51
|
Xiong X, Kuang H, Ansari S, Liu T, Gong J, Wang S, Zhao XY, Ji Y, Li C, Guo L, Zhou L, Chen Z, Leon-Mimila P, Chung MT, Kurabayashi K, Opp J, Campos-Pérez F, Villamil-Ramírez H, Canizales-Quinteros S, Lyons R, Lumeng CN, Zhou B, Qi L, Huertas-Vazquez A, Lusis AJ, Xu XZS, Li S, Yu Y, Li JZ, Lin JD. Landscape of Intercellular Crosstalk in Healthy and NASH Liver Revealed by Single-Cell Secretome Gene Analysis. Mol Cell 2020; 75:644-660.e5. [PMID: 31398325 DOI: 10.1016/j.molcel.2019.07.028] [Citation(s) in RCA: 454] [Impact Index Per Article: 113.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 04/15/2019] [Accepted: 07/19/2019] [Indexed: 12/27/2022]
Abstract
Cell-cell communication via ligand-receptor signaling is a fundamental feature of complex organs. Despite this, the global landscape of intercellular signaling in mammalian liver has not been elucidated. Here we perform single-cell RNA sequencing on non-parenchymal cells isolated from healthy and NASH mouse livers. Secretome gene analysis revealed a highly connected network of intrahepatic signaling and disruption of vascular signaling in NASH. We uncovered the emergence of NASH-associated macrophages (NAMs), which are marked by high expression of triggering receptors expressed on myeloid cells 2 (Trem2), as a feature of mouse and human NASH that is linked to disease severity and highly responsive to pharmacological and dietary interventions. Finally, hepatic stellate cells (HSCs) serve as a hub of intrahepatic signaling via HSC-derived stellakines and their responsiveness to vasoactive hormones. These results provide unprecedented insights into the landscape of intercellular crosstalk and reprogramming of liver cells in health and disease.
Collapse
Affiliation(s)
- Xuelian Xiong
- Ministry of Education Key Laboratory of Metabolism and Molecular Medicine, Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China; Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Henry Kuang
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Sahar Ansari
- Department of Human Genetics, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Tongyu Liu
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Jianke Gong
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Molecular and Integrative Physiology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA; International Research Center for Sensory Biology and Technology of MOST, Key Laboratory of Molecular Biophysics of MOE, and College of Life Science and Technology, and Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shuai Wang
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xu-Yun Zhao
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Yewei Ji
- Department of Molecular and Integrative Physiology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Chuan Li
- Department of Immunology, School of Medicine, University of Connecticut, Farmington, CT 06030, USA
| | - Liang Guo
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Linkang Zhou
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Zhimin Chen
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Paola Leon-Mimila
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, Los Angeles, CA, USA
| | - Meng Ting Chung
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48105, USA
| | - Katsuo Kurabayashi
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48105, USA
| | - Judy Opp
- University of Michigan DNA Sequencing Core, University of Michigan, Ann Arbor, MI 48109, USA
| | - Francisco Campos-Pérez
- Clínica Integral de Cirugía para la Obesidad y Enfermedades Metabólicas, Hospital General Dr. Rubén Lénero, Mexico City, Mexico
| | - Hugo Villamil-Ramírez
- Facultad de Química, UNAM/Instituto Nacional de Medicina Genómica (INMEGEN), Unidad de Genómica de Poblaciones Aplicada a la Salud, Mexico City, Mexico
| | - Samuel Canizales-Quinteros
- Facultad de Química, UNAM/Instituto Nacional de Medicina Genómica (INMEGEN), Unidad de Genómica de Poblaciones Aplicada a la Salud, Mexico City, Mexico
| | - Robert Lyons
- University of Michigan DNA Sequencing Core, University of Michigan, Ann Arbor, MI 48109, USA
| | - Carey N Lumeng
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Beiyan Zhou
- Department of Immunology, School of Medicine, University of Connecticut, Farmington, CT 06030, USA
| | - Ling Qi
- Department of Molecular and Integrative Physiology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Adriana Huertas-Vazquez
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, Los Angeles, CA, USA
| | - Aldons J Lusis
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, Los Angeles, CA, USA
| | - X Z Shawn Xu
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Molecular and Integrative Physiology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Siming Li
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Yonghao Yu
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jun Z Li
- Department of Human Genetics, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Jiandie D Lin
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA.
| |
Collapse
|
52
|
Zhang B, Li R, Zhang Y, Gao X. Differential role of triggering receptors expressed on myeloid cells 2 R47H in 3 neurodegenerative diseases based on a systematic review and meta-analysis. Medicine (Baltimore) 2020; 99:e18921. [PMID: 32000403 PMCID: PMC7004756 DOI: 10.1097/md.0000000000018921] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Recent studies have suggested that the potential functional polymorphism R47H in triggering receptors expressed on myeloid cells 2 (TREM2) is associated with several neurodegenerative diseases, however, the results remain inconclusive. This meta-analysis aimed to investigate the association between TREM2 R47H and the risk for 3 typical neurodegenerative diseases: Alzheimer disease (AD), Parkinson disease (PD), and amyotrophic lateral sclerosis (ALS). METHODS A literature review was carried out using PubMed, Medline, and Embase. Data analysis was conducted using Stata 15.0 software. The pooled odds ratio (ORs) and 95% confidence interval (CIs) were calculated. RESULTS A total of 35 articles were identified as eligible: 22 on AD, 3 on ALS, 7 on PD, 2 on AD and ALS, and 1 on ALS and PD. The AD set included 23,092 cases and 30,920 controls, the ALS set included 7391 cases and 12,442 controls, and the PD set included 8498 patients and 9161 controls. We found that R47H was associated with an increased risk of AD in the total pooled population (P < .001, OR = 4.02, 95% CI = 3.15-5.13). However, this significant difference existed for Caucasian people (OR = 4.16, 95% CI = 3.24-5.33) but not for Asian or African people. Moreover, we did not find any significant differences in minor allele frequency distribution between the PD and control groups or between the ALS and control groups, not only for the total pooled population but also for the subgroups of different ethnicities. CONCLUSION Our study suggested that R47H in the TREM2 gene leads to an increased risk for developing AD, but not for ALS and PD, which adds evidence to the notion that diverse pathogenesis may be involved in different neurogenerative diseases.
Collapse
Affiliation(s)
- Bin Zhang
- Department of Neurology, the First Hospital of Yulin, Yulin, Shaanxi
| | - Rui Li
- Department of Neurology, the First Hospital of Yulin, Yulin, Shaanxi
| | - Yufan Zhang
- Department of Neurology, the First Hospital of Yulin, Yulin, Shaanxi
| | - Xia Gao
- Department of Geriatrics, Dazhou Central Hospital, Dazhou, Sichuan, China
| |
Collapse
|
53
|
Olive C, Ibanez L, Farias FHG, Wang F, Budde JP, Norton JB, Gentsch J, Morris JC, Li Z, Dube U, Del-Aguila J, Bergmann K, Bradley J, Benitez BA, Harari O, Fagan A, Ances B, Cruchaga C, Fernandez MV. Examination of the Effect of Rare Variants in TREM2, ABI3, and PLCG2 in LOAD Through Multiple Phenotypes. J Alzheimers Dis 2020; 77:1469-1482. [PMID: 32894242 PMCID: PMC7927150 DOI: 10.3233/jad-200019] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND Rare variants in PLCG2 (p.P522R), ABI3 (p.S209F), and TREM2 (p.R47H, p.R62H) have been associated with late onset Alzheimer's disease (LOAD) risk in Caucasians. After the initial report, several studies have found positive results in cohorts of different ethnic background and with different phenotype. OBJECTIVE In this study, we aim to evaluate the association of rare coding variants in PLCG2, ABI3, and TREM2 with LOAD risk and their effect at different time points of the disease. METHODS We used a European American cohort to assess the association of the variants prior onset (using CSF Aβ42, tau, and pTau levels, and amyloid imaging as endophenotypes) and after onset (measured as rate of memory decline). RESULTS We confirm the association with LOAD risk of TREM2 p.R47H, p.R62H and ABI3 p.S209F variants, and the protective effect of PLCG2 p.P522R. In addition, ABI3 and TREM2 gene-sets showed significant association with LOAD risk. TREM2 p.R47H and PLCG2 p.P522R variants were also statistically associated with increase of amyloid imaging and AD progression, respectively. We did not observe any association of ABI3 p.S209F with any of the other AD endophenotypes. CONCLUSION The results of this study highlight the importance of including biomarkers and alternative phenotypes to better understand the role of novel candidate genes with the disease.
Collapse
Affiliation(s)
- Claudia Olive
- Neurogenomics and Informatics Center, Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Laura Ibanez
- Neurogenomics and Informatics Center, Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Fabiana H. Geraldo Farias
- Neurogenomics and Informatics Center, Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Fengxian Wang
- Neurogenomics and Informatics Center, Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - John P. Budde
- Neurogenomics and Informatics Center, Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Joanne B. Norton
- Neurogenomics and Informatics Center, Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Jen Gentsch
- Neurogenomics and Informatics Center, Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - John C. Morris
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
- Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Zeran Li
- Neurogenomics and Informatics Center, Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Umber Dube
- Neurogenomics and Informatics Center, Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Jorge Del-Aguila
- Neurogenomics and Informatics Center, Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Kristy Bergmann
- Neurogenomics and Informatics Center, Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Joseph Bradley
- Neurogenomics and Informatics Center, Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Bruno A. Benitez
- Neurogenomics and Informatics Center, Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Oscar Harari
- Neurogenomics and Informatics Center, Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Anne Fagan
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Beau Ances
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Carlos Cruchaga
- Neurogenomics and Informatics Center, Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| | - Maria Victoria Fernandez
- Neurogenomics and Informatics Center, Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
54
|
Peng G, Qiu J, Liu H, Zhou M, Huang S, Guo W, Lin Y, Chen X, Li Z, Li G, Zhang W, Zhang Y, Li X, Wu Z, Wei L, Yang X, Zhu X, Mo M, Xu P. Analysis of Cerebrospinal Fluid Soluble TREM2 and Polymorphisms in Sporadic Parkinson's Disease in a Chinese Population. J Mol Neurosci 2019; 70:294-301. [PMID: 31833018 DOI: 10.1007/s12031-019-01424-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Accepted: 10/18/2019] [Indexed: 01/20/2023]
Abstract
BACKGROUND Triggering receptor expressed on myeloid cells 2 (TREM2) is a microglial surface receptor that mediates the degradation disorder of amyloid β (Aβ) in Alzheimer's disease. However, the role of TREM2 in Parkinson's disease (PD) and α-Synclein (α-Syn) degradation is largely unknown. METHODS In this case-control study on Chinese population, we sequenced for polymorphisms in exon 2 of the TREM2 gene in 1,292 individuals, PD cases (n = 612), healthy controls (n = 680) by Sanger sequence, and compared the distribution of allelic frequencies between the two groups by the Fisher's exact test. Additionally, we developed and used the enzyme-linked immunosorbent assay to evaluated soluble TREM2 (sTREM2) levels in the cerebrospinal fluid (CSF), and plasma in partial of sequenced groups (55 PD and 40 healthy controls) analyzed their relationship with total a-syn (t-a-Syn). RESULTS Two novel variants were detected in exon 2 of the TREM2 gene, namely, p.S81 N, p.G58D; however, these were not significantly associated with PD (612 PD and 680 healthy controls). sTREM2 in CSF was significantly upregulated in PD patients compared to healthy controls (433.1 ± 24.7 pg/mL vs. 275.2 ± 17.9 pg/mL, p < 0.0001), but not in plasma (281.7 ± 29.3 pg/mL vs. 257.8 ± 16.5 pg/mL, p = 0.805). In PD patients, sTREM2 was positively correlated with t-α-syn (r = 0.62, p = 0.0001) in CSF, but not in plasma (r = 0.02, p = 0.89). CONCLUSIONS Although it may not indicate that exon 2 polymorphisms of TREM2 play a role in the pathogenesis of PD in the Chinese population, our findings described above highlight the relevance of CSF sTREM2 as a promising biomarker and are extremely possible to the therapeutic target for PD in the future.
Collapse
Affiliation(s)
- Guoyou Peng
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China
| | - Jiewen Qiu
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China
| | - Hanqun Liu
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China
| | - Miaomiao Zhou
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China
| | - Shuxuan Huang
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China
| | - Wenyuan Guo
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China
| | - Yuwan Lin
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China
| | - Xiang Chen
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China
| | - Zhe Li
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China
| | - Guihua Li
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China
| | - Wenlong Zhang
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China
| | - Yunlong Zhang
- Department of Physiology, Guangzhou Medical University, Guangzhou, 511436, China.,Neuroscience Center, Guangzhou Medical University, Guangzhou, 511436, China
| | - Xingjian Li
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China
| | - Zhuohua Wu
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China
| | - Lei Wei
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Xinling Yang
- Department of Neurology, The Second Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, China
| | - Xiaoqin Zhu
- Department of Physiology, Guangzhou Medical University, Guangzhou, 511436, China. .,Neuroscience Center, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Mingshu Mo
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China.
| | - Pingyi Xu
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China. .,Neuroscience Center, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
55
|
Dean HB, Roberson ED, Song Y. Neurodegenerative Disease-Associated Variants in TREM2 Destabilize the Apical Ligand-Binding Region of the Immunoglobulin Domain. Front Neurol 2019; 10:1252. [PMID: 32021611 PMCID: PMC6985895 DOI: 10.3389/fneur.2019.01252] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 11/11/2019] [Indexed: 01/22/2023] Open
Abstract
Single nucleotide variations in Triggering Receptor Expressed on Myeloid Cells 2 (TREM2) have been linked to both late-onset Alzheimer's disease and behavioral variant frontotemporal dementia (FTD), the latter presenting either in isolation or with cystic bone lesions in a condition called Nasu-Hakola disease. Models of the extracellular domain of TREM2 show that Nasu-Hakola disease-associated mutations are grossly inactivating by truncation, frameshift, or unfolding, that Alzheimer's disease (AD)-associated variants localize to a putative ligand-interacting region (PLIR) on the extracellular surface, and that FTD-associated variants are found in the hydrophobic core. However, while these disease-associated residues are predicted to play some role in disrupting ligand binding to the extracellular domain of TREM2, how they ultimately lead to disease remains unknown. Here, we used in silico molecular modeling to investigate all-atom models of TREM2 and characterize the effects on conformation and dynamical motion of AD-associated R47H and R62H as well as FTD-associated T96K, D86V, and T66M variants compared to the benign N68K variant and the common variant. Our model, which is based on a published 2.2 Å resolution crystal structure of the TREM2 extracellular domain, finds that both AD- and FTD-associated variants cause localized instability in three loops adjacent to the PLIR that correspond to the complementarity-determining regions (CDRs) of antibodies. This instability ultimately disrupts tethering between these CDRs and the core of the immunoglobulin domain, exposing a group of otherwise-buried, negatively charged residues. This instability and exposure of negatively charged residues is most severe following introduction of the T66M variant that has been described as causing FTD even in the heterozygous state and is less severe following introduction of variants that are less strongly tied to FTD or of those associated with AD. Thus, our results provide further evidence that the proposed loss-of-function caused by neurodegenerative disease-associated variants may be driven by altered conformational stability of the ligand-interacting CDR and, ultimately, loss of affinity or specificity for TREM2 ligands.
