1
|
Moon HJ, Luo Y, Chugh D, Zhao L. Human apolipoprotein E glycosylation and sialylation: from structure to function. Front Mol Neurosci 2024; 17:1399965. [PMID: 39169951 PMCID: PMC11335735 DOI: 10.3389/fnmol.2024.1399965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 06/28/2024] [Indexed: 08/23/2024] Open
Abstract
Human apolipoprotein E (ApoE) was first identified as a polymorphic gene in the 1970s; however, the genetic association of ApoE genotypes with late-onset sporadic Alzheimer's disease (sAD) was only discovered 20 years later. Since then, intensive research has been undertaken to understand the molecular effects of ApoE in the development of sAD. Despite three decades' worth of effort and over 10,000 papers published, the greatest mystery in the ApoE field remains: human ApoE isoforms differ by only one or two amino acid residues; what is responsible for their significantly distinct roles in the etiology of sAD, with ApoE4 conferring the greatest genetic risk for sAD whereas ApoE2 providing exceptional neuroprotection against sAD. Emerging research starts to point to a novel and compelling hypothesis that the sialoglycans posttranslationally appended to human ApoE may serve as a critical structural modifier that alters the biology of ApoE, leading to the opposing impacts of ApoE isoforms on sAD and likely in the peripheral systems as well. ApoE has been shown to be posttranslationally glycosylated in a species-, tissue-, and cell-specific manner. Human ApoE, particularly in brain tissue and cerebrospinal fluid (CSF), is highly glycosylated, and the glycan chains are exclusively attached via an O-linkage to serine or threonine residues. Moreover, studies have indicated that human ApoE glycans undergo sialic acid modification or sialylation, a structural alteration found to be more prominent in ApoE derived from the brain and CSF than plasma. However, whether the sialylation modification of human ApoE has a biological role is largely unexplored. Our group recently first reported that the three major isoforms of human ApoE in the brain undergo varying degrees of sialylation, with ApoE2 exhibiting the most abundant sialic acid modification, whereas ApoE4 is the least sialylated. Our findings further indicate that the sialic acid moiety on human ApoE glycans may serve as a critical modulator of the interaction of ApoE with amyloid β (Aβ) and downstream Aβ pathogenesis, a prominent pathologic feature in AD. In this review, we seek to provide a comprehensive summary of this exciting and rapidly evolving area of ApoE research, including the current state of knowledge and opportunities for future exploration.
Collapse
Affiliation(s)
- Hee-Jung Moon
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, United States
| | - Yan Luo
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, United States
| | - Diksha Chugh
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, United States
| | - Liqin Zhao
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, United States
- Neuroscience Graduate Program, University of Kansas, Lawrence, KS, United States
| |
Collapse
|
2
|
Yassaghi Y, Nazerian Y, Ghasemi M, Nazerian A, Sayehmiri F, Perry G, Gholami Pourbadie H. Microglial modulation as a therapeutic strategy in Alzheimer's disease: Focus on microglial preconditioning approaches. J Cell Mol Med 2024; 28:e18554. [PMID: 39103747 DOI: 10.1111/jcmm.18554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 06/15/2024] [Accepted: 07/05/2024] [Indexed: 08/07/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive disease that causes an impairment of learning and memory. Despite the highly complex pathogenesis of AD, amyloid beta (Aβ) deposition and neurofibrillary tangles (NFTs) formation are the main hallmarks of AD. Neuroinflammation also has a crucial role in the development of AD. As the central nervous system's innate immune cells, microglial cells are activated in AD and induce inflammation by producing pro-inflammatory mediators. However, microglial activation is not always deleterious. M2-activated microglial cells are considered anti-inflammatory cells, which develop neuroprotection. Various approaches are proposed for managing AD, yet no effective therapy is available for this disorder. Considering the potential protective role of M2 microglia in neurodegenerative disorders and the improvement of these disorders by preconditioning approaches, it can be suggested that preconditioning of microglial cells may be beneficial for managing AD progression. Therefore, this study review microglial preconditioning approaches for preventing and improving AD.
