiang Medical University, Anyang, P.R. China
| |
Collapse
|
21
|
Revuelta M, Urrutia J, Villarroel A, Casis O. Microglia-Mediated Inflammation and Neural Stem Cell Differentiation in Alzheimer's Disease: Possible Therapeutic Role of K V1.3 Channel Blockade. Front Cell Neurosci 2022; 16:868842. [PMID: 35530176 PMCID: PMC9070300 DOI: 10.3389/fncel.2022.868842] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 03/31/2022] [Indexed: 12/14/2022] Open
Abstract
Increase of deposits of amyloid β peptides in the extracellular matrix is landmark during Alzheimer’s Disease (AD) due to the imbalance in the production vs. clearance. This accumulation of amyloid β deposits triggers microglial activation. Microglia plays a dual role in AD, a protective role by clearing the deposits of amyloid β peptides increasing the phagocytic response (CD163, IGF-1 or BDNF) and a cytotoxic role, releasing free radicals (ROS or NO) and proinflammatory cytokines (TNF-α, IL-1β) in response to reactive gliosis activated by the amyloid β aggregates. Microglia activation correlated with an increase KV1.3 channels expression, protein levels and current density. Several studies highlight the importance of KV1.3 in the activation of inflammatory response and inhibition of neural progenitor cell proliferation and neuronal differentiation. However, little is known about the pathways of this activation in neural stem cells differentiation and proliferation and the role in amyloid β accumulation. In recent studies using in vitro cells derived from mice models, it has been demonstrated that KV1.3 blockers inhibit microglia-mediated neurotoxicity in culture reducing the expression and production of the pro-inflammatory cytokines IL-1β and TNF-α through the NF-kB and p38MAPK pathway. Overall, we conclude that KV1.3 blockers change the course of AD development, reducing microglial cytotoxic activation and increasing neural stem cell differentiation. However, further investigations are needed to establish the specific pathway and to validate the use of this blocker as therapeutic treatment in Alzheimer patients.
Collapse
Affiliation(s)
- Miren Revuelta
- Department of Physiology, Faculty of Medicine and Nursery, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Janire Urrutia
- Department of Physiology, Faculty of Medicine and Nursery, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Alvaro Villarroel
- Instituto Biofisika, Consejo Superior de Investigaciones Científicas (CSIC)-University of the Basque Country/Euskal Herriko Unibertsitatea (UPV/EHU), Leioa, Spain
| | - Oscar Casis
- Department of Physiology, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
| |
Collapse
|
22
|
Xiang J, Meng X, Zhao Y, Wu FX, Li M. HyMM: hybrid method for disease-gene prediction by integrating multiscale module structure. Brief Bioinform 2022; 23:6547263. [PMID: 35275996 DOI: 10.1093/bib/bbac072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 01/18/2022] [Accepted: 02/13/2022] [Indexed: 11/12/2022] Open
Abstract
MOTIVATION Identifying disease-related genes is an important issue in computational biology. Module structure widely exists in biomolecule networks, and complex diseases are usually thought to be caused by perturbations of local neighborhoods in the networks, which can provide useful insights for the study of disease-related genes. However, the mining and effective utilization of the module structure is still challenging in such issues as a disease gene prediction. RESULTS We propose a hybrid disease-gene prediction method integrating multiscale module structure (HyMM), which can utilize multiscale information from local to global structure to more effectively predict disease-related genes. HyMM extracts module partitions from local to global scales by multiscale modularity optimization with exponential sampling, and estimates the disease relatedness of genes in partitions by the abundance of disease-related genes within modules. Then, a probabilistic model for integration of gene rankings is designed in order to integrate multiple predictions derived from multiscale module partitions and network propagation, and a parameter estimation strategy based on functional information is proposed to further enhance HyMM's predictive power. By a series of experiments, we reveal the importance of module partitions at different scales, and verify the stable and good performance of HyMM compared with eight other state-of-the-arts and its further performance improvement derived from the parameter estimation. CONCLUSIONS The results confirm that HyMM is an effective framework for integrating multiscale module structure to enhance the ability to predict disease-related genes, which may provide useful insights for the study of the multiscale module structure and its application in such issues as a disease-gene prediction.
Collapse
Affiliation(s)
- Ju Xiang
- Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha 410083, China; Department of Basic Medical Sciences & Academician Workstation, Changsha Medical University, Changsha, Hunan 410219, China
| | - Xiangmao Meng
- School of Computer Science and Engineering, Central South University, Changsha 410083, China
| | - Yichao Zhao
- School of Computer Science and Engineering, Central South University, Changsha, Hunan 410083, China
| | - Fang-Xiang Wu
- Division of Biomedical Engineering and Department of Mechanical Engineering, University of Saskatchewan, Saskatoon, SK, S7N 5A9, Canada
| | - Min Li
- Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha 410083, China
| |
Collapse
|
23
|
Li Z, Zhang W, Gao F, Tang Q, Kang D, Shen Y. Different Complement Activation Pathways Underly Cognitive Impairment and Type 2 Diabetes Mellitus Combined With Cognitive Impairment. Front Aging Neurosci 2022; 14:810335. [PMID: 35370615 PMCID: PMC8967361 DOI: 10.3389/fnagi.2022.810335] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 01/31/2022] [Indexed: 11/17/2022] Open
Abstract
Background The immune response and the complement system are associated with cognitive impairment and diabetes mellitus, respectively. Activation of the complement system in these diseases occurs mainly through either the classical pathway or the alternative pathway. However, the specific complement proteins involved in the development of the type 2 diabetes mellitus (T2DM) and cognitive impairment are still unclear. Here, we investigated complement proteins in serum from patients with T2DM, cognitive impairment, or both T2DM and cognitive impairment. Objective To investigate the levels of serum immune complement proteins in patients with T2DM, cognitive impairment, or T2DM combined with cognitive impairment and the associations between these complement proteins and risk factors for T2DM or cognitive impairment. Methods Clinical markers were collected from blood samples of 264 participants. Luminex multiplex assays were used to detect serum complement proteins. All statistical analyses were performed using Prism or R studio. Results There was a difference in serum levels of the complement proteins C1q, C3, C3b, and FH between the three different groups. Hyperglycemia was significantly correlated with elevated C3b or reduced C3, C1q, and FH. In addition, hyperlipidemia was positively correlated with elevated levels of C3, C4, C1q, and FH proteins. There was an association between C1q, C3, C4, and FH and β-pancreas cell function, whereas only FH was associated with insulin resistance. Higher serum C1q was significantly associated with an increased risk of cognitive impairment. Conclusion Serum levels of complement proteins were closely associated with hyperglycemia and hyperlipidemia. We found that classical complement pathway activation mainly occurred in the cognitive impairment only group, whereas the alternative pathway may reflect T2DM and T2DM with cognitive impairment.