Collapse
Affiliation(s)
- Hunter B Dean
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States.,Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, United States.,Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, United States.,Alzheimer's Disease Center, University of Alabama at Birmingham, Birmingham, AL, United States.,Medical Scientist Training Program, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Erik D Roberson
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, United States.,Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, United States.,Alzheimer's Disease Center, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Yuhua Song
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
56
|
Schneider SA, Tahirovic S, Hardy J, Strupp M, Bremova-Ertl T. Do heterozygous mutations of Niemann-Pick type C predispose to late-onset neurodegeneration: a review of the literature. J Neurol 2019; 268:2055-2064. [PMID: 31701332 DOI: 10.1007/s00415-019-09621-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 10/30/2019] [Accepted: 11/02/2019] [Indexed: 12/28/2022]
Abstract
BACKGROUND/METHODS Monogenic diseases are important models for the study of neurodegenerative diseases, such as Parkinson's disease (PD) and dementia. Notably, for some disorders, homozygosity is associated with a complex metabolic disease, while heterozygosity predisposes to late-onset neurodegeneration. For instance, biallelic glucocerebrosidase gene mutations cause Gaucher's disease, while heterozygous mutations are a common genetic risk factor for late-onset PD. Little is known about similar risks of related diseases, such as Niemann-Pick type C (NPC). Given that both conditions map into related, i.e., lysosomal, pathways, we hypothesize a similar risk of single-NPC gene mutations. Indeed, there is increasing evidence based on clinical observations in humans and animal studies. Here we review the current knowledge of NPC heterozygosity. RESULTS Family history studies suggest a high proportion of late-onset neurodegenerative diseases in NPC families. We identified 19 cases with heterozygous NPC mutations in the literature who presented with a neurodegenerative disease, including levodopa-responsive PD, atypical parkinsonism (PSP, CBD), dystonia or dementia with a mean age at onset of about 57 years (range 8-87). Consistent splenomegaly and mildly abnormal filipin staining results have also been reported in heterozygous gene mutation carriers. Imaging and pathological data support this notion. DISCUSSION/CONCLUSION This finding has wider implications in so far as NPC-related forms of Parkinsonian syndromes, dementia, motor neuron disease and other neurodegenerative disorders may benefit from NPC-mechanistic therapies, in particular related to lysosomal dysfunction. Further research is warranted to generate systematic data of heterozygous mutation carriers, including longitudinal data.
Collapse
Affiliation(s)
- Susanne A Schneider
- Department of Neurology, Ludwig-Maximilians-University, Marchioninistr 15, 81377, Munich, Germany.
| | - Sabina Tahirovic
- German Center for Neurodegenerative Diseases (DZNE) Within the Helmholtz Association, Feodor-Lynen-Strasse 17, Munich, Germany
| | - John Hardy
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | - Michael Strupp
- Department of Neurology, Ludwig-Maximilians-University, Marchioninistr 15, 81377, Munich, Germany
| | - Tatiana Bremova-Ertl
- Department of Neurology, Ludwig-Maximilians-University, Marchioninistr 15, 81377, Munich, Germany.,Department of Neurology, Inselspital, University Hospital Bern, Bern, Switzerland
| |
Collapse
|
57
|
Karsak M, Glebov K, Scheffold M, Bajaj T, Kawalia A, Karaca I, Rading S, Kornhuber J, Peters O, Diez-Fairen M, Frölich L, Hüll M, Wiltfang J, Scherer M, Riedel-Heller S, Schneider A, Heneka MT, Fliessbach K, Sharaf A, Thiele H, Lennarz M, Jessen F, Maier W, Kubisch C, Ignatova Z, Nürnberg P, Pastor P, Walter J, Ramirez A. A rare heterozygous TREM2 coding variant identified in familial clustering of dementia affects an intrinsically disordered protein region and function of TREM2. Hum Mutat 2019; 41:169-181. [PMID: 31464095 DOI: 10.1002/humu.23904] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 08/12/2019] [Accepted: 08/25/2019] [Indexed: 11/11/2022]
Abstract
Rare coding variants in the triggering receptor expressed on myeloid cells-2 (TREM2) gene have been associated with Alzheimer disease (AD) and homozygous TREM2 loss-of-function variants have been reported in families with monogenic frontotemporal-like dementia with/without bone abnormalities. In a whole-exome sequencing study of a family with probable AD-type dementia without pathogenic variants in known autosomal dominant dementia disease genes and negative for the apolipoprotein E (APOE) ε4 allele, we identified an extremely rare TREM2 coding variant, that is, a glycine-to-tryptophan substitution at amino acid position 145 (NM_018965.3:c.433G>T/p.[Gly145Trp]). This alteration is found in only 1 of 251,150 control alleles in gnomAD. It was present in both severely affected as well as in another putatively affected and one 61 years old as yet unaffected family member suggesting incomplete penetrance and/or a variable age of onset. Gly145 maps to an intrinsically disordered region (IDR) of TREM2 between the immunoglobulin-like and transmembrane domain. Subsequent cellular studies showed that the variant led to IDR shortening and structural changes of the mutant protein resulting in an impairment of cellular responses upon receptor activation. Our results, suggest that a p.(Gly145Trp)-induced structural disturbance and functional impairment of TREM2 may contribute to the pathogenesis of an AD-like form of dementia.
Collapse
Affiliation(s)
- Meliha Karsak
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | | | - Marina Scheffold
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.,Institute of Pharmacology and Toxicology, University of Ulm, Ulm, Germany
| | - Thomas Bajaj
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Cologne, Cologne, Germany
| | - Amit Kawalia
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Cologne, Cologne, Germany
| | - Ilker Karaca
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn, Bonn, Germany
| | - Sebastian Rading
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen and Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Oliver Peters
- Department of Psychiatry, Charité University Medicine, Berlin, Germany
| | - Monica Diez-Fairen
- Department of Neurology, Memory and Movement Disorders Units, University Hospital Mutua de Terrassa, Terrassa, Barcelona, Spain.,Fundació Docència i Recerca Mútua Terrassa, University Hospital Mútua de Terrassa, Terrassa, Barcelona, Spain
| | - Lutz Frölich
- Department of Geriatric Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Michael Hüll
- Center for Psychiatry, Clinic for Geriatric Psychiatry and Psychotherapy Emmendingen and Department of Psychiatry and Psychotherapy, University of Freiburg, Freiburg, Germany
| | - Jens Wiltfang
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany.,German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Martin Scherer
- Department of Primary Medical Care, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Steffi Riedel-Heller
- Institute of Social Medicine, Occupational Health and Public Health, University of Leipzig, Leipzig, Germany
| | - Anja Schneider
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn, Bonn, Germany.,German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Michael T Heneka
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn, Bonn, Germany.,German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Klaus Fliessbach
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn, Bonn, Germany
| | - Ahmed Sharaf
- Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Holger Thiele
- Cologne Center for Genomics (CCG), University of Cologne, Cologne, Germany
| | - Martina Lennarz
- Department of Psychiatry and Psychotherapy, University of Bonn, Bonn, Germany
| | - Frank Jessen
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Department of Psychiatry and Psychotherapy, Medical Faculty, University of Cologne, Cologne, Germany
| | - Wolfgang Maier
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn, Bonn, Germany.,German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Christian Kubisch
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Zoya Ignatova
- Institute for Biochemistry and Molecular Biology, University of Hamburg, Hamburg, Germany
| | - Peter Nürnberg
- Cologne Center for Genomics (CCG), University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Pau Pastor
- Department of Neurology, Memory and Movement Disorders Units, University Hospital Mutua de Terrassa, Terrassa, Barcelona, Spain.,Fundació Docència i Recerca Mútua Terrassa, University Hospital Mútua de Terrassa, Terrassa, Barcelona, Spain
| | - Jochen Walter
- Department of Neurology, University of Bonn, Bonn, Germany
| | - Alfredo Ramirez
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Cologne, Cologne, Germany.,Department of Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn, Bonn, Germany
| |
Collapse
|
58
|
Redaelli V, Salsano E, Colleoni L, Corbetta P, Tringali G, Del Sole A, Giaccone G, Rossi G. Frontotemporal Dementia and Chorea Associated with a Compound Heterozygous TREM2 Mutation. J Alzheimers Dis 2019; 63:195-201. [PMID: 29578490 DOI: 10.3233/jad-180018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Frontotemporal dementia (FTD) is clinically characterized by behavioral changes, language impairment, and executive dysfunction. FTD usually belongs to the frontotemporal lobar degeneration (FTLD) disease group, and its familial forms are dominantly inherited and linked to a group of genes relevant to frontal and temporal brain pathology, such as MAPT, GRN, C9ORF72, TARDBP, CHMP2B, VCP, and FUS. However, FTD can also be associated with different clinical or pathological phenotypes caused by mutations in other genes, whose heredity can be dominant or recessive. In this work we report on a familial case of FTD characterized by behavioral changes and aphasia, very early onset and very long duration, choreic movements, and white matter lesions at magnetic resonance imaging. We performed a wide-range genetic analysis, using a next generation sequencing approach, to evaluate a number of genes involved in neurodegeneration. We found a previously unreported compound heterozygous mutation in TREM2, that is commonly associated with the recessively inherited Nasu-Hakola disease. We discuss the differential diagnosis to be taken into account in cases of FTD presenting with atypical features.
Collapse
Affiliation(s)
- Veronica Redaelli
- Unit of Neurology V and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Ettore Salsano
- Unit of Neurodegenerative and Neurometabolic Rare Diseases, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Lara Colleoni
- Unit of Neurology V and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Paola Corbetta
- Unit of Neurology V and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Giovanni Tringali
- Unit of Neurosurgery III, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Angelo Del Sole
- Department of Diagnostic Services, Unit of Nuclear Medicine, ASST Santi Paolo e Carlo, Milan, Italy
| | - Giorgio Giaccone
- Unit of Neurology V and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| | - Giacomina Rossi
- Unit of Neurology V and Neuropathology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milano, Italy
| |
Collapse
|
59
|
Tian ML, Ni XN, Li JQ, Tan CC, Cao XP, Tan L. A Candidate Regulatory Variant at the TREM Gene Cluster Confer Alzheimer's Disease Risk by Modulating Both Amyloid-β Pathology and Neuronal Degeneration. Front Neurosci 2019; 13:742. [PMID: 31379492 PMCID: PMC6650562 DOI: 10.3389/fnins.2019.00742] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 07/04/2019] [Indexed: 11/13/2022] Open
Abstract
Background: rs9357347 located at the triggering receptor expressed on myeloid cells (TREM) gene cluster could increase TREM2 and TREM-like transcript 1 (TREML1) brain gene expression, which is considered to play a protective role against Alzheimer's disease (AD). Objectives: To investigate the role of rs9357347 in AD pathogenesis by exploring the effects of rs9357347 on AD specific biomarkers. Methods: This study analyzed the association of rs9357347 with AD-related cerebrospinal fluid (CSF) and neuroimaging markers from 201 cognitively normal (CN) older adults, 349 elders with mild cognitive impairment (MCI), and 172 elders with AD dementia from the Alzheimer's Disease Neuroimaging Initiative (ADNI). We next analyzed the association in 259 amyloid-β positive (Aβ+) elders and 117 amyloid-β negative (Aβ-) elders (Aβ+: CSF Aβ1-42 ≤ 192 pg/ml; Aβ-: CSF Aβ1-42 > 192 pg/ml). Associations were tested using multiple linear regression models at baseline. Furthermore, multiple mixed-effects models were used in a longitudinal study which lasted 4 years. Results: At baseline, we found that rs9357347 had association with CSF Aβ1-42 in CN group (β = 0.357, P = 0.009). In AD group, rs9357347 was associated with total tau (T-tau) level (β = -0.436, P = 0.007). Moreover, the strong influence exerted by rs9357347 on T-tau was also seen in Aβ+ group (β = -0.202, P = 0.036). In the longitudinal study, rs9357347 was also found to be associated with Aβ1-42 in CN group (β = 0.329, P = 0.023). In AD group, the mutation of rs9357347 was associated with slower accumulation of T-tau (β = -0.472, P = 0.002) and tau phosphorylated at threonine 181 [P-tau 181 (β = -0.330, P = 0.019)]. Furthermore, the obvious influence exerted by rs9357347 on T-tau was also seen in Aβ+ group (β = -0.241, P = 0.013). Conclusion: This study suggested that rs9357347 reduced the risk of AD by modulating both amyloid-β pathology and neuronal degeneration.