Collapse
Affiliation(s)
- Younes Yassaghi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Yasaman Nazerian
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mobina Ghasemi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Fatemeh Sayehmiri
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - George Perry
- Department of Neuroscience, Development, and Regenerative Biology, University of Texas at San Antonio, San Antonio, Texas, USA
| | | |
Collapse
|
3
|
Faraji P, Kühn H, Ahmadian S. Multiple Roles of Apolipoprotein E4 in Oxidative Lipid Metabolism and Ferroptosis During the Pathogenesis of Alzheimer's Disease. J Mol Neurosci 2024; 74:62. [PMID: 38958788 PMCID: PMC11222241 DOI: 10.1007/s12031-024-02224-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 04/14/2024] [Indexed: 07/04/2024]
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disease worldwide and has a great socio-economic impact. Modified oxidative lipid metabolism and dysregulated iron homeostasis have been implicated in the pathogenesis of this disorder, but the detailed pathophysiological mechanisms still remain unclear. Apolipoprotein E (APOE) is a lipid-binding protein that occurs in large quantities in human blood plasma, and a polymorphism of the APOE gene locus has been identified as risk factors for AD. The human genome involves three major APOE alleles (APOE2, APOE3, APOE4), which encode for three subtly distinct apolipoprotein E isoforms (APOE2, APOE3, APOE4). The canonic function of these apolipoproteins is lipid transport in blood and brain, but APOE4 allele carriers have a much higher risk for AD. In fact, about 60% of clinically diagnosed AD patients carry at least one APOE4 allele in their genomes. Although the APOE4 protein has been implicated in pathophysiological key processes of AD, such as extracellular beta-amyloid (Aβ) aggregation, mitochondrial dysfunction, neuroinflammation, formation of neurofibrillary tangles, modified oxidative lipid metabolism, and ferroptotic cell death, the underlying molecular mechanisms are still not well understood. As for all mammalian cells, iron plays a crucial role in neuronal functions and dysregulation of iron homeostasis has also been implicated in the pathogenesis of AD. Imbalances in iron homeostasis and impairment of the hydroperoxy lipid-reducing capacity induce cellular dysfunction leading to neuronal ferroptosis. In this review, we summarize the current knowledge on APOE4-related oxidative lipid metabolism and the potential role of ferroptosis in the pathogenesis of AD. Pharmacological interference with these processes might offer innovative strategies for therapeutic interventions.
Collapse
Affiliation(s)
- Parisa Faraji
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
- Department of Biochemistry, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Hartmut Kühn
- Department of Biochemistry, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany.
| | - Shahin Ahmadian
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran.
| |
Collapse
|
4
|