Collapse
Affiliation(s)
- Zhenxing Li
- Department of Neurology and Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Weiwei Zhang
- Department of Neurology and Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Feng Gao
- Department of Neurology and Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Qiqiang Tang
- Department of Neurology and Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- *Correspondence: Yong Shen,
| | - Dongmei Kang
- Department of Geriatric Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- *Correspondence: Yong Shen,
| | - Yong Shen
- Department of Neurology and Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- *Correspondence: Yong Shen,
| |
Collapse
|
24
|
Uchida K. Waste Clearance in the Brain and Neuroinflammation: A Novel Perspective on Biomarker and Drug Target Discovery in Alzheimer's Disease. Cells 2022; 11:cells11050919. [PMID: 35269541 PMCID: PMC8909773 DOI: 10.3390/cells11050919] [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: 01/05/2022] [Revised: 02/26/2022] [Accepted: 03/04/2022] [Indexed: 02/06/2023] Open
Abstract
Alzheimer’s disease (AD) is a multifactorial disease with a heterogeneous etiology. The pathology of Alzheimer’s disease is characterized by amyloid-beta and hyperphosphorylated tau, which are necessary for disease progression. Many clinical trials on disease-modifying drugs for AD have failed to indicate their clinical benefits. Recent advances in fundamental research have indicated that neuroinflammation plays an important pathological role in AD. Damage- and pathogen-associated molecular patterns in the brain induce neuroinflammation and inflammasome activation, causing caspase-1-dependent glial and neuronal cell death. These waste products in the brain are eliminated by the glymphatic system via perivascular spaces, the blood-brain barrier, and the blood–cerebrospinal fluid barrier. Age-related vascular dysfunction is associated with an impairment of clearance and barrier functions, leading to neuroinflammation. The proteins involved in waste clearance in the brain and peripheral circulation may be potential biomarkers and drug targets in the early stages of cognitive impairment. This short review focuses on waste clearance dysfunction in AD pathobiology and discusses the improvement of waste clearance as an early intervention in prodromal AD and preclinical stages of dementia.
Collapse
Affiliation(s)
- Kazuhiko Uchida
- Faculty of Medicine, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba 305-8575, Ibaraki, Japan; ; Tel.: +81-29-853-3210; Fax: +81-50-3730-7456
- Institute for Biomedical Research, MCBI, 4-9-29 Matsushiro, Tsukuba 305-0035, Ibaraki, Japan
| |
Collapse
|
25
|
Soares Martins T, Marçalo R, da Cruz E Silva CB, Trindade D, Catita J, Amado F, Melo T, Rosa IM, Vogelgsang J, Wiltfang J, da Cruz E Silva OAB, Henriques AG. Novel Exosome Biomarker Candidates for Alzheimer's Disease Unravelled Through Mass Spectrometry Analysis. Mol Neurobiol 2022; 59:2838-2854. [PMID: 35212939 PMCID: PMC9016047 DOI: 10.1007/s12035-022-02762-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 01/28/2022] [Indexed: 01/03/2023]
Abstract
Exosomes are small extracellular vesicles (EVs) present in human biofluids that can transport specific disease-associated molecules. Consequently blood-derived exosomes have emerged as important peripheral biomarker sources for a wide range of diseases, among them Alzheimer’s disease (AD). Although there is no effective cure for AD, an accurate diagnosis, relying on easily accessible peripheral biofluids, is still necessary to discriminate this disease from other dementias, test potential therapies and even monitor rate of disease progression. The ultimate goal is to produce a cost-effective and widely available alternative, which can also be employed as a first clinical screen. In this study, EVs with exosome-like characteristics were isolated from serum of Controls and AD cases through precipitation- and column-based methods, followed by mass spectrometry analysis. The resulting proteomes were characterized by Gene Ontology (GO) and multivariate analyses. Although GO terms were similar for exosomes’ proteomes of Controls and ADs, using both methodologies, a clear segregation of disease cases was obtained when using the precipitation-based method. Nine significantly different abundant proteins were identified between Controls and AD cases, representing putative biomarker candidate targets. Among them are AACT and C4BPα, two Aβ-binding proteins, whose exosome levels were further validated in individuals from independent cohorts using antibody-based approaches. The findings discussed represent an important contribution to the identification of novel exosomal biomarker candidates useful as potential blood-based tools for AD diagnosis.
Collapse
Affiliation(s)
- Tânia Soares Martins
- Neuroscience and Signalling Group, Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro (UA), 3810-193, Aveiro, Portugal
| | - Rui Marçalo
- Neuroscience and Signalling Group, Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro (UA), 3810-193, Aveiro, Portugal
| | - Cristóvão B da Cruz E Silva
- Laboratory of Instrumentation and Experimental Particle Physics-LIP, Av. Elias Garcia 14-1º, 1000-149, Lisbon, Portugal
| | - Dário Trindade
- Neuroscience and Signalling Group, Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro (UA), 3810-193, Aveiro, Portugal
| | - José Catita
- CEBIMED-Faculty of Health Sciences, University Fernando Pessoa, 4249-004, Porto, Portugal.,Paralab SA, 4420-437, Gondomar, Portugal
| | - Francisco Amado
- Department of Chemistry, QOPNA (Organic Chemistry Natural and Agrofood Products and LAVQ REQUIMTE), University of Aveiro, 3810-193, Aveiro, Portugal
| | - Tânia Melo
- Department of Chemistry, QOPNA (Organic Chemistry Natural and Agrofood Products and LAVQ REQUIMTE), University of Aveiro, 3810-193, Aveiro, Portugal
| | - Ilka Martins Rosa
- Neuroscience and Signalling Group, Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro (UA), 3810-193, Aveiro, Portugal
| | - Jonathan Vogelgsang
- Department of Psychiatry and Psychotherapy, University Medical Center Goettingen (UMG), Georg-August University, Von-Siebold-Str. 5, 37075, Goettingen, Germany.,Translational Neuroscience Laboratory, McLean Hospital, Harvard Medical School, Belmont, MA, 02478, USA
| | - Jens Wiltfang
- Neuroscience and Signalling Group, Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro (UA), 3810-193, Aveiro, Portugal.,Department of Psychiatry and Psychotherapy, University Medical Center Goettingen (UMG), Georg-August University, Von-Siebold-Str. 5, 37075, Goettingen, Germany.,German Center for Neurodegenerative Diseases (DZNE), Von-Siebold-Str. 3a, 37075, Goettingen, Germany
| | - Odete A B da Cruz E Silva
- Neuroscience and Signalling Group, Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro (UA), 3810-193, Aveiro, Portugal
| | - Ana Gabriela Henriques
- Neuroscience and Signalling Group, Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro (UA), 3810-193, Aveiro, Portugal.