Collapse
Affiliation(s)
- Mei-Ling Tian
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Xiao-Neng Ni
- Department of Neurology, Qingdao Municipal Hospital, Nanjing Medical University, Nanjing, China
| | - Jie-Qiong Li
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Chen-Chen Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Xi-Peng Cao
- Clinical Research Center, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Nanjing Medical University, Nanjing, China
- Clinical Research Center, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | | |
Collapse
|
60
|
Szatmari B, Balicza P, Nemeth G, Molnar MJ. The Panomics Approach in Neurodegenerative Disorders. Curr Med Chem 2019; 26:1712-1720. [PMID: 28685677 DOI: 10.2174/0929867324666170705120038] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 05/15/2017] [Accepted: 05/15/2017] [Indexed: 12/20/2022]
Abstract
BACKGROUND The molecular genetic technologies revolutionized the diagnostics of many disorders. Thanks to the new molecular techniques and the rapid improvement of the information technologies the number of mendelien inherited disorders has increased rapidly in the last five years. The omics era brought radical changes in the understanding of complex disorders and the underlying pathomechanisms. However, in most complex disorders the genome wide association studies could not clarify the genetic background even for disorders where a very strong heritability had been observed. OBJECTIVE In this paper the changing concept of the neurodegenerative disorders is discussed. The traditional classification of these disorders was purely based on clinical symptoms and morphological signs in the last century. Identifying the signature lesions of various neurodegenerative disorders may reveal a common pathological pathway in these disorders. New neuroimaging methods provided additional tools to assess pathological pathways in vivo already in the early stages of the diseases. Visualizing in vivo amyloid deposits and neuroinflammation improved our understanding of their role in various neurodegenerative disorders. Genetics may be the most precise way to identify the background of these disorders. However, there is only limited number of cases where true association can be proved between the disorder and the genetic mutations. Most of the neurodegenerative disorders seem to be multifactorial and cannot be traced back to one single cause. CONCLUSION In conclusion, shifting from a classification based on symptomatology only to a modern multidisciplinary approach, based on the constantly evolving panomics findings, would improve our understanding of neurodegenerative diseases and could be the basis of novel therapeutic research.
Collapse
Affiliation(s)
| | - Peter Balicza
- Institute of Genomic Medicine and Rare Disorders, Semmelweis University, Budapest, Hungary
| | - Gyorgy Nemeth
- Medical Division, Gedeon Richter Plc., Budapest, Hungary
| | - Maria Judit Molnar
- Institute of Genomic Medicine and Rare Disorders, Semmelweis University, Budapest, Hungary
| |
Collapse
|
61
|
An update on the interactions between Alzheimer's disease, autophagy and inflammation. Gene 2019; 705:157-166. [DOI: 10.1016/j.gene.2019.04.040] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 04/14/2019] [Accepted: 04/15/2019] [Indexed: 12/20/2022]
|
62
|
Lessard CB, Malnik SL, Zhou Y, Ladd TB, Cruz PE, Ran Y, Mahan TE, Chakrabaty P, Holtzman DM, Ulrich JD, Colonna M, Golde TE. High-affinity interactions and signal transduction between Aβ oligomers and TREM2. EMBO Mol Med 2019; 10:emmm.201809027. [PMID: 30341064 PMCID: PMC6220267 DOI: 10.15252/emmm.201809027] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Rare coding variants in the triggering receptor expressed on myeloid cells 2 (TREM2) are associated with increased risk for Alzheimer's disease (AD), but how they confer this risk remains uncertain. We assessed binding of TREM2, AD‐associated TREM2 variants to various forms of Aβ and APOE in multiple assays. TREM2 interacts directly with various forms of Aβ, with highest affinity interactions observed between TREM2 and soluble Aβ42 oligomers. High‐affinity binding of TREM2 to Aβ oligomers is characterized by very slow dissociation. Pre‐incubation with Aβ is shown to block the interaction of APOE. In cellular assays, AD‐associated variants of TREM2 reduced the amount of Aβ42 internalized, and in NFAT assay, the R47H and R62H variants decreased NFAT signaling activity in response to Aβ42. These studies demonstrate i) a high‐affinity interaction between TREM2 and Aβ oligomers that can block interaction with another TREM2 ligand and ii) that AD‐associated TREM2 variants bind Aβ with equivalent affinity but show loss of function in terms of signaling and Aβ internalization.
Collapse
Affiliation(s)
- Christian B Lessard
- Center for Translational Research in Neurodegenerative Disease, Department of Neuroscience, University of Florida, Gainesville, FL, USA
| | - Samuel L Malnik
- Center for Translational Research in Neurodegenerative Disease, Department of Neuroscience, University of Florida, Gainesville, FL, USA
| | - Yingyue Zhou
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Thomas B Ladd
- Center for Translational Research in Neurodegenerative Disease, Department of Neuroscience, University of Florida, Gainesville, FL, USA
| | - Pedro E Cruz
- Center for Translational Research in Neurodegenerative Disease, Department of Neuroscience, University of Florida, Gainesville, FL, USA
| | - Yong Ran
- Center for Translational Research in Neurodegenerative Disease, Department of Neuroscience, University of Florida, Gainesville, FL, USA
| | - Thomas E Mahan
- Department of Neurology, Hope Center for Neurological Disorders, Knight ADRC, Washington University School of Medicine, St. Louis, MO, USA
| | - Paramita Chakrabaty
- Center for Translational Research in Neurodegenerative Disease, Department of Neuroscience, University of Florida, Gainesville, FL, USA.,McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Knight ADRC, Washington University School of Medicine, St. Louis, MO, USA
| | - Jason D Ulrich
- Department of Neurology, Hope Center for Neurological Disorders, Knight ADRC, Washington University School of Medicine, St. Louis, MO, USA
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Todd E Golde
- Center for Translational Research in Neurodegenerative Disease, Department of Neuroscience, University of Florida, Gainesville, FL, USA .,McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| |
Collapse
|
63
|
Microglia-neuron crosstalk: Signaling mechanism and control of synaptic transmission. Semin Cell Dev Biol 2019; 94:138-151. [PMID: 31112798 DOI: 10.1016/j.semcdb.2019.05.017] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 04/17/2019] [Accepted: 05/16/2019] [Indexed: 12/13/2022]
Abstract
The continuous crosstalk between microglia and neurons is required for microglia housekeeping functions and contributes to brain homeostasis. Through these exchanges, microglia take part in crucial brain functions, including development and plasticity. The alteration of neuron-microglia communication contributes to brain disease states with consequences, ranging from synaptic function to neuronal survival. This review focuses on the signaling pathways responsible for neuron-microglia crosstalk, highlighting their physiological roles and their alteration or specific involvement in disease. In particular, we discuss studies, establishing how these signaling allow microglial cells to control relevant physiological functions during brain development, including synaptic formation and circuit refinement. In addition, we highlight how microglia and neurons interact functionally to regulate highly dynamical synaptic functions. Microglia are able to release several signaling molecules involved in the regulation of synaptic activity and plasticity. On the other side, molecules of neuronal origin control microglial processes motility in an activity-dependent manner. Indeed, the continuous crosstalk between microglia and neurons is required for the sensing and housekeeping functions of microglia and contributes to the maintenance of brain homeostasis and, particularly, to the sculpting of neuronal connections during development. These interactions lay on the delicate edge between physiological processes and homeostasis alteration in pathology and are themselves altered during neuroinflammation. The full description of these processes could be fundamental for understanding brain functioning in health and disease.
Collapse
|
64
|
Del-Aguila JL, Benitez BA, Li Z, Dube U, Mihindukulasuriya KA, Budde JP, Farias FHG, Fernández MV, Ibanez L, Jiang S, Perrin RJ, Cairns NJ, Morris JC, Harari O, Cruchaga C. TREM2 brain transcript-specific studies in AD and TREM2 mutation carriers. Mol Neurodegener 2019; 14:18. [PMID: 31068200 PMCID: PMC6505298 DOI: 10.1186/s13024-019-0319-3] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 04/26/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Low frequency coding variants in TREM2 are associated with Alzheimer disease (AD) risk and cerebrospinal fluid (CSF) TREM2 protein levels are different between AD cases and controls. Similarly, TREM2 risk variant carriers also exhibit differential CSF TREM2 levels. TREM2 has three different alternative transcripts, but most of the functional studies only model the longest transcript. No studies have analyzed TREM2 expression levels or alternative splicing in brains from AD and cognitively normal individuals. We wanted to determine whether there was differential expression of TREM2 in sporadic-AD cases versus AD-TREM2 carriers vs sex- and aged-matched normal controls; and if this differential expression was due to a particular TREM2 transcript. METHODS We analyzed RNA-Seq data from parietal lobe brain tissue from AD cases with TREM2 variants (n = 33), AD cases (n = 195) and healthy controls (n = 118), from three independent datasets using Kallisto and the R package tximport to determine the read count for each transcript and quantified transcript abundance as transcripts per million. RESULTS The three TREM2 transcripts were expressed in brain cortex in the three datasets. We demonstrate for the first time that the transcript that lacks the transmembrane domain and encodes a soluble form of TREM2 (sTREM2) has an expression level around 60% of the canonical transcript, suggesting that around 25% of the sTREM2 protein levels could be explained by this transcript. We did not observe a difference in the overall TREM2 expression level between cases and controls. However, the isoform which lacks the 5' exon, but includes the transmembrane domain, was significantly lower in TREM2- p.R62H carriers than in AD cases (p = 0.007). CONCLUSION Using bulk RNA-Seq data from three different cohorts, we were able to quantify the expression level of the three TREM2 transcripts, demonstrating: (1) all three transcripts of them are highly expressed in the human cortex, (2) that up to 25% of the sTREM2 may be due to the expression of a specific isoform and not TREM2 cleavage; and (3) that TREM2 risk variants do not affect expression levels, suggesting that the effect of the TREM2 variants on CSF levels occurs at post-transcriptional level.
Collapse
Affiliation(s)
- Jorge L. Del-Aguila
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO USA
- NeuroGenomics and Informatics, Washington University School of Medicine, St. Louis, MO USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO USA
| | - Bruno A. Benitez
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO USA
- NeuroGenomics and Informatics, Washington University School of Medicine, St. Louis, MO USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO USA
| | - Zeran Li
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO USA
- NeuroGenomics and Informatics, Washington University School of Medicine, St. Louis, MO USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO USA
| | - Umber Dube
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO USA
- NeuroGenomics and Informatics, Washington University School of Medicine, St. Louis, MO USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO USA
| | - Kathie A. Mihindukulasuriya
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO USA
- NeuroGenomics and Informatics, Washington University School of Medicine, St. Louis, MO USA
| | - John P. Budde
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO USA
- NeuroGenomics and Informatics, Washington University School of Medicine, St. Louis, MO USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO USA
| | - Fabiana H. G. Farias
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO USA
- NeuroGenomics and Informatics, Washington University School of Medicine, St. Louis, MO USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO USA
| | - Maria Victoria Fernández
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO USA
- NeuroGenomics and Informatics, Washington University School of Medicine, St. Louis, MO USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO USA
| | - Laura Ibanez
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO USA
- NeuroGenomics and Informatics, Washington University School of Medicine, St. Louis, MO USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO USA
| | - Shan Jiang
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO USA
| | - Richard J. Perrin
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO USA
- Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO USA
| | - Nigel J. Cairns
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO USA
- Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO USA
| | - John C. Morris
- Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO USA
- Department of Neurology, Washington University School of Medicine, St. Louis, MO USA
| | - Oscar Harari
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO USA
- NeuroGenomics and Informatics, Washington University School of Medicine, St. Louis, MO USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO USA
- Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO USA
- NeuroGenomics and Informatics, Washington University School of Medicine, St. Louis, MO USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO USA
- Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO USA
| |
Collapse
|
65
|
McQuade A, Blurton-Jones M. Microglia in Alzheimer's Disease: Exploring How Genetics and Phenotype Influence Risk. J Mol Biol 2019; 431:1805-1817. [PMID: 30738892 PMCID: PMC6475606 DOI: 10.1016/j.jmb.2019.01.045] [Citation(s) in RCA: 134] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 01/29/2019] [Accepted: 01/29/2019] [Indexed: 01/25/2023]
Abstract
Research into the function of microglia has dramatically accelerated during the last few years, largely due to recent genetic findings implicating microglia in virtually every neurodegenerative disorder. In Alzheimer's disease (AD), a majority of risk loci discovered through genome-wide association studies were found in or near genes expressed most highly in microglia leading to the hypothesis that microglia play a much larger role in disease progression than previously thought. From this body of work produced in the last several years, we find that almost every function of microglia has been proposed to influence the progression of AD from altered phagocytosis and synaptic pruning to cytokine secretion and changes in trophic support. By studying key Alzheimer's risk genes such as TREM2, CD33, ABCA7, and MS4A6A, we will be able to distinguish true disease-modulatory pathways from the full range of microglial-related functions. To successfully carry out these experiments, more advanced microglial models are needed. Microglia are quite sensitive to their local environment, suggesting the need to more fully recapitulate an in vivo environment to study this highly plastic cell type. Likely only by combining the above approaches will the field fully elucidate the molecular pathways that regulate microglia and influence neurodegeneration, in turn uncovering potential new targets for future therapeutic development.