Ghosh S, Tamilselvi S, Williams C, Jayaweera SW, Iashchishyn IA, Šulskis D, Gilthorpe JD, Olofsson A, Smirnovas V, Svedružić ŽM, Morozova-Roche LA. ApoE Isoforms Inhibit Amyloid Aggregation of Proinflammatory Protein S100A9. Int J Mol Sci 2024; 25:2114. [PMID: 38396791 PMCID: PMC10889306 DOI: 10.3390/ijms25042114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 01/31/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Increasing evidence suggests that the calcium-binding and proinflammatory protein S100A9 is an important player in neuroinflammation-mediated Alzheimer's disease (AD). The amyloid co-aggregation of S100A9 with amyloid-β (Aβ) is an important hallmark of this pathology. Apolipoprotein E (ApoE) is also known to be one of the important genetic risk factors of AD. ApoE primarily exists in three isoforms, ApoE2 (Cys112/Cys158), ApoE3 (Cys112/Arg158), and ApoE4 (Arg112/Arg158). Even though the difference lies in just two amino acid residues, ApoE isoforms produce differential effects on the neuroinflammation and activation of the microglial state in AD. Here, we aim to understand the effect of the ApoE isoforms on the amyloid aggregation of S100A9. We found that both ApoE3 and ApoE4 suppress the aggregation of S100A9 in a concentration-dependent manner, even at sub-stoichiometric ratios compared to S100A9. These interactions lead to a reduction in the quantity and length of S100A9 fibrils. The inhibitory effect is more pronounced if ApoE isoforms are added in the lipid-free state versus lipidated ApoE. We found that, upon prolonged incubation, S100A9 and ApoE form low molecular weight complexes with stochiometric ratios of 1:1 and 2:1, which remain stable under SDS-gel conditions. These complexes self-assemble also under the native conditions; however, their interactions are transient, as revealed by glutaraldehyde cross-linking experiments and molecular dynamics (MD) simulation. MD simulation demonstrated that the lipid-binding C-terminal domain of ApoE and the second EF-hand calcium-binding motif of S100A9 are involved in these interactions. We found that amyloids of S100A9 are cytotoxic to neuroblastoma cells, and the presence of either ApoE isoforms does not change the level of their cytotoxicity. A significant inhibitory effect produced by both ApoE isoforms on S100A9 amyloid aggregation can modulate the amyloid-neuroinflammatory cascade in AD.
Collapse
Affiliation(s)
- Shamasree Ghosh
- Department of Medical Biochemistry and Biophysics, Umeå University, SE-90187 Umeå, Sweden; (S.G.); (S.T.); (I.A.I.)
| | - Shanmugam Tamilselvi
- Department of Medical Biochemistry and Biophysics, Umeå University, SE-90187 Umeå, Sweden; (S.G.); (S.T.); (I.A.I.)
| | - Chloe Williams
- Department of Medical and Translational Biology, Umeå University, SE-90187 Umeå, Sweden; (C.W.); (J.D.G.)
| | - Sanduni W. Jayaweera
- Department of Clinical Microbiology, Umeå University, SE-90187 Umeå, Sweden; (S.W.J.); (A.O.)
| | - Igor A. Iashchishyn
- Department of Medical Biochemistry and Biophysics, Umeå University, SE-90187 Umeå, Sweden; (S.G.); (S.T.); (I.A.I.)
| | - Darius Šulskis
- Institute of Biotechnology, Life Sciences Center, Vilnius University, LT-10257 Vilnius, Lithuania; (D.Š.); (V.S.)
| | - Jonathan D. Gilthorpe
- Department of Medical and Translational Biology, Umeå University, SE-90187 Umeå, Sweden; (C.W.); (J.D.G.)
| | - Anders Olofsson
- Department of Clinical Microbiology, Umeå University, SE-90187 Umeå, Sweden; (S.W.J.); (A.O.)
| | - Vytautas Smirnovas
- Institute of Biotechnology, Life Sciences Center, Vilnius University, LT-10257 Vilnius, Lithuania; (D.Š.); (V.S.)
| | | | - Ludmilla A. Morozova-Roche
- Department of Medical Biochemistry and Biophysics, Umeå University, SE-90187 Umeå, Sweden; (S.G.); (S.T.); (I.A.I.)