| |
Collapse
|
26
|
Ghosh P, Singh R, Ganeshpurkar A, Pokle AV, Singh RB, Singh SK, Kumar A. Cellular and molecular influencers of neuroinflammation in Alzheimer's disease: Recent concepts & roles. Neurochem Int 2021; 151:105212. [PMID: 34656693 DOI: 10.1016/j.neuint.2021.105212] [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: 03/26/2021] [Revised: 09/22/2021] [Accepted: 10/10/2021] [Indexed: 01/21/2023]
Abstract
Alzheimer's disease (AD), an extremely common neurodegenerative disorder of the older generation, is one of the leading causes of death globally. Besides the conventional hallmarks i.e. Amyloid-β (Aβ) plaques and neurofibrillary tangles (NFTs), neuroinflammation also serves as a major contributing factor in the pathogenesis of AD. There are mounting evidences to support the fundamental role of cellular (microglia, astrocytes, mast cells, and T-cells) and molecular (cytokines, chemokines, caspases, and complement proteins) influencers of neuroinflammation in producing/promoting neurodegeneration and dementia in AD. Genome-wide association studies (GWAS) have revealed the involvement of various single nucleotide polymorphisms (SNPs) of genes related to neuroinflammation with the risk of developing AD. Modulating the release of the neuroinflammatory molecules and targeting their relevant mechanisms may have beneficial effects on the onset, progress and severity of the disease. Here, we review the distinct role of various mediators and modulators of neuroinflammation that impact the pathogenesis and progression of AD as well as incite further research efforts for the treatment of AD through a neuroinflammatory approach.
Collapse
Affiliation(s)
- Powsali Ghosh
- Pharmaceutical Chemistry Research Laboratory 1, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Ravi Singh
- Pharmaceutical Chemistry Research Laboratory 1, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Ankit Ganeshpurkar
- Pharmaceutical Chemistry Research Laboratory 1, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Ankit Vyankatrao Pokle
- Pharmaceutical Chemistry Research Laboratory 1, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Ravi Bhushan Singh
- Institute of Pharmacy Harischandra PG College, Bawanbigha, Varanasi, India
| | - Sushil Kumar Singh
- Pharmaceutical Chemistry Research Laboratory 1, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Ashok Kumar
- Pharmaceutical Chemistry Research Laboratory 1, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India.
| |
Collapse
|
27
|
Hao J, Qiao Y, Li T, Yang J, Song Y, Jia L, Jia J. Investigating Changes in the Serum Inflammatory Factors in Alzheimer's Disease and Their Correlation with Cognitive Function. J Alzheimers Dis 2021; 84:835-842. [PMID: 34602472 DOI: 10.3233/jad-210552] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Serum levels of inflammatory factors, such as C3, C4, C-reactive protein (CRP), immunoglobulin (Ig) G, IgA, and IgM, in patients with Alzheimer's disease (AD) and their correlation with cognitive function remain unexplored. OBJECTIVE To investigate the expression of serum inflammatory factors in patients with AD and its correlation with cognitive function. METHODS Serum levels of C3, C4, CRP, IgG, IgA, and IgM in 200 patients with AD (mild, moderate, and severe) and those in 174 normal controls were assessed. Spearman's rank correlation analysis was used to explore the relationships among biomarker levels, cognitive function, and activities of daily living (ADL). RESULTS Among these inflammatory factors, C3 and CRP levels were significantly lower, and IgG and IgA levels were significantly higher in the AD group than in the control group (p < 0.05). There were no significant differences in C4 and IgM levels between the two groups (p > 0.05). In all participants, CRP level was positively correlated with the Mini-Mental State Examination and Montreal Cognitive Assessment scores (p < 0.05). In the AD group, IgA level was negatively associated with ADL scores (p < 0.05). No significant correlation was detected between the other factors and different cognitive scores (p > 0.05). CONCLUSION Inflammatory factors C3, CRP, IgG, and IgA have the potential to serve as biomarkers for AD. Furthermore, serum IgA was not only correlated with AD but also with ADL. These results support the hypothesis that inflammation is involved in the occurrence and development of AD.