Collapse
Affiliation(s)
- Amanda McQuade
- Department of Neurobiology & Behavior, University of California Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA 92697, USA; Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA 92697, USA
| | - Mathew Blurton-Jones
- Department of Neurobiology & Behavior, University of California Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA 92697, USA; Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
66
|
Hammond TR, Marsh SE, Stevens B. Immune Signaling in Neurodegeneration. Immunity 2019; 50:955-974. [PMID: 30995509 PMCID: PMC6822103 DOI: 10.1016/j.immuni.2019.03.016] [Citation(s) in RCA: 208] [Impact Index Per Article: 41.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 03/17/2019] [Accepted: 03/18/2019] [Indexed: 02/07/2023]
Abstract
Neurodegenerative diseases of the central nervous system progressively rob patients of their memory, motor function, and ability to perform daily tasks. Advances in genetics and animal models are beginning to unearth an unexpected role of the immune system in disease onset and pathogenesis; however, the role of cytokines, growth factors, and other immune signaling pathways in disease pathogenesis is still being examined. Here we review recent genetic risk and genome-wide association studies and emerging mechanisms for three key immune pathways implicated in disease, the growth factor TGF-β, the complement cascade, and the extracellular receptor TREM2. These immune signaling pathways are important under both healthy and neurodegenerative conditions, and recent work has highlighted new functional aspects of their signaling. Finally, we assess future directions for immune-related research in neurodegeneration and potential avenues for immune-related therapies.
Collapse
Affiliation(s)
- Timothy R Hammond
- Boston Children's Hospital, F.M. Kirby Neurobiology Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Samuel E Marsh
- Boston Children's Hospital, F.M. Kirby Neurobiology Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Beth Stevens
- Boston Children's Hospital, F.M. Kirby Neurobiology Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA.
| |
Collapse
|
67
|
Pagan FL, Hebron ML, Wilmarth B, Torres‐Yaghi Y, Lawler A, Mundel EE, Yusuf N, Starr NJ, Arellano J, Howard HH, Peyton M, Matar S, Liu X, Fowler AJ, Schwartz SL, Ahn J, Moussa C. Pharmacokinetics and pharmacodynamics of a single dose Nilotinib in individuals with Parkinson's disease. Pharmacol Res Perspect 2019; 7:e00470. [PMID: 30906562 PMCID: PMC6412143 DOI: 10.1002/prp2.470] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 02/07/2019] [Accepted: 02/07/2019] [Indexed: 01/17/2023] Open
Abstract
Nilotinib is a broad-based tyrosine kinase inhibitor with the highest affinity to inhibit Abelson (c-Abl) and discoidin domain receptors (DDR1/2). Preclinical evidence indicates that Nilotinib reduces the level of brain alpha-synuclein and attenuates inflammation in models of Parkinson's disease (PD). We previously showed that Nilotinib penetrates the blood-brain barrier (BBB) and potentially improves clinical outcomes in individuals with PD and dementia with Lewy bodies (DLB). We performed a physiologically based population pharmacokinetic/pharmacodynamic (popPK/PD) study to determine the effects of Nilotinib in a cohort of 75 PD participants. Participants were randomized (1:1:1:1:1) into five groups (n = 15) and received open-label random single dose (RSD) 150:200:300:400 mg Nilotinib vs placebo. Plasma and cerebrospinal fluid (CSF) were collected at 1, 2, 3, and 4 hours after Nilotinib administration. The results show that Nilotinib enters the brain in a dose-independent manner and 200 mg Nilotinib increases the level of 3,4-Dihydroxyphenylacetic acid (DOPAC) and homovanillic acid (HVA), suggesting alteration to dopamine metabolism. Nilotinib significantly reduces plasma total alpha-synuclein and appears to reduce CSF oligomeric: total alpha-synuclein ratio. Furthermore, Nilotinib significantly increases the CSF level of triggering receptors on myeloid cells (TREM)-2, suggesting an anti-inflammatory effect. Taken together, 200 mg Nilotinib appears to be an optimal single dose that concurrently reduces inflammation and engages surrogate disease biomarkers, including dopamine metabolism and alpha-synuclein.
Collapse
Affiliation(s)
- Fernando L. Pagan
- Translational Neurotherapeutics ProgramLaboratory for Dementia and ParkinsonismDepartment of NeurologyGeorgetown University Medical CenterWashingtonDistrict of Columbia
- Movement Disorders ClinicDepartment of NeurologyMedStar Georgetown University HospitalWashingtonDistrict of Columbia
| | - Michaeline L. Hebron
- Translational Neurotherapeutics ProgramLaboratory for Dementia and ParkinsonismDepartment of NeurologyGeorgetown University Medical CenterWashingtonDistrict of Columbia
| | - Barbara Wilmarth
- Translational Neurotherapeutics ProgramLaboratory for Dementia and ParkinsonismDepartment of NeurologyGeorgetown University Medical CenterWashingtonDistrict of Columbia
- Movement Disorders ClinicDepartment of NeurologyMedStar Georgetown University HospitalWashingtonDistrict of Columbia
| | - Yasar Torres‐Yaghi
- Translational Neurotherapeutics ProgramLaboratory for Dementia and ParkinsonismDepartment of NeurologyGeorgetown University Medical CenterWashingtonDistrict of Columbia
- Movement Disorders ClinicDepartment of NeurologyMedStar Georgetown University HospitalWashingtonDistrict of Columbia
| | - Abigail Lawler
- Translational Neurotherapeutics ProgramLaboratory for Dementia and ParkinsonismDepartment of NeurologyGeorgetown University Medical CenterWashingtonDistrict of Columbia
| | - Elizabeth E. Mundel
- Translational Neurotherapeutics ProgramLaboratory for Dementia and ParkinsonismDepartment of NeurologyGeorgetown University Medical CenterWashingtonDistrict of Columbia
- Movement Disorders ClinicDepartment of NeurologyMedStar Georgetown University HospitalWashingtonDistrict of Columbia
| | - Nadia Yusuf
- Translational Neurotherapeutics ProgramLaboratory for Dementia and ParkinsonismDepartment of NeurologyGeorgetown University Medical CenterWashingtonDistrict of Columbia
- Movement Disorders ClinicDepartment of NeurologyMedStar Georgetown University HospitalWashingtonDistrict of Columbia
| | - Nathan J. Starr
- Translational Neurotherapeutics ProgramLaboratory for Dementia and ParkinsonismDepartment of NeurologyGeorgetown University Medical CenterWashingtonDistrict of Columbia
- Movement Disorders ClinicDepartment of NeurologyMedStar Georgetown University HospitalWashingtonDistrict of Columbia
| | - Joy Arellano
- Movement Disorders ClinicDepartment of NeurologyMedStar Georgetown University HospitalWashingtonDistrict of Columbia
| | - Helen H. Howard
- Movement Disorders ClinicDepartment of NeurologyMedStar Georgetown University HospitalWashingtonDistrict of Columbia
| | - Margo Peyton
- Translational Neurotherapeutics ProgramLaboratory for Dementia and ParkinsonismDepartment of NeurologyGeorgetown University Medical CenterWashingtonDistrict of Columbia
| | - Sara Matar
- Translational Neurotherapeutics ProgramLaboratory for Dementia and ParkinsonismDepartment of NeurologyGeorgetown University Medical CenterWashingtonDistrict of Columbia
| | - Xiaoguang Liu
- Translational Neurotherapeutics ProgramLaboratory for Dementia and ParkinsonismDepartment of NeurologyGeorgetown University Medical CenterWashingtonDistrict of Columbia
| | - Alan J. Fowler
- Translational Neurotherapeutics ProgramLaboratory for Dementia and ParkinsonismDepartment of NeurologyGeorgetown University Medical CenterWashingtonDistrict of Columbia
| | - Sorell L. Schwartz
- Department of PharmacologyGeorgetown University Medical CenterWashingtonDistrict of Columbia
| | - Jaeil Ahn
- Department of Biostatistics, Bioinformatics and BiomathematicsGeorgetown University Medical CenterWashingtonDistrict of Columbia
| | - Charbel Moussa
- Translational Neurotherapeutics ProgramLaboratory for Dementia and ParkinsonismDepartment of NeurologyGeorgetown University Medical CenterWashingtonDistrict of Columbia
| |
Collapse
|
68
|
Ohara T, Hata J, Tanaka M, Honda T, Yamakage H, Yoshida D, Inoue T, Hirakawa Y, Kusakabe T, Shibata M, Teraoka T, Kitazono T, Kanba S, Satoh-Asahara N, Ninomiya T. Serum Soluble Triggering Receptor Expressed on Myeloid Cells 2 as a Biomarker for Incident Dementia: The Hisayama Study. Ann Neurol 2018; 85:47-58. [PMID: 30485483 DOI: 10.1002/ana.25385] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 11/22/2018] [Accepted: 11/23/2018] [Indexed: 12/28/2022]
Abstract
OBJECTIVE To investigate the association between serum soluble triggering receptor expressed on myeloid cells 2 (sTREM2), a soluble type of an innate immune receptor expressed on the microglia, and the risk of dementia. METHODS A total of 1,349 Japanese community residents aged 60 and older without dementia were followed prospectively for 10 years (2002-2012). Serum sTREM2 levels were quantified by using an enzyme-linked immunosorbent assay and divided into quartiles. Cox proportional hazards model was used to estimate the hazard ratios (HRs) of serum sTREM2 levels on the risk of dementia. RESULTS During the follow-up, 300 subjects developed all-cause dementia; 193 had Alzheimer's disease (AD), and 85 had vascular dementia (VaD). The age- and sex-adjusted incidences of all-cause dementia, AD, and VaD elevated significantly with higher serum sTREM2 levels (all p for trend < 0.012). These associations were not altered after adjustment for confounding factors, including high-sensitive C-reactive protein. Subjects with the highest quartile of serum sTREM2 levels had significantly higher multivariable-adjusted risks of developing all-cause dementia, AD, and VaD than those with the lowest quartile (HR = 2.03, 95% confidence interval [CI] = 1.39-2.97, p < 0.001 for all-cause dementia; HR = 1.62, 95% CI = 1.02-2.55, p = 0.04 for AD; HR = 2.85, 95% CI = 1.35-6.02, p = 0.006 for VaD). No significant heterogeneity in the association of serum sTREM2 levels with the development of dementia was observed among the other risk factor subgroups (all p for heterogeneity > 0.11). INTERPRETATION The present findings suggest a significant association between increased serum sTREM2 levels and the risk of developing all-cause dementia, AD, and VaD in the general elderly Japanese population. ANN NEUROL 2019;85:47-58.
Collapse
Affiliation(s)
- Tomoyuki Ohara
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka.,Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University, Fukuoka
| | - Jun Hata
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University, Fukuoka.,Department of Center for Cohort Studies, Graduate School of Medical Sciences, Kyushu University, Fukuoka.,Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka
| | - Masashi Tanaka
- Department of Endocrinology, Metabolism, and Hypertension Research, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Kyoto
| | - Takanori Honda
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University, Fukuoka
| | - Hajime Yamakage
- Department of Endocrinology, Metabolism, and Hypertension Research, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Kyoto
| | - Daigo Yoshida
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University, Fukuoka.,Department of Center for Cohort Studies, Graduate School of Medical Sciences, Kyushu University, Fukuoka
| | - Takayuki Inoue
- Department of Endocrinology, Metabolism, and Hypertension Research, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Kyoto
| | - Yoichiro Hirakawa
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University, Fukuoka.,Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka
| | - Toru Kusakabe
- Department of Endocrinology, Metabolism, and Hypertension Research, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Kyoto
| | - Mao Shibata
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University, Fukuoka.,Department of Center for Cohort Studies, Graduate School of Medical Sciences, Kyushu University, Fukuoka
| | | | - Takanari Kitazono
- Department of Center for Cohort Studies, Graduate School of Medical Sciences, Kyushu University, Fukuoka.,Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka
| | - Shigenobu Kanba
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka
| | - Noriko Satoh-Asahara
- Department of Endocrinology, Metabolism, and Hypertension Research, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Kyoto
| | - Toshiharu Ninomiya
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University, Fukuoka.,Department of Center for Cohort Studies, Graduate School of Medical Sciences, Kyushu University, Fukuoka
| |
Collapse
|
69
|
The Role of APOE and TREM2 in Alzheimer's Disease-Current Understanding and Perspectives. Int J Mol Sci 2018; 20:ijms20010081. [PMID: 30587772 PMCID: PMC6337314 DOI: 10.3390/ijms20010081] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 12/19/2018] [Accepted: 12/21/2018] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is the leading cause of dementia worldwide. The extracellular deposits of Amyloid beta (Aβ) in the brain-called amyloid plaques, and neurofibrillary tangles-intracellular tau aggregates, are morphological hallmarks of the disease. The risk for AD is a complicated interplay between aging, genetic risk factors, and environmental influences. One of the Apolipoprotein E (APOE) alleles-APOEε4, is the major genetic risk factor for late-onset AD (LOAD). APOE is the primary cholesterol carrier in the brain, and plays an essential role in lipid trafficking, cholesterol homeostasis, and synaptic stability. Recent genome-wide association studies (GWAS) have identified other candidate LOAD risk loci, as well. One of those is the triggering receptor expressed on myeloid cells 2 (TREM2), which, in the brain, is expressed primarily by microglia. While the function of TREM2 is not fully understood, it promotes microglia survival, proliferation, and phagocytosis, making it important for cell viability and normal immune functions in the brain. Emerging evidence from protein binding assays suggests that APOE binds to TREM2 and APOE-containing lipoproteins in the brain as well as periphery, and are putative ligands for TREM2, thus raising the possibility of an APOE-TREM2 interaction modulating different aspects of AD pathology, potentially in an isoform-specific manner. This review is focusing on the interplay between APOE isoforms and TREM2 in association with AD pathology.