| |
Collapse
|
5
|
Wang X, Li H, Sheng Y, He B, Liu Z, Li W, Yu S, Wang J, Zhang Y, Chen J, Qin L, Meng X. The function of sphingolipids in different pathogenesis of Alzheimer's disease: A comprehensive review. Biomed Pharmacother 2024; 171:116071. [PMID: 38183741 DOI: 10.1016/j.biopha.2023.116071] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 12/11/2023] [Accepted: 12/21/2023] [Indexed: 01/08/2024] Open
Abstract
Sphingolipids (SPLs) represent a highly diverse and structurally complex lipid class. The discussion of SPL metabolism-related issues is of importance in understanding the neuropathological progression of Alzheimer's disease (AD). AD is characterized by the accumulation of extracellular deposits of the amyloid β-peptide (Aβ) and intraneuronal aggregates of the microtubule-associated protein tau. Critical roles of Aβ oligomer deposited and ganglioside GM1 could be formed as "seed" from insoluble GAβ polymer in initiating the pathogenic process, while tau might also mediate SPLs and their toxicity. The interaction between ceramide and α-Synuclein (α-Syn) accelerates the aggregation of ferroptosis and exacerbates the pathogenesis of AD. For instance, reducing the levels of SPLs can mitigate α-Syn accumulation and inhibit AD progression. Meanwhile, loss of SPLs may inhibit the expression of APOE4 and confer protection against AD, while the loss of APOE4 expression also disrupts SPLs homeostasis. Moreover, the heightened activation of sphingomyelinase promotes the ferroptosis signaling pathway, leading to exacerbated AD symptoms. Ferroptosis plays a vital role in the pathological progression of AD by influencing Aβ, tau, APOE, and α-Syn. Conversely, the development of AD also exacerbates the manifestation of ferroptosis and SPLs. We are compiling the emerging techniques (Derivatization and IM-MS) of sphingolipidomics, to overcome the challenges of AD diagnosis and treatment. In this review, we examined the intricate neuro-mechanistic interactions between SPLs and Aβ, tau, α-Syn, APOE, and ferroptosis, mediating the onset of AD. Furthermore, our findings highlight the potential of targeting SPLs as underexplored avenue for devising innovative therapeutic strategies against AD.
Collapse
Affiliation(s)
- Xinyi Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China
| | - Huaqiang Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China
| | - Yunjie Sheng
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China
| | - Bingqian He
- Academy of Chinese Medical Science, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China
| | - Zeying Liu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China
| | - Wanli Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China
| | - Shujie Yu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China
| | - Jiajing Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China
| | - Yixin Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China
| | - Jianyu Chen
- Fujian University of Traditional Chinese Medicine, School of Pharmacy, Fuzhou, Fujian 350122, PR China.
| | - Luping Qin
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China.
| | - Xiongyu Meng
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, Zhejiang Province, PR China.
| |
Collapse
|
6
|
Ramachandran AK, Das S, Shenoy GG, Mudgal J, Joseph A. Relation between Apolipoprotein E in Alzheimer's Disease and SARS-CoV-2 and their Treatment Strategy: A Review. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:9-20. [PMID: 36573058 DOI: 10.2174/1871527322666221226145141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 10/27/2022] [Accepted: 11/01/2022] [Indexed: 12/28/2022]
Abstract
COVID-19, which primarily affects the pulmonary system, turned out to be a global pandemic, whereas the effects on other systems are still unknown. SARS-CoV-2, binds to angiotensinconverting enzyme 2 (ACE2) receptors in the lungs, causing pneumonia-like symptoms. The same ACE receptors are also present in organs other than the lungs. Therefore, there is a need to study the impact of coronavirus on other human body organs. Recently, UK Biobank reports on the genetic risk factor of the virus attack. A double mutation in the apolipoprotein E (APOE4) allele has shown a significant role in COVID-19. The same APOE4 mutation has already been proven to hold a key role in developing early-onset Alzheimer's disease (EOAD). Despite this data, Alzheimer's disease is believed to be a comorbidity of COVID-19. Previous virus attacks on the same viral family, Coronaviridae, produced neurological effects like neurodegeneration, neuronal inflammation, and other central nervous system-related dysfunctions. Since the long-term implications of COVID-19 are unknown, more research into the impact of the virus on the central nervous system is needed. Both COVID-19 and AD share a common genetic factor, so that AD patients may have a greater risk of SARS-CoV-2. Here, in this review, we have briefly discussed the role of APOE4 in the pathogenesis of AD and SARS-CoV-2, along with their treatment strategy, current scenario, and possible future directions.