Collapse
Affiliation(s)
- Jing Hao
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, P.R. China
| | - Yuchen Qiao
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, P.R. China
| | - Tingting Li
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, P.R. China
| | - Jianwei Yang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, P.R. China
| | - Yang Song
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, P.R. China
| | - Longfei Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, P.R. China
| | - Jianping Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, P.R. China.,Beijing Key Laboratory of Geriatric Cognitive Disorders, Beijing, P.R. China.,Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, Beijing, P.R. China.,Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, P.R. China.,Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, P.R. China
| |
Collapse
|
28
|
The complement cascade in Alzheimer's disease: a systematic review and meta-analysis. Mol Psychiatry 2021; 26:5532-5541. [PMID: 31628417 DOI: 10.1038/s41380-019-0536-8] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 09/10/2019] [Accepted: 09/20/2019] [Indexed: 12/17/2022]
Abstract
Genetic evidence implicates a causal role for the complement pathway in Alzheimer's disease (AD). Since studies have shown inconsistent differences in cerebrospinal fluid (CSF) and peripheral blood complement protein concentrations between AD patients and healthy elderly, this study sought to summarize the clinical data. Original peer-reviewed articles measuring CSF and/or blood concentrations of complement or complement regulator protein concentrations in AD and healthy elderly control (HC) groups were included. Of 2966 records identified, means and standard deviations from 86 studies were summarized as standardized mean differences (SMD) by random effects meta-analyses. In CSF, concentrations of clusterin (NAD/NHC = 625/577, SMD = 0.53, Z8 = 8.81, p < 0.005; I2 < 0.005%) and complement component 3 (C3; NAD/NHC = 299/522, SMD = 0.45, Z3 = 3.21, p < 0.005; I2 = 68.40%) were significantly higher in AD, but differences in C1q, C-reactive protein (CRP), serum amyloid protein (SAP), and factor H concentrations were not significant. In peripheral blood, concentrations of CRP were elevated in AD (NAD/NHC = 3404/3332, SMD = 0.44, Z43 = 3.43, p < 0.005; I2 = 93.81%), but differences between groups in C3, C4, C1-inhibitor, SAP, factor H and clusterin concentrations were not significant, and inconsistent between studies. Of 64 complement pathway proteins or regulators in the quantitative synthesis, trends in C1q, factor B, C4a, and late-stage complement pathway components (e.g. C9) in blood, C4 in CSF, and the membrane attack complex in blood and CSF, might be investigated further. The results collectively support elevated complement pathway activity in AD, which was best characterized by increased CSF clusterin concentrations and less consistently by CSF C3 concentrations. Complement activity related to an AD diagnosis was not reflected consistently by the peripheral blood proteins investigated.
Collapse
|
29
|
Wu KM, Zhang YR, Huang YY, Dong Q, Tan L, Yu JT. The role of the immune system in Alzheimer's disease. Ageing Res Rev 2021; 70:101409. [PMID: 34273589 DOI: 10.1016/j.arr.2021.101409] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 07/09/2021] [Accepted: 07/12/2021] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder where the accumulation of amyloid plaques and the formation of tau tangles are the prominent pathological hallmarks. Increasing preclinical and clinical studies have revealed that different components of the immune system may act as important contributors to AD etiology and pathogenesis. The recognition of misfolded Aβ and tau by immune cells can trigger a series of complex immune responses in AD, and then lead to neuroinflammation and neurodegeneration. In parallel, genome-wide association studies have also identified several immune related loci associated with increased - risk of AD by interfering with the function of immune cells. Other immune related factors, such as impaired immunometabolism, defective meningeal lymphatic vessels and autoimmunity might also be involved in the pathogenesis of AD. Here, we review the data showing the alterations of immune cells in the AD trajectory and seek to demonstrate the crosstalk between the immune cell dysfunction and AD pathology. We then discuss the most relevant research findings in regards to the influences of gene susceptibility of immune cells for AD. We also consider impaired meningeal lymphatics, immunometabolism and autoimmune mechanisms in AD. In addition, immune related biomarkers and immunotherapies for AD are also mentioned in order to offer novel insights for future research.
Collapse
|
30
|
Byrne RAJ, Torvell M, Daskoulidou N, Fathalla D, Kokkali E, Carpanini SM, Morgan BP. Novel Monoclonal Antibodies Against Mouse C1q: Characterisation and Development of a Quantitative ELISA for Mouse C1q. Mol Neurobiol 2021; 58:4323-4336. [PMID: 34002346 PMCID: PMC8487419 DOI: 10.1007/s12035-021-02419-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 04/30/2021] [Indexed: 12/20/2022]
Abstract
Recent studies have identified roles for complement in synaptic pruning, both physiological during development and pathological in Alzheimer's disease (AD). These reports suggest that C1q initiates complement activation on synapses and C3 fragments then tag them for removal by microglia. There is an urgent need to characterise these processes in rodent AD models; this requires the development of reagents and methods for detection and quantification of rodent C1q in fluids and pathological tissues. These will enable better evaluation of the role of C1q in disease and its value as disease biomarker. We describe the generation in C1q-deficient mice of novel monoclonal antibodies against mouse and rat C1q that enabled development of a sensitive, specific, and quantitative ELISA for mouse and rat C1q capable of measuring C1q in biological fluids and tissue extracts. Serum C1q levels were measured in wild-type (WT), C1q knockout (KO), C3 KO, C7 KO, Crry KO, and 3xTg and APPNL-G-F AD model mice through ageing. C1q levels significantly decreased in WT, APPNL-G-F, and C7 KO mice with ageing. C1q levels were reduced in APPNL-G-F compared to WT at all ages and in 3xTg at 12 months; C3 KO and C7 KO, but not Crry KO mice, also demonstrated significantly lower C1q levels compared to matched WT. In brain homogenates, C1q levels increased with age in both WT and APPNL-G-F mice. This robust and adaptable assay for quantification of mouse and rat C1q provides a vital tool for investigating the expression of C1q in rodent models of AD and other complement-driven pathologies.
Collapse
Affiliation(s)
- Robert A J Byrne
- UK Dementia Research Institute Cardiff, Hadyn Ellis Building, Cardiff University, Maindy Road, Cardiff, CF244HQ, UK.,Division of Infection and Immunity and Systems Immunity Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Heath Park, Cardiff, CF144XN, UK
| | - Megan Torvell
- UK Dementia Research Institute Cardiff, Hadyn Ellis Building, Cardiff University, Maindy Road, Cardiff, CF244HQ, UK.,Division of Infection and Immunity and Systems Immunity Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Heath Park, Cardiff, CF144XN, UK
| | - Nikoleta Daskoulidou
- UK Dementia Research Institute Cardiff, Hadyn Ellis Building, Cardiff University, Maindy Road, Cardiff, CF244HQ, UK.,Division of Infection and Immunity and Systems Immunity Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Heath Park, Cardiff, CF144XN, UK
| | - Dina Fathalla
- UK Dementia Research Institute Cardiff, Hadyn Ellis Building, Cardiff University, Maindy Road, Cardiff, CF244HQ, UK.,Division of Infection and Immunity and Systems Immunity Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Heath Park, Cardiff, CF144XN, UK
| | - Eirini Kokkali
- School of Optometry and Visual Sciences, Cardiff University, Maindy Road, Cardiff, CF244HQ, UK
| | - Sarah M Carpanini
- UK Dementia Research Institute Cardiff, Hadyn Ellis Building, Cardiff University, Maindy Road, Cardiff, CF244HQ, UK.,Division of Infection and Immunity and Systems Immunity Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Heath Park, Cardiff, CF144XN, UK
| | - B Paul Morgan
- UK Dementia Research Institute Cardiff, Hadyn Ellis Building, Cardiff University, Maindy Road, Cardiff, CF244HQ, UK. .,Division of Infection and Immunity and Systems Immunity Research Institute, School of Medicine, Cardiff University, Hadyn Ellis Building, Heath Park, Cardiff, CF144XN, UK.