Collapse
|
70
|
Gratuze M, Leyns CEG, Holtzman DM. New insights into the role of TREM2 in Alzheimer's disease. Mol Neurodegener 2018; 13:66. [PMID: 30572908 PMCID: PMC6302500 DOI: 10.1186/s13024-018-0298-9] [Citation(s) in RCA: 288] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 11/28/2018] [Indexed: 12/22/2022] Open
Abstract
Alzheimer's disease (AD) is the leading cause of dementia. The two histopathological markers of AD are amyloid plaques composed of the amyloid-β (Aβ) peptide, and neurofibrillary tangles of aggregated, abnormally hyperphosphorylated tau protein. The majority of AD cases are late-onset, after the age of 65, where a clear cause is still unknown. However, there are likely different multifactorial contributors including age, enviornment, biology and genetics which can increase risk for the disease. Genetic predisposition is considerable, with heritability estimates of 60-80%. Genetic factors such as rare variants of TREM2 (triggering receptor expressed on myeloid cells-2) strongly increase the risk of developing AD, confirming the role of microglia in AD pathogenesis. In the last 5 years, several studies have dissected the mechanisms by which TREM2, as well as its rare variants affect amyloid and tau pathologies and their consequences in both animal models and in human studies. In this review, we summarize increases in our understanding of the involvement of TREM2 and microglia in AD development that may open new therapeutic strategies targeting the immune system to influence AD pathogenesis.
Collapse
Affiliation(s)
- Maud Gratuze
- Department of Neurology, St. Louis, USA
- Hope Center for Neurological Disorders, St. Louis, USA
- Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Cheryl E. G. Leyns
- Department of Neurology, St. Louis, USA
- Hope Center for Neurological Disorders, St. Louis, USA
- Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - David M. Holtzman
- Department of Neurology, St. Louis, USA
- Hope Center for Neurological Disorders, St. Louis, USA
- Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110 USA
| |
Collapse
|
71
|
García JC, Bustos RH. The Genetic Diagnosis of Neurodegenerative Diseases and Therapeutic Perspectives. Brain Sci 2018; 8:brainsci8120222. [PMID: 30551598 PMCID: PMC6316116 DOI: 10.3390/brainsci8120222] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 11/26/2018] [Accepted: 12/07/2018] [Indexed: 12/12/2022] Open
Abstract
Genetics has led to a new focus regarding approaches to the most prevalent diseases today. Ascertaining the molecular secrets of neurodegenerative diseases will lead to developing drugs that will change natural history, thereby affecting the quality of life and mortality of patients. The sequencing of candidate genes in patients suffering neurodegenerative pathologies is faster, more accurate, and has a lower cost, thereby enabling algorithms to be proposed regarding the risk of neurodegeneration onset in healthy persons including the year of onset and neurodegeneration severity. Next generation sequencing has resulted in an explosion of articles regarding the diagnosis of neurodegenerative diseases involving exome sequencing or sequencing a whole gene for correlating phenotypical expression with genetic mutations in proteins having key functions. Many of them occur in neuronal glia, which can trigger a proinflammatory effect leading to defective proteins causing sporadic or familial mutations. This article reviews the genetic diagnosis techniques and the importance of bioinformatics in interpreting results from neurodegenerative diseases. Risk scores must be established in the near future regarding diseases with a high incidence in healthy people for defining prevention strategies or an early start for giving drugs in the absence of symptoms.
Collapse
Affiliation(s)
- Julio-César García
- Evidence-Based Therapeutics Group, Department of Clinical Pharmacology, Universidad de La Sabana, Chía 140013, Colombia.
- Department of Clinical Pharmacology, Clínica Universidad de La Sabana, Chía 140013, Colombia.
| | - Rosa-Helena Bustos
- Evidence-Based Therapeutics Group, Department of Clinical Pharmacology, Universidad de La Sabana, Chía 140013, Colombia.
| |
Collapse
|
72
|
Zheng H, Cheng B, Li Y, Li X, Chen X, Zhang YW. TREM2 in Alzheimer's Disease: Microglial Survival and Energy Metabolism. Front Aging Neurosci 2018; 10:395. [PMID: 30532704 PMCID: PMC6265312 DOI: 10.3389/fnagi.2018.00395] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 11/13/2018] [Indexed: 12/16/2022] Open
Abstract
Alzheimer’s disease (AD) is the leading cause of age-related dementia among the elderly population. Recent genetic studies have identified rare variants of the gene encoding the triggering receptor expressed on myeloid cells-2 (TREM2) as significant genetic risk factors in late-onset AD (LOAD). TREM2 is specifically expressed in brain microglia and modulates microglial functions in response to key AD pathologies such as amyloid-β (Aβ) plaques and tau tangles. In this review article, we discuss recent research progress in our understanding on the role of TREM2 in microglia and its relevance to AD pathologies. In addition, we discuss evidence describing new TREM2 ligands and the role of TREM2 signaling in microglial survival and energy metabolism. A comprehensive understanding of TREM2 function in the pathogenesis of AD offers a unique opportunity to explore the potential of this microglial receptor as an alternative target in AD therapy.
Collapse
Affiliation(s)
- Honghua Zheng
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Medical College, Xiamen University, Xiamen, China.,Shenzhen Research Institute, Xiamen University, Shenzhen, China
| | - Baoying Cheng
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Medical College, Xiamen University, Xiamen, China
| | - Yanfang Li
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Medical College, Xiamen University, Xiamen, China.,Shenzhen Research Institute, Xiamen University, Shenzhen, China
| | - Xin Li
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Medical College, Xiamen University, Xiamen, China
| | - Xiaofen Chen
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Medical College, Xiamen University, Xiamen, China.,Shenzhen Research Institute, Xiamen University, Shenzhen, China
| | - Yun-Wu Zhang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Medical College, Xiamen University, Xiamen, China
| |
Collapse
|
73
|
Katsumoto A, Takeuchi H, Takahashi K, Tanaka F. Microglia in Alzheimer's Disease: Risk Factors and Inflammation. Front Neurol 2018; 9:978. [PMID: 30498474 PMCID: PMC6249341 DOI: 10.3389/fneur.2018.00978] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 10/30/2018] [Indexed: 01/28/2023] Open
Abstract
Microglia are resident immune cells in the central nervous system (CNS) that originate from myeloid progenitor cells in the embryonic yolk sac and are maintained independently of circulating monocytes throughout life. In the healthy state, microglia are highly dynamic and control the environment by rapidly extending and retracting their processes. When the CNS is inflamed, microglia can give rise to macrophages, but the regulatory mechanisms underlying this process have not been fully elucidated. Recent genetic studies have suggested that microglial function is compromised in Alzheimer's disease (AD), and that environmental factors such as diet and brain injury also affect microglial activation. In addition, studies of triggering receptor expressed on myeloid cells 2-deficiency in AD mice revealed heterogeneous microglial reactions at different disease stages, complicating the therapeutic strategy for AD. In this paper, we describe the relationship between genetic and environmental risk factors and the roles of microglia in AD pathogenesis, based on studies performed in human patients and animal models. We also discuss the mechanisms of inflammasomes and neurotransmitters in microglia, which accelerate the development of amyloid-β and tau pathology.
Collapse
Affiliation(s)
- Atsuko Katsumoto
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Hideyuki Takeuchi
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Keita Takahashi
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Fumiaki Tanaka
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| |
Collapse
|
74
|
Deconstructing and targeting the genomic architecture of human neurodegeneration. Nat Neurosci 2018; 21:1310-1317. [PMID: 30258235 DOI: 10.1038/s41593-018-0240-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 08/23/2018] [Indexed: 01/17/2023]
Abstract
The field of neurodegenerative disease research has seen tremendous advances over the last two decades as new technologies and analytic methods have enabled well-powered human genomic studies. Driven first by genetic studies and more recently by transcriptomic and epigenomic studies of proper size, we have uncovered a large repertoire of loci, genes, and molecular features that are implicated in discrete, syndromically defined neurodegenerative conditions, such as Alzheimer's disease, amyotrophic lateral sclerosis, frontotemporal dementia, multiple sclerosis, and Parkinson's disease. As we begin to understand the impact of these genomic features in each disease, we also appreciate that many aging individuals accumulate each of these pathologies without fulfilling criteria for syndromic diagnoses, that other pathologies are common in individuals with a given diagnosis, and that there may be shared protective factors against central nervous system injury. Thus, we now need to bring these disparate observations together into a person-centered approach that considers all neurodegenerative and protective processes simultaneously to modulate the trajectory of cognitive and functional decline that comes with brain aging.
Collapse
|
75
|
Hickman S, Izzy S, Sen P, Morsett L, El Khoury J. Microglia in neurodegeneration. Nat Neurosci 2018; 21:1359-1369. [PMID: 30258234 DOI: 10.1038/s41593-018-0242-x] [Citation(s) in RCA: 1008] [Impact Index Per Article: 168.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 08/06/2018] [Indexed: 12/13/2022]
Abstract
The neuroimmune system is involved in development, normal functioning, aging, and injury of the central nervous system. Microglia, first described a century ago, are the main neuroimmune cells and have three essential functions: a sentinel function involved in constant sensing of changes in their environment, a housekeeping function that promotes neuronal well-being and normal operation, and a defense function necessary for responding to such changes and providing neuroprotection. Microglia use a defined armamentarium of genes to perform these tasks. In response to specific stimuli, or with neuroinflammation, microglia also have the capacity to damage and kill neurons. Injury to neurons in Alzheimer's, Parkinson's, Huntington's, and prion diseases, as well as in amyotrophic lateral sclerosis, frontotemporal dementia, and chronic traumatic encephalopathy, results from disruption of the sentinel or housekeeping functions and dysregulation of the defense function and neuroinflammation. Pathways associated with such injury include several sensing and housekeeping pathways, such as the Trem2, Cx3cr1 and progranulin pathways, which act as immune checkpoints to keep the microglial inflammatory response under control, and the scavenger receptor pathways, which promote clearance of injurious stimuli. Peripheral interference from systemic inflammation or the gut microbiome can also alter progression of such injury. Initiation or exacerbation of neurodegeneration results from an imbalance between these microglial functions; correcting such imbalance may be a potential mode for therapy.
Collapse
Affiliation(s)
- Suzanne Hickman
- Center for Immunology & Inflammatory Diseases, Massachusetts General Hospital, and Harvard Medical School, Boston, MA, USA
| | - Saef Izzy
- Center for Immunology & Inflammatory Diseases, Massachusetts General Hospital, and Harvard Medical School, Boston, MA, USA
| | - Pritha Sen
- Center for Immunology & Inflammatory Diseases, Massachusetts General Hospital, and Harvard Medical School, Boston, MA, USA
| | - Liza Morsett
- Center for Immunology & Inflammatory Diseases, Massachusetts General Hospital, and Harvard Medical School, Boston, MA, USA
| | - Joseph El Khoury
- Center for Immunology & Inflammatory Diseases, Massachusetts General Hospital, and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
76
|
Konishi H, Kiyama H. Microglial TREM2/DAP12 Signaling: A Double-Edged Sword in Neural Diseases. Front Cell Neurosci 2018; 12:206. [PMID: 30127720 PMCID: PMC6087757 DOI: 10.3389/fncel.2018.00206] [Citation(s) in RCA: 185] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 06/25/2018] [Indexed: 12/21/2022] Open
Abstract
Microglia are activated after neuronal injury and in neurodegenerative diseases, and trigger neuroinflammation in the central nervous system (CNS). Microglia-derived neuroinflammation has both beneficial and detrimental effects on neurons. Because the timing and magnitude of microglial activation is thought to be a critical determinant of neuronal fate, understanding the molecular mechanisms underlying microglial activation is required to enable establishment of microglia-targeted therapies for neural diseases. Plasma membrane receptors play primary roles as activators of microglia and in this review, we focus on a receptor complex involving triggering receptor expressed on myeloid cells 2 (TREM2) and DNAX-activating protein of 12 kDa (DAP12), both of which are causative genes for Nasu-Hakola disease, a dementia with bone cysts. Recent transcriptome approaches demonstrated TREM2/DAP12 signaling as the principal regulator that transforms microglia from a homeostatic to a neural disease-associated state. Furthermore, animal model studies revealed critical roles for TREM2/DAP12 in the regulation of microglial activity, including survival, phagocytosis, and cytokine production, not only in Alzheimer's disease but also in other neural diseases, such as Parkinson's disease, demyelinating disease, ischemia, and peripheral nerve injury. Intriguingly, while TREM2/DAP12-mediated microglial activation is detrimental for some diseases, including peripheral nerve injury, it is beneficial for other diseases. As the role of activated microglia differs among disease models, TREM2/DAP12 signaling may result in different outcomes in different diseases. In this review we discuss recent perspectives on the role of TREM2/DAP12 in microglia and their contribution to neural diseases.
Collapse
Affiliation(s)
- Hiroyuki Konishi
- Department of Functional Anatomy and Neuroscience, Nagoya University Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Hiroshi Kiyama
- Department of Functional Anatomy and Neuroscience, Nagoya University Graduate School of Medicine, Nagoya University, Nagoya, Japan
| |
Collapse
|
77
|
The role of TREM2 in Alzheimer's disease and other neurodegenerative disorders. Lancet Neurol 2018; 17:721-730. [PMID: 30033062 DOI: 10.1016/s1474-4422(18)30232-1] [Citation(s) in RCA: 163] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 06/04/2018] [Accepted: 06/06/2018] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease is a genetically complex disorder; rare variants in the triggering receptor expressed on myeloid cells 2 (TREM2) gene have been shown to as much as triple an individual's risk of developing Alzheimer's disease. TREM2 is a transmembrane receptor expressed in cells of the myeloid lineage, and its association with Alzheimer's disease supports the involvement of immune and inflammatory pathways in the cause of the disease, rather than as a consequence of the disease. TREM2 variants associated with Alzheimer's disease induce partial loss of function of the TREM2 protein and alter the behaviour of microglial cells, including their response to amyloid plaques. TREM2 variants have also been shown to cause polycystic lipomembranous osteodysplasia with sclerosing leukoencephalopathy and frontotemporal dementia. Although the low frequency of TREM2 variants makes it difficult to establish robust genotype-phenotype correlations, such studies are essential to enable a comprehensive understanding of the role of TREM2 in different neurological diseases, with the ultimate goal of developing novel therapeutic approaches.