Collapse
Affiliation(s)
- Anu Kunnath Ramachandran
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Subham Das
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
- Manipal McGill Centre for Infectious Diseases, Prasanna School of Public Health, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Gurupur Gautham Shenoy
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Jayesh Mudgal
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Alex Joseph
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| |
Collapse
|
7
|
Giil LM, Hanseth S, Bojovic O, Nordrehaug JE, Ueland PM, Meyer K, Tell GS. The inverse association between the apolipoprotein E ε4 allele and C-reactive protein levels is stronger in persons with obesity and diabetes. Scand J Immunol 2024; 99:e13323. [PMID: 37801334 DOI: 10.1111/sji.13323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 08/09/2023] [Accepted: 08/10/2023] [Indexed: 10/07/2023]
Abstract
BACKGROUND C-reactive protein (CRP) is lower in patients who carry the apolipoprotein E epsilon 4 allele variant (APOEε4) of the APOE gene. This could however be explained by other factors observed in APOEε4 carriers, such as lower body mass index (BMI), possibly less diabetes and more use of statins, all associated with CRP concentrations. OBJECTIVES To assess the association between CRP and APOEε4 stratified by BMI, statin use and diabetes. METHODS We included 2700 community-dwelling older adults from the Hordaland health study with genotyping of the APOE gene by a one-step polymerase chain reaction and CRP measured using immuno-MALDI-TOF MS. Differences in CRP concentrations by APOE (ε4 vs no ε4) were assessed using the Mann-Whitney U tests, also stratified by statin use, diabetes and BMI categories. Finally, we performed linear regression with log (CRP) as the outcome and APOEε4 together with statin use, diabetes, BMI and their respective interactions. RESULTS CRP was higher in APOEε4 carriers irrespective of BMI, diabetes and statin use. In APOEε4 non-carriers, CRP was elevated with diabetes and obesity as expected. However, this was attenuated or even reversed in APOEε4 carriers. Such differences were not observed for statin use. CONCLUSIONS Statin use, obesity or diabetes did not confound the known association between the APOEε4 allele and lower CRP. Our data suggest that CRP is less responsive to inflammatory cues involved in diabetes and obesity in APOEε4 carriers. Epidemiological studies should take note of these relationships, as CRP, APOEε4, diabetes and obesity are both linked to neurodegenerative and cardiovascular disease.
Collapse
Affiliation(s)
- Lasse Melvaer Giil
- Neuro-SysMed, Department of Internal Medicine, Haraldsplass Deaconess Hospital, Bergen, Norway
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Silja Hanseth
- Neuro-SysMed, Department of Internal Medicine, Haraldsplass Deaconess Hospital, Bergen, Norway
| | - Ognjen Bojovic
- Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | | | | | | | - Grethe S Tell
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway
| |
Collapse
|
8
|
Huang Z. Evidence that Alzheimer's Disease Is a Disease of Competitive Synaptic Plasticity Gone Awry. J Alzheimers Dis 2024; 99:447-470. [PMID: 38669548 PMCID: PMC11119021 DOI: 10.3233/jad-240042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Abstract
Mounting evidence indicates that a physiological function of amyloid-β (Aβ) is to mediate neural activity-dependent homeostatic and competitive synaptic plasticity in the brain. I have previously summarized the lines of evidence supporting this hypothesis and highlighted the similarities between Aβ and anti-microbial peptides in mediating cell/synapse competition. In cell competition, anti-microbial peptides deploy a multitude of mechanisms to ensure both self-protection and competitor elimination. Here I review recent studies showing that similar mechanisms are at play in Aβ-mediated synapse competition and perturbations in these mechanisms underpin Alzheimer's disease (AD). Specifically, I discuss evidence that Aβ and ApoE, two crucial players in AD, co-operate in the regulation of synapse competition. Glial ApoE promotes self-protection by increasing the production of trophic monomeric Aβ and inhibiting its assembly into toxic oligomers. Conversely, Aβ oligomers, once assembled, promote the elimination of competitor synapses via direct toxic activity and amplification of "eat-me" signals promoting the elimination of weak synapses. I further summarize evidence that neuronal ApoE may be part of a gene regulatory network that normally promotes competitive plasticity, explaining the selective vulnerability of ApoE expressing neurons in AD brains. Lastly, I discuss evidence that sleep may be key to Aβ-orchestrated plasticity, in which sleep is not only induced by Aβ but is also required for Aβ-mediated plasticity, underlining the link between sleep and AD. Together, these results strongly argue that AD is a disease of competitive synaptic plasticity gone awry, a novel perspective that may promote AD research.