| |
Collapse
|
31
|
Moin ASM, Al-Qaissi A, Sathyapalan T, Atkin SL, Butler AE. Glucose excursions in type 2 diabetes modulate amyloid-related proteins associated with dementia. J Transl Med 2021; 19:131. [PMID: 33789659 PMCID: PMC8011191 DOI: 10.1186/s12967-021-02797-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 03/18/2021] [Indexed: 12/02/2022] Open
Affiliation(s)
- Abu Saleh Md Moin
- Diabetes Research Center (DRC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), PO Box 34110, Doha, Qatar
| | - Ahmed Al-Qaissi
- Academic Endocrinology, Diabetes and Metabolism, Hull York Medical School, Hull, UK.,Leeds Medical School, Leeds, UK
| | | | | | - Alexandra E Butler
- Diabetes Research Center (DRC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), PO Box 34110, Doha, Qatar.
| |
Collapse
|
32
|
Bray MJC, Richey LN, Bryant BR, Krieg A, Jahed S, Tobolowsky W, LoBue C, Peters ME. Traumatic brain injury alters neuropsychiatric symptomatology in all-cause dementia. Alzheimers Dement 2021; 17:686-691. [PMID: 33470043 DOI: 10.1002/alz.12225] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 10/02/2020] [Accepted: 10/07/2020] [Indexed: 11/08/2022]
Abstract
INTRODUCTION Traumatic brain injury (TBI) may alter the course of neuropsychiatric symptom (NPS) onset during dementia development. The connection among TBI, NPS, and dementia progression is of increasing interest to researchers and clinicians. METHODS Incidence of NPS was examined in participants with normal cognition who progressed to all-cause dementia based on whether TBI history was present (n = 130) or absent (n = 849). Survival analyses were used to examine NPS incidence across 7.6 ± 3.0 years of follow-up. RESULTS Participants with TBI history had increased prevalence and incidence of apathy (44.7% vs 29.9%, P = .0062; HRadj. = 1.708, P = .0018) and motor disturbances (17.2% vs 9.5%, P = .0458; HRadj. = 2.023, P = .0168), controlling for demographics and type of dementia diagnosis. Earlier anxiety onset was associated with TBI (692 days prior to dementia diagnosis vs 161 days, P = .0265). DISCUSSION History of TBI is associated with increased risk for and earlier onset of NPS in the trajectory of dementia development.
Collapse
Affiliation(s)
- Michael J C Bray
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Lisa N Richey
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Barry R Bryant
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Akshay Krieg
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sahar Jahed
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - William Tobolowsky
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Christian LoBue
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Matthew E Peters
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
33
|
Handy A, Lord J, Green R, Xu J, Aarsland D, Velayudhan L, Hye A, Dobson R, Proitsi P, on behalf of the Alzheimer’s Disease Neuroimaging initiative, AddNeuroMed, and the GERAD1 Consortium. Assessing Genetic Overlap and Causality Between Blood Plasma Proteins and Alzheimer's Disease. J Alzheimers Dis 2021; 83:1825-1839. [PMID: 34459398 PMCID: PMC8609677 DOI: 10.3233/jad-210462] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/30/2021] [Indexed: 12/21/2022]
Abstract
BACKGROUND Blood plasma proteins have been associated with Alzheimer's disease (AD), but understanding which proteins are on the causal pathway remains challenging. OBJECTIVE Investigate the genetic overlap between candidate proteins and AD using polygenic risk scores (PRS) and interrogate their causal relationship using bi-directional Mendelian randomization (MR). METHODS Following a literature review, 31 proteins were selected for PRS analysis. PRS were constructed for prioritized proteins with and without the apolipoprotein E region (APOE+/-PRS) and tested for association with AD status across three cohorts (n = 6,244). An AD PRS was also tested for association with protein levels in one cohort (n = 410). Proteins showing association with AD were taken forward for MR. RESULTS For APOE ɛ3, apolipoprotein B-100, and C-reactive protein (CRP), protein APOE+ PRS were associated with AD below Bonferroni significance (pBonf, p < 0.00017). No protein APOE- PRS or AD PRS (APOE+/-) passed pBonf. However, vitamin D-binding protein (protein PRS APOE-, p = 0.009) and insulin-like growth factor-binding protein 2 (AD APOE- PRS p = 0.025, protein APOE- PRS p = 0.045) displayed suggestive signals and were selected for MR. In bi-directional MR, none of the five proteins demonstrated a causal association (p < 0.05) in either direction. CONCLUSION Apolipoproteins and CRP PRS are associated with AD and provide a genetic signal linked to a specific, accessible risk factor. While evidence of causality was limited, this study was conducted in a moderate sample size and provides a framework for larger samples with greater statistical power.