Collapse
|
78
|
Li JT, Zhang Y. TREM2 regulates innate immunity in Alzheimer's disease. J Neuroinflammation 2018; 15:107. [PMID: 29655369 PMCID: PMC5899410 DOI: 10.1186/s12974-018-1148-y] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Accepted: 04/04/2018] [Indexed: 11/10/2022] Open
Abstract
Recent research has shown that the triggering receptor expressed on myeloid cells 2 (TREM2) in microglia is closely related to the pathogenesis of Alzheimer's disease (AD). The mechanism of this relationship, however, remains unclear. TREM2 is part of the TREM family of receptors, which are expressed primarily in myeloid cells, including monocytes, dendritic cells, and microglia. The TREM family members are cell surface glycoproteins with an immunoglobulin-like extracellular domain, a transmembrane region and a short cytoplasmic tail region. The present article reviews the following: (1) the structure, function, and variant site analysis of the Trem2 gene; (2) the metabolism of TREM2 in peripheral blood and cerebrospinal fluid; and (3) the possible underlying mechanism by which TREM2 regulates innate immunity and participates in AD.
Collapse
Affiliation(s)
- Jiang-Tao Li
- College of Life Sciences and Bioengineering, Beijing Jiaotong University, No. 3 Shangyuan Residence, Haidian District, Beijing, 100044, China
| | - Ying Zhang
- College of Life Sciences and Bioengineering, Beijing Jiaotong University, No. 3 Shangyuan Residence, Haidian District, Beijing, 100044, China.
| |
Collapse
|
79
|
Hampel H, Vergallo A, Aguilar LF, Benda N, Broich K, Cuello AC, Cummings J, Dubois B, Federoff HJ, Fiandaca M, Genthon R, Haberkamp M, Karran E, Mapstone M, Perry G, Schneider LS, Welikovitch LA, Woodcock J, Baldacci F, Lista S. Precision pharmacology for Alzheimer’s disease. Pharmacol Res 2018; 130:331-365. [DOI: 10.1016/j.phrs.2018.02.014] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Revised: 02/11/2018] [Accepted: 02/12/2018] [Indexed: 12/12/2022]
|
80
|
Sekiya M, Wang M, Fujisaki N, Sakakibara Y, Quan X, Ehrlich ME, De Jager PL, Bennett DA, Schadt EE, Gandy S, Ando K, Zhang B, Iijima KM. Integrated biology approach reveals molecular and pathological interactions among Alzheimer's Aβ42, Tau, TREM2, and TYROBP in Drosophila models. Genome Med 2018; 10:26. [PMID: 29598827 PMCID: PMC5875009 DOI: 10.1186/s13073-018-0530-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 03/12/2018] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Cerebral amyloidosis, neuroinflammation, and tauopathy are key features of Alzheimer's disease (AD), but interactions among these features remain poorly understood. Our previous multiscale molecular network models of AD revealed TYROBP as a key driver of an immune- and microglia-specific network that was robustly associated with AD pathophysiology. Recent genetic studies of AD further identified pathogenic mutations in both TREM2 and TYROBP. METHODS In this study, we systematically examined molecular and pathological interactions among Aβ, tau, TREM2, and TYROBP by integrating signatures from transgenic Drosophila models of AD and transcriptome-wide gene co-expression networks from two human AD cohorts. RESULTS Glial expression of TREM2/TYROBP exacerbated tau-mediated neurodegeneration and synergistically affected pathways underlying late-onset AD pathology, while neuronal Aβ42 and glial TREM2/TYROBP synergistically altered expression of the genes in synaptic function and immune modules in AD. CONCLUSIONS The comprehensive pathological and molecular data generated through this study strongly validate the causal role of TREM2/TYROBP in driving molecular networks in AD and AD-related phenotypes in flies.
Collapse
Affiliation(s)
- Michiko Sekiya
- 0000 0004 1791 9005grid.419257.cDepartment of Alzheimer’s Disease Research, National Center for Geriatrics and Gerontology, 7-430 Morioka-cho, Obu, Aichi 474-8511 Japan
| | - Minghui Wang
- 0000 0001 0670 2351grid.59734.3cDepartment of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Room 8-111, Box 1498, New York, NY 10029 USA ,0000 0001 0670 2351grid.59734.3cIcahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY USA
| | - Naoki Fujisaki
- 0000 0004 1791 9005grid.419257.cDepartment of Alzheimer’s Disease Research, National Center for Geriatrics and Gerontology, 7-430 Morioka-cho, Obu, Aichi 474-8511 Japan ,0000 0001 0728 1069grid.260433.0Department of Experimental Gerontology, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Japan
| | - Yasufumi Sakakibara
- 0000 0004 1791 9005grid.419257.cDepartment of Alzheimer’s Disease Research, National Center for Geriatrics and Gerontology, 7-430 Morioka-cho, Obu, Aichi 474-8511 Japan
| | - Xiuming Quan
- 0000 0004 1791 9005grid.419257.cDepartment of Alzheimer’s Disease Research, National Center for Geriatrics and Gerontology, 7-430 Morioka-cho, Obu, Aichi 474-8511 Japan
| | - Michelle E. Ehrlich
- 0000 0001 0670 2351grid.59734.3cDepartment of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Room 8-111, Box 1498, New York, NY 10029 USA ,0000 0001 0670 2351grid.59734.3cDepartment of Neurology, Alzheimer’s Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY USA ,0000 0001 0670 2351grid.59734.3cDepartment of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Philip L. De Jager
- 0000 0001 2285 2675grid.239585.0Center for translational & Computational Neuroimmunology, Department of Neurology, The Neurological Institute of New York, Columbia University Medical Center, New York, NY USA ,grid.66859.34Broad Institute, Cambridge, MA USA
| | - David A. Bennett
- 0000 0001 0705 3621grid.240684.cRush Alzheimer’s Disease Research Center and Department of Neurology, Rush University Medical Center, 1750 W. Congress Parkway, Chicago, IL 60612 USA
| | - Eric E. Schadt
- 0000 0001 0670 2351grid.59734.3cDepartment of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Room 8-111, Box 1498, New York, NY 10029 USA ,0000 0001 0670 2351grid.59734.3cIcahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY USA
| | - Sam Gandy
- 0000 0001 0670 2351grid.59734.3cDepartment of Neurology, Alzheimer’s Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY USA ,0000 0001 0670 2351grid.59734.3cDepartment of Psychiatry and Alzheimer’s Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY USA ,0000 0004 5907 0628grid.480949.8Center for NFL Neurological Care, Department of Neurology, New York, NY USA ,0000 0004 0420 1184grid.274295.fJames J. Peters VA Medical Center, 130 West Kingsbridge Road, New York, NY USA
| | - Kanae Ando
- 0000 0001 1090 2030grid.265074.2Department of Biological Sciences, Graduate School of Science and Engineering, Tokyo Metropolitan University, Tokyo, Japan
| | - Bin Zhang
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Room 8-111, Box 1498, New York, NY, 10029, USA. .,Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA. .,Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, One Gustave L Levy Place, New York, NY, USA.
| | - Koichi M. Iijima
- 0000 0004 1791 9005grid.419257.cDepartment of Alzheimer’s Disease Research, National Center for Geriatrics and Gerontology, 7-430 Morioka-cho, Obu, Aichi 474-8511 Japan ,0000 0001 0728 1069grid.260433.0Department of Experimental Gerontology, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Japan
| |
Collapse
|
81
|
Peplonska B, Berdynski M, Mandecka M, Barczak A, Kuzma-Kozakiewicz M, Barcikowska M, Zekanowski C. TREM2 variants in neurodegenerative disorders in the Polish population. Homozygosity and compound heterozygosity in FTD patients. Amyotroph Lateral Scler Frontotemporal Degener 2018; 19:407-412. [PMID: 29557178 DOI: 10.1080/21678421.2018.1451894] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Activation of the TREM2 receptor on microglia stimulates phagocytosis and decreases the microglial proinflammatory response. Mutations in exon 2 of the TREM2 gene have been reported to be associated with various neurodegenerative diseases characterized by chronic inflammation. The aim of our study was to evaluate exon 2 of TREM2 gene variants as a putative genetic risk factor for Alzheimer's disease (AD), frontotemporal dementia (FTD), and amyotrophic lateral sclerosis (ALS) in the Polish population. The results were interpreted using previously published data, especially highlighting differences in the prevalence of the variants among Caucasian subpopulations across different geographic regions. The DNA sequence of exon 2 of TREM2 was analyzed in 811 subjects (274 AD, 135 FTD, 194 ALS patients, and 208 neurologically healthy controls). Nine heterozygous variants were detected, including two novel ones: p.G29 = and c.41-2_3insA, found respectively in a control and an ALS patient. Additionally, we identified one homozygous and two compound heterozygous FTD patients. We confirm previous data that homozygous and compound heterozygous TREM2 mutations can be causative for FTD.
Collapse
Affiliation(s)
- Beata Peplonska
- a Department of Neurodegenerative Disorders , Mossakowski Medical Research Centre Polish Academy of Sciences , Warsaw , Poland
| | - Mariusz Berdynski
- a Department of Neurodegenerative Disorders , Mossakowski Medical Research Centre Polish Academy of Sciences , Warsaw , Poland
| | - Monika Mandecka
- a Department of Neurodegenerative Disorders , Mossakowski Medical Research Centre Polish Academy of Sciences , Warsaw , Poland
| | - Anna Barczak
- a Department of Neurodegenerative Disorders , Mossakowski Medical Research Centre Polish Academy of Sciences , Warsaw , Poland
| | - Magdalena Kuzma-Kozakiewicz
- b Department of Neurology , and.,c Neurodegenerative Diseases Research Group Medical University of Warsaw , Warsaw , Poland
| | - Maria Barcikowska
- a Department of Neurodegenerative Disorders , Mossakowski Medical Research Centre Polish Academy of Sciences , Warsaw , Poland
| | - Cezary Zekanowski
- a Department of Neurodegenerative Disorders , Mossakowski Medical Research Centre Polish Academy of Sciences , Warsaw , Poland
| |
Collapse
|
82
|
Bekris LM, Khrestian M, Dyne E, Shao Y, Pillai JA, Rao SM, Bemiller SM, Lamb B, Fernandez HH, Leverenz JB. Soluble TREM2 and biomarkers of central and peripheral inflammation in neurodegenerative disease. J Neuroimmunol 2018; 319:19-27. [PMID: 29685286 DOI: 10.1016/j.jneuroim.2018.03.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 03/08/2018] [Accepted: 03/08/2018] [Indexed: 01/22/2023]
Abstract
Alzheimer's disease (AD) has been genetically and pathologically associated with neuroinflammation. Triggering receptor expressed on myeloid cells 2 (TREM2) is a microglial receptor involved in innate immunity. TREM2 rare protein coding genetic variants have been linked to AD. A soluble TREM2 (sTREM2) cleavage product is elevated in AD. It is unclear whether there is a relationship between elevated sTREM2 and markers of inflammation. The hypothesis of this investigation was that central and peripheral inflammation play a role in sTREM2 levels in AD. A consistent association of peripheral or central markers of inflammation and CSF sTREM2 levels was not found, suggesting a limited impact of general inflammation on sTREM2 levels. An association between peripheral sTREM2 levels and CSF sTREM2, as well as an association between CSF sTREM2 and a marker of blood brain barrier integrity, was observed in AD, suggesting a potential role of peripheral TREM2 in central TREM2 biology.
Collapse
Affiliation(s)
- L M Bekris
- Cleveland Clinic, Genomic Medicine Institute, Cleveland, OH, United States.
| | - M Khrestian
- Cleveland Clinic, Genomic Medicine Institute, Cleveland, OH, United States
| | - E Dyne
- Kent State University, Departoment of Biomedicine, Kent, OH, United States
| | - Y Shao
- Cleveland Clinic, Genomic Medicine Institute, Cleveland, OH, United States
| | - J A Pillai
- Cleveland Clinic, Lou Ruvo Center for Brain Health, Cleveland, OH, United States
| | - S M Rao
- Cleveland Clinic, Lou Ruvo Center for Brain Health, Cleveland, OH, United States
| | - S M Bemiller
- Indiana University, Department of Neuroscience, Indianapolis, IN, United States
| | - B Lamb
- Indiana University, Department of Neuroscience, Indianapolis, IN, United States
| | - H H Fernandez
- Cleveland Clinic, Center for Neurological Restoration, Cleveland, OH, United States
| | - J B Leverenz
- Cleveland Clinic, Lou Ruvo Center for Brain Health, Cleveland, OH, United States
| |
Collapse
|
83
|
Chen H, Kankel MW, Su SC, Han SWS, Ofengeim D. Exploring the genetics and non-cell autonomous mechanisms underlying ALS/FTLD. Cell Death Differ 2018; 25:648-662. [PMID: 29459769 DOI: 10.1038/s41418-018-0060-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 11/27/2017] [Accepted: 11/28/2017] [Indexed: 12/11/2022] Open
Abstract
Although amyotrophic lateral sclerosis (ALS), also known as Lou Gehrig's disease, was first described in 1874, a flurry of genetic discoveries in the last 10 years has markedly increased our understanding of this disease. These findings have not only enhanced our knowledge of mechanisms leading to ALS, but also have revealed that ALS shares many genetic causes with another neurodegenerative disease, frontotemporal lobar dementia (FTLD). In this review, we survey how recent genetic studies have bridged our mechanistic understanding of these two related diseases and how the genetics behind ALS and FTLD point to complex disorders, implicating non-neuronal cell types in disease pathophysiology. The involvement of non-neuronal cell types is consistent with a non-cell autonomous component in these diseases. This is further supported by studies that identified a critical role of immune-associated genes within ALS/FTLD and other neurodegenerative disorders. The molecular functions of these genes support an emerging concept that various non-autonomous functions are involved in neurodegeneration. Further insights into such a mechanism(s) will ultimately lead to a better understanding of potential routes of therapeutic intervention. Facts ALS and FTLD are severe neurodegenerative disorders on the same disease spectrum. Multiple cellular processes including dysregulation of RNA homeostasis, imbalance of proteostasis, contribute to ALS/FTLD pathogenesis. Aberrant function in non-neuronal cell types, including microglia, contributes to ALS/FTLD. Strong neuroimmune and neuroinflammatory components are associated with ALS/FTLD patients. Open Questions Why can patients with similar mutations have different disease manifestations, i.e., why do C9ORF72 mutations lead to motor neuron loss in some patients while others exhibit loss of neurons in the frontotemporal lobe? Do ALS causal mutations result in microglial dysfunction and contribute to ALS/FTLD pathology? How do microglia normally act to mitigate neurodegeneration in ALS/FTLD? To what extent do cellular signaling pathways mediate non-cell autonomous communications between distinct central nervous system (CNS) cell types during disease? Is it possible to therapeutically target specific cell types in the CNS?