Collapse
Affiliation(s)
- Alex Handy
- University College London, Institute of Health Informatics, London, UK
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, London, UK
| | - Jodie Lord
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, London, UK
| | - Rebecca Green
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, London, UK
- NIHR Maudsley Biomedical Research Centre, South London and Maudsley NHS Trust, London, UK
| | - Jin Xu
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, London, UK
- Institute of Pharmaceutical Science, King’s College London, UK
| | - Dag Aarsland
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, London, UK
- Center for Age-Related Medicine, Stavanger University Hospital, Norway
| | - Latha Velayudhan
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, London, UK
| | - Abdul Hye
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, London, UK
| | - Richard Dobson
- University College London, Institute of Health Informatics, London, UK
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, London, UK
- NIHR Maudsley Biomedical Research Centre, South London and Maudsley NHS Trust, London, UK
- Health Data Research UK London, University College London, London, UK
- NIHR Biomedical Research Centre at University College London Hospitals NHS Foundation Trust, London, UK
| | - Petroula Proitsi
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, London, UK
| | | | | |
Collapse
|
34
|
Kanmogne M, Klein RS. Neuroprotective versus Neuroinflammatory Roles of Complement: From Development to Disease. Trends Neurosci 2020; 44:97-109. [PMID: 33190930 DOI: 10.1016/j.tins.2020.10.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 09/21/2020] [Accepted: 10/08/2020] [Indexed: 12/12/2022]
Abstract
Complement proteins are ancient components of innate immunity that have emerged as crucial regulators of neural networks. We discuss these roles in the context of the CNS development, acute CNS viral infections, and post-infectious and noninfectious CNS disorders, with an emphasis on microglia-mediated loss of synapses. Despite extensive examples that implicate classical complement proteins and their receptors in CNS dysfunction, recent data suggest that they exert neuroprotective roles in CNS homeostasis through continued refinement of synaptic connections. Thorough understanding of the mechanisms involved in these processes may lead to novel targets for the treatment of CNS diseases involving aberrant complement-mediated synapse loss.
Collapse
Affiliation(s)
- Marlene Kanmogne
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Robyn S Klein
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
35
|
Yang J, Wise L, Fukuchi KI. TLR4 Cross-Talk With NLRP3 Inflammasome and Complement Signaling Pathways in Alzheimer's Disease. Front Immunol 2020; 11:724. [PMID: 32391019 PMCID: PMC7190872 DOI: 10.3389/fimmu.2020.00724] [Citation(s) in RCA: 226] [Impact Index Per Article: 45.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 03/30/2020] [Indexed: 01/02/2023] Open
Abstract
Amyloid plaques, mainly composed of abnormally aggregated amyloid β-protein (Aβ) in the brain parenchyma, and neurofibrillary tangles (NFTs), consisting of hyperphosphorylated tau protein aggregates in neurons, are two pathological hallmarks of Alzheimer's disease (AD). Aβ fibrils and tau aggregates in the brain are closely associated with neuroinflammation and synapse loss, characterized by activated microglia and dystrophic neurites. Genome-wide genetic association studies revealed important roles of innate immune cells in the pathogenesis of late-onset AD by recognizing a dozen genetic risk loci that modulate innate immune activities. Furthermore, microglia, brain resident innate immune cells, have been increasingly recognized to play key, opposing roles in AD pathogenesis by either eliminating toxic Aβ aggregates and enhancing neuronal plasticity or producing proinflammatory cytokines, reactive oxygen species, and synaptotoxicity. Aggregated Aβ binds to toll-like receptor 4 (TLR4) and activates microglia, resulting in increased phagocytosis and cytokine production. Complement components are associated with amyloid plaques and NFTs. Aggregated Aβ can activate complement, leading to synapse pruning and loss by microglial phagocytosis. Systemic inflammation can activate microglial TLR4, NLRP3 inflammasome, and complement in the brain, leading to neuroinflammation, Aβ accumulation, synapse loss and neurodegeneration. The host immune response has been shown to function through complex crosstalk between the TLR, complement and inflammasome signaling pathways. Accordingly, targeting the molecular mechanisms underlying the TLR-complement-NLRP3 inflammasome signaling pathways can be a preventive and therapeutic approach for AD.
Collapse
Affiliation(s)
- Junling Yang
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, IL, United States
| | - Leslie Wise
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, IL, United States
| | - Ken-Ichiro Fukuchi
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, IL, United States
| |
Collapse
|
36
|
Shim K, Begum R, Yang C, Wang H. Complement activation in obesity, insulin resistance, and type 2 diabetes mellitus. World J Diabetes 2020; 11:1-12. [PMID: 31938469 PMCID: PMC6927818 DOI: 10.4239/wjd.v11.i1.1] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 11/07/2019] [Accepted: 11/26/2019] [Indexed: 02/05/2023] Open
Abstract
Amplified inflammatory reaction has been observed to be involved in cardiometabolic diseases such as obesity, insulin resistance, diabetes, dyslipidemia, and atherosclerosis. The complement system was originally viewed as a supportive first line of defense against microbial invaders, and research over the past decade has come to appreciate that the functions of the complement system extend beyond the defense and elimination of microbes, involving in such diverse processes as clearance of the immune complexes, complementing T and B cell immune functions, tissue regeneration, and metabolism. The focus of this review is to summarize the role of the activation of complement system and the initiation and progression of metabolic disorders including obesity, insulin resistance and diabetes mellitus. In addition, we briefly describe the interaction of the activation of the complement system with diabetic complications such as diabetic retinopathy, nephropathy and neuropathy, highlighting that targeting complement system therapeutics could be one of possible routes to slow down those aforementioned diabetic complications.
Collapse
Affiliation(s)
- Kyumin Shim
- Department of Basic Science, California Northstate University College of Medicine, Elk Grove, CA 95757, United States
| | - Rayhana Begum
- Department of Pharmacy, Primeasia University, Dhaka 1213, Bangladesh
| | - Catherine Yang
- Department of Basic Science, California Northstate University College of Medicine, Elk Grove, CA 95757, United States
- California Northstate University College of Graduate Studies, Elk Grove, CA 95757, United States
| | - Hongbin Wang
- Department of Basic Science, California Northstate University College of Medicine, Elk Grove, CA 95757, United States
- California Northstate University College of Graduate Studies, Elk Grove, CA 95757, United States
- Department of Pharmaceutical and Biomedical Sciences, California Northstate University College of Pharmacy, Elk Grove, CA 95757, United States
| |
Collapse
|
37
|
Shi L, Winchester LM, Liu BY, Killick R, Ribe EM, Westwood S, Baird AL, Buckley NJ, Hong S, Dobricic V, Kilpert F, Franke A, Kiddle S, Sattlecker M, Dobson R, Cuadrado A, Hye A, Ashton NJ, Morgan AR, Bos I, Vos SJ, ten Kate M, Scheltens P, Vandenberghe R, Gabel S, Meersmans K, Engelborghs S, De Roeck EE, Sleegers K, Frisoni GB, Blin O, Richardson JC, Bordet R, Molinuevo JL, Rami L, Wallin A, Kettunen P, Tsolaki M, Verhey F, Lleó A, Alcolea D, Popp J, Peyratout G, Martinez-Lage P, Tainta M, Johannsen P, Teunissen CE, Freund-Levi Y, Frölich L, Legido-Quigley C, Barkhof F, Blennow K, Rasmussen KL, Nordestgaard BG, Frikke-Schmidt R, Nielsen SF, Soininen H, Vellas B, Kloszewska I, Mecocci P, Zetterberg H, Morgan BP, Streffer J, Visser PJ, Bertram L, Nevado-Holgado AJ, Lovestone S. Dickkopf-1 Overexpression in vitro Nominates Candidate Blood Biomarkers Relating to Alzheimer's Disease Pathology. J Alzheimers Dis 2020; 77:1353-1368. [PMID: 32831200 PMCID: PMC7683080 DOI: 10.3233/jad-200208] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/15/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Previous studies suggest that Dickkopf-1 (DKK1), an inhibitor of Wnt signaling, plays a role in amyloid-induced toxicity and hence Alzheimer's disease (AD). However, the effect of DKK1 expression on protein expression, and whether such proteins are altered in disease, is unknown. OBJECTIVE We aim to test whether DKK1 induced protein signature obtained in vitro were associated with markers of AD pathology as used in the amyloid/tau/neurodegeneration (ATN) framework as well as with clinical outcomes. METHODS We first overexpressed DKK1 in HEK293A cells and quantified 1,128 proteins in cell lysates using aptamer capture arrays (SomaScan) to obtain a protein signature induced by DKK1. We then used the same assay to measure the DKK1-signature proteins in human plasma in two large cohorts, EMIF (n = 785) and ANM (n = 677). RESULTS We identified a 100-protein signature induced by DKK1 in vitro. Subsets of proteins, along with age and apolipoprotein E ɛ4 genotype distinguished amyloid pathology (A + T-N-, A+T+N-, A+T-N+, and A+T+N+) from no AD pathology (A-T-N-) with an area under the curve of 0.72, 0.81, 0.88, and 0.85, respectively. Furthermore, we found that some signature proteins (e.g., Complement C3 and albumin) were associated with cognitive score and AD diagnosis in both cohorts. CONCLUSIONS Our results add further evidence for a role of DKK regulation of Wnt signaling in AD and suggest that DKK1 induced signature proteins obtained in vitro could reflect theATNframework as well as predict disease severity and progression in vivo.
Collapse
Affiliation(s)
- Liu Shi
- Department of Psychiatry, University of Oxford, UK
| | | | | | - Richard Killick
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience Institute, London, UK
| | | | | | | | | | - Shengjun Hong
- Lübeck Interdisciplinary Platform for Genome Analytics, Institutes of Neurogenetics and Cardiogenetics, University of Lübeck, Lübeck, Germany
| | - Valerija Dobricic
- Lübeck Interdisciplinary Platform for Genome Analytics, Institutes of Neurogenetics and Cardiogenetics, University of Lübeck, Lübeck, Germany
| | - Fabian Kilpert
- Lübeck Interdisciplinary Platform for Genome Analytics, Institutes of Neurogenetics and Cardiogenetics, University of Lübeck, Lübeck, Germany
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Steven Kiddle
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience Institute, London, UK
- MRC Social, Genetic and Developmental Psychiatry Centre, King’s College London, UK
| | - Martina Sattlecker
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience Institute, London, UK
- MRC Social, Genetic and Developmental Psychiatry Centre, King’s College London, UK
| | - Richard Dobson
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
- Institute of Health Informatics, University College London, London, UK
| | - Antonio Cuadrado
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Investigación Sanitaria La Paz (IdiPaz), Instituto de Investigaciones Biomédicas Alberto Sols UAM-CSIC, and Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, Madrid, Spain
- ”Victor Babes” National Institute of Pathology, Bucharest, Romania
| | - Abdul Hye
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience Institute, London, UK
| | - Nicholas J. Ashton
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Institute Clinical Neuroscience Institute, London, UK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS Foundation, London, UK
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | | | - Isabelle Bos
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Centrum Limburg, Maastricht University, Maastricht, the Netherlands
- Alzheimer Center, VU University Medical Center, Amsterdam, the Netherlands
| | - Stephanie J.B. Vos
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Centrum Limburg, Maastricht University, Maastricht, the Netherlands
| | - Mara ten Kate
- Alzheimer Center, VU University Medical Center, Amsterdam, the Netherlands
| | - Philip Scheltens
- Alzheimer Center, VU University Medical Center, Amsterdam, the Netherlands
| | | | - Silvy Gabel
- University Hospital Leuven, Leuven, Belgium
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Belgium
| | - Karen Meersmans
- University Hospital Leuven, Leuven, Belgium
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven, Belgium
| | - Sebastiaan Engelborghs
- Center for Neurosciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
- Department of Neurology, UZ Brussel, Brussels, Belgium
| | - Ellen E. De Roeck
- Center for Neurosciences, Vrije Universiteit Brussel (VUB), Brussels, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Kristel Sleegers
- Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
- Neurodegenerative Brain Diseases Group, Center for Molecular Neurology, VIB, Belgium
| | - Giovanni B. Frisoni
- University of Geneva, Geneva, Switzerland
- IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Olivier Blin
- AIX Marseille University, INS, Ap-hm, Marseille, France
| | | | | | - José L. Molinuevo
- Alzheimer’s disease & other cognitive disorders unit, Hospital Clínic, Barcelona, Spain
- BarcelonaBeta Brain Research Center, Universitat Pompeu Fabra, Barcelona, Spain
| | - Lorena Rami
- BarcelonaBeta Brain Research Center, Universitat Pompeu Fabra, Barcelona, Spain