Collapse
Affiliation(s)
- Hongbo Chen
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.,Department of Cell Biology, Harvard Medical School, 240 Longwood Avenue, Boston, MA, 02115, USA
| | - Mark W Kankel
- Biogen Inc., 225 Binney Street, Cambridge, MA, 02142, USA
| | - Susan C Su
- Biogen Inc., 225 Binney Street, Cambridge, MA, 02142, USA
| | - Steve W S Han
- Biogen Inc., 225 Binney Street, Cambridge, MA, 02142, USA.,Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.,GSK, Upper Providence, PA, 19426, USA
| | - Dimitry Ofengeim
- Biogen Inc., 225 Binney Street, Cambridge, MA, 02142, USA. .,Sanofi Neuroscience, Framingham, MA, USA.
| |
Collapse
|
84
|
Condello C, Yuan P, Grutzendler J. Microglia-Mediated Neuroprotection, TREM2, and Alzheimer's Disease: Evidence From Optical Imaging. Biol Psychiatry 2018; 83:377-387. [PMID: 29169609 PMCID: PMC5767550 DOI: 10.1016/j.biopsych.2017.10.007] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 10/05/2017] [Accepted: 10/06/2017] [Indexed: 12/19/2022]
Abstract
Recent genetic studies have provided overwhelming evidence of the involvement of microglia-related molecular networks in the pathophysiology of Alzheimer's disease (AD). However, the precise mechanisms by which microglia alter the course of AD neuropathology remain poorly understood. Here we discuss current evidence of the neuroprotective functions of microglia with a focus on optical imaging studies that have revealed a role of these cells in the encapsulation of amyloid deposits ("microglia barrier"). This barrier modulates the degree of plaque compaction, amyloid fibril surface area, and insulation from adjacent axons thereby reducing neurotoxicity. We discuss findings implicating genetic variants of the microglia receptor, triggering receptor expressed on myeloid cells 2, in the increased risk of late onset AD. We provide evidence that increased AD risk may be at least partly mediated by deficient microglia polarization toward amyloid deposits, resulting in ineffective plaque encapsulation and reduced plaque compaction, which is associated with worsened axonal pathology. Finally, we propose possible avenues for therapeutic targeting of plaque-associated microglia with the goal of enhancing the microglia barrier and potentially reducing disease progression.
Collapse
Affiliation(s)
- Carlo Condello
- Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California, San Francisco, CA 94158, USA,Department of Neurology, University of California, San Francisco, CA 94158, USA
| | - Peng Yuan
- Department of Biology, Stanford University, Palo Alto, CA 94305, USA
| | - Jaime Grutzendler
- Department of Neurology, Yale School of Medicine, New Haven, Connecticut; Department of Neuroscience, Yale School of Medicine, New Haven, Connecticut.
| |
Collapse
|
85
|
Raha AA, Henderson JW, Stott SRW, Vuono R, Foscarin S, Friedland RP, Zaman SH, Raha-Chowdhury R. Neuroprotective Effect of TREM-2 in Aging and Alzheimer's Disease Model. J Alzheimers Dis 2018; 55:199-217. [PMID: 27662313 DOI: 10.3233/jad-160663] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Neuroinflammation and activation of innate immunity are early events in neurodegenerative diseases including Alzheimer's disease (AD). Recently, a rare mutation in the gene Triggering receptor expressed on myeloid cells 2 (TREM2) has been associated with a substantial increase in the risk of developing late onset AD. To uncover the molecular mechanisms underlying this association, we investigated the RNA and protein expression of TREM2 in APP/PS1 transgenic mice. Our findings suggest that TREM2 not only plays a critical role in inflammation, but is also involved in neuronal cell survival and in neurogenesis. We have shown that TREM2 is a soluble protein transported by macrophages through ventricle walls and choroid plexus, and then enters the brain parenchyma via radial glial cells. TREM2 protein is essential for neuroplasticity and myelination. During the late stages of life, a lack of TREM2 protein may accelerate aging processes and neuronal cell loss and reduce microglial activity, ultimately leading to neuroinflammation. As inflammation plays a major role in neurodegenerative diseases, a lack of TREM2 could be a missing link between immunomodulation and neuroprotection.
Collapse
Affiliation(s)
- Animesh Alexander Raha
- John Van Geest Centre for Brain Repair, Department of Clinical Neuroscience, University of Cambridge, Cambridge, UK
| | - James W Henderson
- John Van Geest Centre for Brain Repair, Department of Clinical Neuroscience, University of Cambridge, Cambridge, UK
| | - Simon R W Stott
- John Van Geest Centre for Brain Repair, Department of Clinical Neuroscience, University of Cambridge, Cambridge, UK
| | - Romina Vuono
- John Van Geest Centre for Brain Repair, Department of Clinical Neuroscience, University of Cambridge, Cambridge, UK
| | - Simona Foscarin
- John Van Geest Centre for Brain Repair, Department of Clinical Neuroscience, University of Cambridge, Cambridge, UK
| | | | - Shahid H Zaman
- Cambridge Intellectual & Developmental Disabilities Research Group, Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Ruma Raha-Chowdhury
- John Van Geest Centre for Brain Repair, Department of Clinical Neuroscience, University of Cambridge, Cambridge, UK
| |
Collapse
|
86
|
Zhang X, Wang W, Li P, Wang X, Ni K. High TREM2 expression correlates with poor prognosis in gastric cancer. Hum Pathol 2018; 72:91-99. [DOI: 10.1016/j.humpath.2017.10.026] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2017] [Revised: 10/18/2017] [Accepted: 10/20/2017] [Indexed: 12/16/2022]
|
87
|
Mecca C, Giambanco I, Donato R, Arcuri C. Microglia and Aging: The Role of the TREM2-DAP12 and CX3CL1-CX3CR1 Axes. Int J Mol Sci 2018; 19:E318. [PMID: 29361745 PMCID: PMC5796261 DOI: 10.3390/ijms19010318] [Citation(s) in RCA: 156] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 01/17/2018] [Accepted: 01/18/2018] [Indexed: 12/21/2022] Open
Abstract
Depending on the species, microglial cells represent 5-20% of glial cells in the adult brain. As the innate immune effector of the brain, microglia are involved in several functions: regulation of inflammation, synaptic connectivity, programmed cell death, wiring and circuitry formation, phagocytosis of cell debris, and synaptic pruning and sculpting of postnatal neural circuits. Moreover, microglia contribute to some neurodevelopmental disorders such as Nasu-Hakola disease (NHD), and to aged-associated neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), and others. There is evidence that human and rodent microglia may become senescent. This event determines alterations in the microglia activation status, associated with a chronic inflammation phenotype and with the loss of neuroprotective functions that lead to a greater susceptibility to the neurodegenerative diseases of aging. In the central nervous system (CNS), Triggering Receptor Expressed on Myeloid Cells 2-DNAX activation protein 12 (TREM2-DAP12) is a signaling complex expressed exclusively in microglia. As a microglial surface receptor, TREM2 interacts with DAP12 to initiate signal transduction pathways that promote microglial cell activation, phagocytosis, and microglial cell survival. Defective TREM2-DAP12 functions play a central role in the pathogenesis of several diseases. The CX3CL1 (fractalkine)-CX3CR1 signaling represents the most important communication channel between neurons and microglia. The expression of CX3CL1 in neurons and of its receptor CX3CR1 in microglia determines a specific interaction, playing fundamental roles in the regulation of the maturation and function of these cells. Here, we review the role of the TREM2-DAP12 and CX3CL1-CX3CR1 axes in aged microglia and the involvement of these pathways in physiological CNS aging and in age-associated neurodegenerative diseases.
Collapse
Affiliation(s)
- Carmen Mecca
- Department of Experimental Medicine, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132 Perugia, Italy.
| | - Ileana Giambanco
- Department of Experimental Medicine, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132 Perugia, Italy.
| | - Rosario Donato
- Department of Experimental Medicine, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132 Perugia, Italy.
- Centro Universitario per la Ricerca sulla Genomica Funzionale, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132 Perugia, Italy.
| | - Cataldo Arcuri
- Department of Experimental Medicine, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132 Perugia, Italy.
| |
Collapse
|
88
|
Su WH, Shi ZH, Liu SL, Wang XD, Liu S, Ji Y. The rs75932628 and rs2234253 polymorphisms of the TREM2 gene were associated with susceptibility to frontotemporal lobar degeneration in Caucasian populations. Ann Hum Genet 2018; 82:177-185. [PMID: 29322490 DOI: 10.1111/ahg.12241] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Revised: 09/30/2017] [Accepted: 11/29/2017] [Indexed: 12/29/2022]
Abstract
Polymorphisms of the triggering receptor expressed on myeloid cells 2 (TREM2) gene have been reported to be potentially associated with the risks of developing frontotemporal lobar degeneration (FTLD), with inconsistent conclusions. This study aims to comprehensively investigate the potential role of TREM2 variants in FTLD risks via a meta-analysis. We included a total of eight eligible articles. For TREM2 rs75932628, we observed a significantly increased FTLD risk in the models of T vs. C [Association Test, odds ratio (OR) = 2.43, 95% confidence interval (CI) = 1.43∼4.14, P = 0.001], CT vs. CC (OR = 2.27, 95% CI = 1.39∼3.71, P = 0.001), CT + TT vs. CC (OR = 2.27, 95% CI = 1.38∼3.71, P = 0.001), and Carrier T vs. C (OR = 2.26, 95% CI = 1.38∼3.69, P = 0.001). Similarly, we observed positive results for TREM2 rs2234253 in all of the genetic models (all OR > 1, P = 0.030). Nevertheless, we did not observe any statistical difference between the case and control groups in the pooled analyses of TREM2 rs142232675 and rs143332484 (all P > 0.05). Our findings identified the rs75932628 and rs2234253 polymorphisms of the TREM2 gene as risk factors for FTLD in Caucasian populations.
Collapse
Affiliation(s)
- Wen-Hua Su
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin, China.,Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Huanhu Hospital, Tianjin, China
| | - Zhi-Hong Shi
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin, China.,Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Huanhu Hospital, Tianjin, China
| | - Shu-Ling Liu
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin, China.,Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Huanhu Hospital, Tianjin, China
| | - Xiao-Dan Wang
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin, China.,Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Huanhu Hospital, Tianjin, China
| | - Shuai Liu
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin, China.,Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Huanhu Hospital, Tianjin, China
| | - Yong Ji
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin, China.,Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Huanhu Hospital, Tianjin, China
| |
Collapse
|
89
|
Scott-Hewitt NJ, Folts CJ, Hogestyn JM, Piester G, Mayer-Pröschel M, Noble MD. Heterozygote galactocerebrosidase (GALC) mutants have reduced remyelination and impaired myelin debris clearance following demyelinating injury. Hum Mol Genet 2018; 26:2825-2837. [PMID: 28575206 DOI: 10.1093/hmg/ddx153] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 04/07/2017] [Indexed: 12/20/2022] Open
Abstract
Genome-wide association studies are identifying multiple genetic risk factors for several diseases, but the functional role of these changes remains mostly unknown. Variants in the galactocerebrosidase (GALC) gene, for example, were identified as a risk factor for Multiple Sclerosis (MS); however, the potential biological relevance of GALC variants to MS remains elusive. We found that heterozygote GALC mutant mice have reduced myelin debris clearance and diminished remyelination after a demyelinating insult. We found no histological or behavioral differences between adult wild-type and GALC +/- animals under normal conditions. Following exposure to the demyelinating agent cuprizone, however, GALC +/- animals had significantly reduced remyelination during recovery. In addition, the microglial phagocytic response and elevation of Trem2, both necessary for clearing damaged myelin, were markedly reduced in GALC +/- animals. These altered responses could be corrected in vitro by treatment with NKH-477, a compound discovered as protective in our previous studies on Krabbe disease, which is caused by mutations in both GALC alleles. Our data are the first to show remyelination defects in individuals with a single mutant GALC allele, suggesting such carriers may have increased vulnerability to myelin damage following injury or disease due to inefficient myelin debris clearance. We thus provide a potential functional link between GALC variants and increased MS susceptibility, particularly due to the failure of remyelination associated with progressive MS. Finally, this work demonstrates that genetic variants identified through genome-wide association studies may contribute significantly to complex diseases, not by driving initial symptoms, but by altering repair mechanisms.
Collapse
Affiliation(s)
- Nicole J Scott-Hewitt
- Department of Biomedical Genetics, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Christopher J Folts
- Department of Biomedical Genetics, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Jessica M Hogestyn
- Department of Biomedical Genetics, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Gavin Piester
- Department of Biochemistry, University of Rochester, Rochester, NY 14642, USA
| | - Margot Mayer-Pröschel
- Department of Biomedical Genetics, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Mark D Noble
- Department of Biomedical Genetics, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| |
Collapse
|
90
|
Genetic Risk Factors for Complex Forms of Alzheimer’s Disease. NEURODEGENER DIS 2018. [DOI: 10.1007/978-3-319-72938-1_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
|
91
|
Scott-Hewitt NJ, Folts CJ, Noble MD. Heterozygous carriers of galactocerebrosidase mutations that cause Krabbe disease have impaired microglial function and defective repair of myelin damage. Neural Regen Res 2018; 13:393-401. [PMID: 29623914 PMCID: PMC5900492 DOI: 10.4103/1673-5374.228712] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
This review addresses two puzzling findings related to mutations in galactocerebrosidase (GALC) that cause Krabbe disease (KD), a severe lysosomal storage disorder characterized by extensive myelin damage in children with mutations in both GALC alleles. First, heterozygous carriers of KD-causing mutations, which include the biological parents of children with KD, exhibit increased risk for developing other diseases. Second, variants in the GALC locus increase the risk of developing multiple sclerosis (MS), another disease characterized by extensive myelin damage. What explains these correlations? In studies on cuprizone-induced myelin damage in heterozygous (GALC+/–) mice carrying one copy of a mutation that causes KD-like disease, the extent of damage was similar in GALC+/– and wild-type (WT) mice. In contrast, GALC+/- mice had striking defects in repair of cuprizone-induced damage. We further found unexpected microglial defects in myelin debris clearance and in the ability to up-regulate the Trem2 microglial protein critical for debris uptake. These defects were rescued by exposure to a lysosomal re-acidifying drug discovered in our studies on KD, and which provides multiple clinically relevant benefits in the twitcher (GALC+/–) mouse model of KD. Thus, heterozygous GALC mutations cause effects on biological function that may help to understand the increased disease risk in heterozygous carriers of such mutations and to understand why GALC variations increase the risk of MS. Our findings indicate that while some genetic risk factors may contribute to complex diseases by increasing the risk of tissue damage, others may do so by compromising tissue repair.
Collapse
Affiliation(s)
- Nicole J Scott-Hewitt
- Department of Biomedical Genetics, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Christopher J Folts
- Department of Biomedical Genetics, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Mark D Noble
- Department of Biomedical Genetics, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| |
Collapse
|
92
|
Let's make microglia great again in neurodegenerative disorders. J Neural Transm (Vienna) 2017; 125:751-770. [PMID: 29027011 DOI: 10.1007/s00702-017-1792-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 09/18/2017] [Indexed: 12/12/2022]
Abstract
All of the common neurodegenerative disorders-Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis and prion diseases-are characterized by accumulation of misfolded proteins that trigger activation of microglia; brain-resident mononuclear phagocytes. This chronic form of neuroinflammation is earmarked by increased release of myriad cytokines and chemokines in patient brains and biofluids. Microglial phagocytosis is compromised early in the disease process, obfuscating clearance of abnormal proteins. This review identifies immune pathologies shared by the major neurodegenerative disorders. The overarching concept is that aberrant innate immune pathways can be targeted for return to homeostasis in hopes of coaxing microglia into clearing neurotoxic misfolded proteins.
Collapse
|
93
|
Discoidin domain receptor inhibition reduces neuropathology and attenuates inflammation in neurodegeneration models. J Neuroimmunol 2017; 311:1-9. [PMID: 28863860 DOI: 10.1016/j.jneuroim.2017.07.009] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 06/23/2017] [Accepted: 07/12/2017] [Indexed: 01/04/2023]
Abstract
The role of cell surface tyrosine kinase collagen-activated receptors known as discoidin domain receptors (DDRs) is unknown in neurodegenerative diseases. We detect up-regulation in DDRs level in post-mortem Alzheimer and Parkinson brains. Lentiviral shRNA knockdown of DDR1 and DDR2 reduces the levels of α-synuclein, tau, and β-amyloid and prevents cell loss in vivo and in vitro. DDR1 and DDR2 knockdown alters brain immunity and significantly reduces the level of triggering receptor expressed on myeloid cells (TREM)-2 and microglia. These studies suggest that DDR1 and DDR2 inhibition is a potential target to clear neurotoxic proteins and reduce inflammation in neurodegeneration.
Collapse
|
94
|
Jay TR, von Saucken VE, Landreth GE. TREM2 in Neurodegenerative Diseases. Mol Neurodegener 2017; 12:56. [PMID: 28768545 PMCID: PMC5541421 DOI: 10.1186/s13024-017-0197-5] [Citation(s) in RCA: 263] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 07/20/2017] [Indexed: 12/12/2022] Open
Abstract
TREM2 variants have been identified as risk factors for Alzheimer's disease (AD) and other neurodegenerative diseases (NDDs). Because TREM2 encodes a receptor exclusively expressed on immune cells, identification of these variants conclusively demonstrates that the immune response can play an active role in the pathogenesis of NDDs. These TREM2 variants also confer the highest risk for developing Alzheimer's disease of any risk factor identified in nearly two decades, suggesting that understanding more about TREM2 function could provide key insights into NDD pathology and provide avenues for novel immune-related NDD biomarkers and therapeutics. The expression, signaling and function of TREM2 in NDDs have been extensively investigated in an effort to understand the role of immune function in disease pathogenesis and progression. We provide a comprehensive review of our current understanding of TREM2 biology, including new insights into the regulation of TREM2 expression, and TREM2 signaling and function across NDDs. While many open questions remain, the current body of literature provides clarity on several issues. While it is still often cited that TREM2 expression is decreased by pro-inflammatory stimuli, it is now clear that this is true in vitro, but inflammatory stimuli in vivo almost universally increase TREM2 expression. Likewise, while TREM2 function is classically described as promoting an anti-inflammatory phenotype, more than half of published studies demonstrate a pro-inflammatory role for TREM2, suggesting that its role in inflammation is much more complex. Finally, these components of TREM2 biology are applied to a discussion of how TREM2 impacts NDD pathologies and the latest assessment of how these findings might be applied to immune-directed clinical biomarkers and therapeutics.
Collapse
Affiliation(s)
- Taylor R. Jay
- Department of Neurosciences, Case Western Reserve University, School of Medicine, 10900 Euclid Avenue, Cleveland, OH 44106 USA
| | - Victoria E. von Saucken
- Department of Neurosciences, Case Western Reserve University, School of Medicine, 10900 Euclid Avenue, Cleveland, OH 44106 USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, 320 W 15th Street, Indianapolis, IN 46202 USA
| | - Gary E. Landreth
- Department of Neurosciences, Case Western Reserve University, School of Medicine, 10900 Euclid Avenue, Cleveland, OH 44106 USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, 320 W 15th Street, Indianapolis, IN 46202 USA
| |
Collapse
|
95
|
Disease Progression-Dependent Effects of TREM2 Deficiency in a Mouse Model of Alzheimer's Disease. J Neurosci 2017; 37:637-647. [PMID: 28100745 DOI: 10.1523/jneurosci.2110-16.2016] [Citation(s) in RCA: 306] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 11/30/2016] [Accepted: 12/05/2016] [Indexed: 11/21/2022] Open
Abstract
Neuroinflammation is an important contributor to Alzheimer's disease (AD) pathogenesis, as underscored by the recent identification of immune-related genetic risk factors for AD, including coding variants in the gene TREM2 (triggering receptor expressed on myeloid cells 2). Understanding TREM2 function promises to provide important insights into how neuroinflammation contributes to AD pathology. However, studies so far have produced seemingly conflicting results, with reports that amyloid pathology can be both decreased and increased in TREM2-deficient AD mouse models. In this study, we unify these previous findings by demonstrating that TREM2 deficiency ameliorates amyloid pathology early, but exacerbates it late in disease progression in the APPPS1-21 mouse model of AD. We also demonstrate that TREM2 deficiency decreases plaque-associated myeloid cell accumulation by reducing cell proliferation, specifically late in pathology. In addition, TREM2 deficiency reduces myeloid cell internalization of amyloid throughout pathology, but decreases inflammation-related gene transcript levels selectively late in disease progression. Together, these results suggest that TREM2 plays distinct functional roles at different stages in AD pathology. SIGNIFICANCE STATEMENT Alzheimer's disease (AD) is a devastating neurodegenerative disorder and there are currently no effective treatments that modify disease progression. However, the recent identification of genetic risk factors for AD promises to provide new insight into AD biology and possible new therapeutic targets. Among these risk factors, variants in the gene TREM2 (triggering receptor expressed on myeloid cells 2) confer greatly elevated risk for developing the disease. We demonstrate that TREM2 deficiency has opposing effects on AD-related pathologies at early and late stages of disease progression, unifying previous work in the field. In addition, we examine how TREM2 affects the function of the brain immune cell populations in which it is expressed throughout disease progression to understand possible mechanisms underlying its differential impacts on pathology.
Collapse
|
96
|
Kopper TJ, Gensel JC. Myelin as an inflammatory mediator: Myelin interactions with complement, macrophages, and microglia in spinal cord injury. J Neurosci Res 2017; 96:969-977. [PMID: 28696010 DOI: 10.1002/jnr.24114] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 06/02/2017] [Accepted: 06/19/2017] [Indexed: 12/24/2022]
Abstract
Spinal cord injury (SCI) triggers chronic intraspinal inflammation consisting of activated resident and infiltrating immune cells (especially microglia/macrophages). The environmental factors contributing to this protracted inflammation are not well understood; however, myelin lipid debris is a hallmark of SCI. Myelin is also a potent macrophage stimulus and target of complement-mediated clearance and inflammation. The downstream effects of these neuroimmune interactions have the potential to contribute to ongoing pathology or facilitate repair. This depends in large part on whether myelin drives pathological or reparative macrophage activation states, commonly referred to as M1 (proinflammatory) or M2 (alternatively) macrophages, respectively. Here we review the processes by which myelin debris may be cleared through macrophage surface receptors and the complement system, how this differentially influences macrophage and microglial activation states, and how the cellular functions of these myelin macrophages and complement proteins contribute to chronic inflammation and secondary injury after SCI.
Collapse
Affiliation(s)
- Timothy J Kopper
- Spinal Cord and Brain Injury Research Center, Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky
| | - John C Gensel
- Spinal Cord and Brain Injury Research Center, Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
97
|
Wang MD, Little J, Gomes J, Cashman NR, Krewski D. Identification of risk factors associated with onset and progression of amyotrophic lateral sclerosis using systematic review and meta-analysis. Neurotoxicology 2017; 61:101-130. [DOI: 10.1016/j.neuro.2016.06.015] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 06/29/2016] [Indexed: 12/11/2022]
|
98
|
Pottier C, Ravenscroft TA, Sanchez-Contreras M, Rademakers R. Genetics of FTLD: overview and what else we can expect from genetic studies. J Neurochem 2017; 138 Suppl 1:32-53. [PMID: 27009575 DOI: 10.1111/jnc.13622] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 02/26/2016] [Accepted: 03/18/2016] [Indexed: 12/11/2022]
Abstract
Frontotemporal lobar degeneration (FTLD) comprises a highly heterogeneous group of disorders clinically associated with behavioral and personality changes, language impairment, and deficits in executive functioning, and pathologically associated with degeneration of frontal and temporal lobes. Some patients present with motor symptoms including amyotrophic lateral sclerosis. Genetic research over the past two decades in FTLD families led to the identification of three common FTLD genes (microtubule-associated protein tau, progranulin, and chromosome 9 open reading frame 72) and a small number of rare FTLD genes, explaining the disease in almost all autosomal dominant FTLD families but only a minority of apparently sporadic patients or patients in whom the family history is less clear. Identification of additional FTLD (risk) genes is therefore highly anticipated, especially with the emerging use of next-generation sequencing. Common variants in the transmembrane protein 106 B were identified as a genetic risk factor of FTLD and disease modifier in patients with known mutations. This review summarizes for each FTLD gene what we know about the type and frequency of mutations, their associated clinical and pathological features, and potential disease mechanisms. We also provide an overview of emerging disease pathways encompassing multiple FTLD genes. We further discuss how FTLD specific issues, such as disease heterogeneity, the presence of an unclear family history and the possible role of an oligogenic basis of FTLD, can pose challenges for future FTLD gene identification and risk assessment of specific variants. Finally, we highlight emerging clinical, genetic, and translational research opportunities that lie ahead. Genetic research led to the identification of three common FTLD genes with rare variants (MAPT, GRN, and C9orf72) and a small number of rare genes. Efforts are now ongoing, which aimed at the identification of rare variants with high risk and/or low frequency variants with intermediate effect. Common risk variants have also been identified, such as TMEM106B. This review discusses the current knowledge on FTLD genes and the emerging disease pathways encompassing multiple FTLD genes.
Collapse
Affiliation(s)
- Cyril Pottier
- Mayo Clinic Jacksonville, Department of Neuroscience, Jacksonville, FL, USA
| | | | | | - Rosa Rademakers
- Mayo Clinic Jacksonville, Department of Neuroscience, Jacksonville, FL, USA
| |
Collapse
|
99
|
Karaca I, Wagner H, Ramirez A. Suche nach Risikogenen bei der Alzheimer-Erkrankung. DER NERVENARZT 2017; 88:744-750. [DOI: 10.1007/s00115-017-0354-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
100
|
Yeh FL, Hansen DV, Sheng M. TREM2, Microglia, and Neurodegenerative Diseases. Trends Mol Med 2017; 23:512-533. [DOI: 10.1016/j.molmed.2017.03.008] [Citation(s) in RCA: 180] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Revised: 02/15/2017] [Accepted: 03/26/2017] [Indexed: 01/17/2023]
|