| | - Anders Wallin
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Memory Clinic at Department of Neuropsychiatry, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Petronella Kettunen
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Magda Tsolaki
- 1st Department of Neurology, AHEPA University Hospital, school of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Makedonia, Greece
| | - Frans Verhey
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Centrum Limburg, Maastricht University, Maastricht, the Netherlands
| | - Alberto Lleó
- Department of Neurology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Daniel Alcolea
- Department of Neurology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Julius Popp
- Department of Psychiatry, University Hospital of Lausanne, Lausanne, Switzerland
- Geriatric Psychiatry, Department of Psychiatry, Geneva University Hospitals, and University of Geneva, Geneva, Switzerland
| | - Gwendoline Peyratout
- Department of Psychiatry, University Hospital of Lausanne, Lausanne, Switzerland
| | | | - Mikel Tainta
- CITA-Alzheimer Foundation, San Sebastian, Spain
- Organización Sanitaria Integrada Goierri – Alto Urola, Osakidetza, Spain
| | - Peter Johannsen
- Danish Dementia Research Centre, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Charlotte E. Teunissen
- Neurochemistry Laboratory, dept of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam, the Netherlands
| | - Yvonne Freund-Levi
- School of Medical Sciences, Örebro University, Örebro, Sweden
- Department of Neurobiology, Care Sciences and Society, Division of Clinical Geriatrics, Karolinska Institute, Stockholm, Sweden
- Department of Old Age Psychiatry, Psychology and Neuroscience, King’s College London, UK
- Department of Psychiatry, Örebro Universitetssjukhus, Örebro, Sweden
| | - Lutz Frölich
- Department of Geriatric Psychiatry, Zentralinstitut für Seelische Gesundheit, University of Heidelberg, Mannheim, Germany
| | - Cristina Legido-Quigley
- Kings College London, London, UK
- The Systems Medicine Group, Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherland
- UCL Institutes of Neurology and Healthcare Engineering, London, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Katrine Laura Rasmussen
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Børge Grønne Nordestgaard
- The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
- The Copenhagen City Heart Study, Frederiksberg Hospital, Copenhagen University Hospital, Frederiksberg, Denmark
| | - Ruth Frikke-Schmidt
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sune Fallgaard Nielsen
- The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
| | - Hilkka Soininen
- Neurology / Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Bruno Vellas
- Toulouse Gerontopole University Hospital, Univeriste Paul Sabatier, INSERM U 558, France
| | | | - Patrizia Mecocci
- Section of Gerontology and Geriatrics, Department of Medicine, University of Perugia, Perugia, Italy
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- UK Dementia Research Institute at UCL, London, United Kingdom
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
| | - B. Paul Morgan
- Dementia Research Institute Cardiff, Cardiff University, Cardiff, UK
| | - Johannes Streffer
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- UCB, Braine-l’Alleud, Belgium, formerly Janssen R&D, LLC. Beerse, Belgium at the time of study conduct
| | - Pieter Jelle Visser
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Centrum Limburg, Maastricht University, Maastricht, the Netherlands
- Alzheimer Center, VU University Medical Center, Amsterdam, the Netherlands
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Lars Bertram
- Lübeck Interdisciplinary Platform for Genome Analytics, Institutes of Neurogenetics and Cardiogenetics, University of Lübeck, Lübeck, Germany
- Department of Psychology, University of Oslo, Oslo, Norway
| | | | - Simon Lovestone
- Department of Psychiatry, University of Oxford, UK
- Currently at Janssen-Cilag UK, formerly at Department of Psychiatry, University of Oxford, UK at the time of the study conduct
| |
Collapse
|
38
|
Carpanini SM, Torvell M, Morgan BP. Therapeutic Inhibition of the Complement System in Diseases of the Central Nervous System. Front Immunol 2019; 10:362. [PMID: 30886620 PMCID: PMC6409326 DOI: 10.3389/fimmu.2019.00362] [Citation(s) in RCA: 142] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 02/12/2019] [Indexed: 12/14/2022] Open
Abstract
The complement system plays critical roles in development, homeostasis, and regeneration in the central nervous system (CNS) throughout life; however, complement dysregulation in the CNS can lead to damage and disease. Complement proteins, regulators, and receptors are widely expressed throughout the CNS and, in many cases, are upregulated in disease. Genetic and epidemiological studies, cerebrospinal fluid (CSF) and plasma biomarker measurements and pathological analysis of post-mortem tissues have all implicated complement in multiple CNS diseases including multiple sclerosis (MS), neuromyelitis optica (NMO), neurotrauma, stroke, amyotrophic lateral sclerosis (ALS), Alzheimer's disease (AD), Parkinson's disease (PD), and Huntington's disease (HD). Given this body of evidence implicating complement in diverse brain diseases, manipulating complement in the brain is an attractive prospect; however, the blood-brain barrier (BBB), critical to protect the brain from potentially harmful agents in the circulation, is also impermeable to current complement-targeting therapeutics, making drug design much more challenging. For example, antibody therapeutics administered systemically are essentially excluded from the brain. Recent protocols have utilized "Trojan horse" techniques to transport therapeutics across the BBB or used osmotic shock or ultrasound to temporarily disrupt the BBB. Most research to date exploring the impact of complement inhibition on CNS diseases has been in animal models, and some of these studies have generated convincing data; for example, in models of MS, NMO, and stroke. There have been a few recent clinical trials of available anti-complement drugs in CNS diseases associated with BBB impairment, for example the use of the anti-C5 monoclonal antibody (mAb) eculizumab in NMO, but for most CNS diseases there have been no human trials of anti-complement therapies. Here we will review the evidence implicating complement in diverse CNS disorders, from acute, such as traumatic brain or spine injury, to chronic, including demyelinating, neuroinflammatory, and neurodegenerative diseases. We will discuss the particular problems of drug access into the CNS and explore ways in which anti-complement therapies might be tailored for CNS disease.
Collapse
Affiliation(s)
- Sarah M Carpanini
- UK Dementia Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Megan Torvell
- UK Dementia Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Bryan Paul Morgan
- UK Dementia Research Institute, Cardiff University, Cardiff, United Kingdom.,Division of Infection and Immunity, School of Medicine, Systems Immunity Research Institute, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|