Collapse
Affiliation(s)
- Zhen Huang
- Departments of Neuroscience and Neurology, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
9
|
Pini L, Salvalaggio A, Wennberg AM, Dimakou A, Matteoli M, Corbetta M. The pollutome-connectome axis: a putative mechanism to explain pollution effects on neurodegeneration. Ageing Res Rev 2023; 86:101867. [PMID: 36720351 DOI: 10.1016/j.arr.2023.101867] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 01/17/2023] [Accepted: 01/26/2023] [Indexed: 01/29/2023]
Abstract
The study of pollutant effects is extremely important to address the epochal challenges we are facing, where world populations are increasingly moving from rural to urban centers, revolutionizing our world into an urban world. These transformations will exacerbate pollution, thus highlighting the necessity to unravel its effect on human health. Epidemiological studies have reported that pollution increases the risk of neurological diseases, with growing evidence on the risk of neurodegenerative disorders. Air pollution and water pollutants are the main chemicals driving this risk. These chemicals can promote inflammation, acting in synergy with genotype vulnerability. However, the biological underpinnings of this association are unknown. In this review, we focus on the link between pollution and brain network connectivity at the macro-scale level. We provide an updated overview of epidemiological findings and studies investigating brain network changes associated with pollution exposure, and discuss the mechanistic insights of pollution-induced brain changes through neural networks. We explain, in detail, the pollutome-connectome axis that might provide the functional substrate for pollution-induced processes leading to cognitive impairment and neurodegeneration. We describe this model within the framework of two pollutants, air pollution, a widely recognized threat, and polyfluoroalkyl substances, a large class of synthetic chemicals which are currently emerging as new neurotoxic source.
Collapse
Affiliation(s)
- Lorenzo Pini
- Department of Neuroscience and Padova Neuroscience Center, University of Padova, Italy; Venetian Institute of Molecular Medicine, VIMM, Padova, Italy.
| | | | - Alexandra M Wennberg
- Unit of Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Anastasia Dimakou
- Department of Neuroscience and Padova Neuroscience Center, University of Padova, Italy
| | - Michela Matteoli
- Neuro Center, IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milano, Italy; CNR Institute of Neuroscience, Milano, Italy
| | - Maurizio Corbetta
- Department of Neuroscience and Padova Neuroscience Center, University of Padova, Italy; Venetian Institute of Molecular Medicine, VIMM, Padova, Italy
| |
Collapse
|
10
|
Zhou S, Li Y, Zhang Z, Yuan Y. An insight into the TAM system in Alzheimer's disease. Int Immunopharmacol 2023; 116:109791. [PMID: 36738678 DOI: 10.1016/j.intimp.2023.109791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/13/2023] [Accepted: 01/24/2023] [Indexed: 02/05/2023]
Abstract
The TAM receptors may help delay the progression of Alzheimer's disease (AD). AD is the most common neurodegenerative disease associated with human aging. The TAM receptors, derived from the first letter of its three constituents -Tyro3, Axl, and Mertk, are associated with immune responses, cellular differentiation and migration, and clearance of apoptotic cells and debris, with the two canonical ligands, Growth Arrest Specific 6 (Gas6) and ProS1. Several kinds of research have indicated the participation of the TAM system in AD pathology. Also, the TAMs regulate multiple features of microglia, the significant sensors of disorder in the central nervous system (CNS). In this review, we describe the biology of the TAM receptors and ligands in the CNS. Then, we discuss the relationship between the TAM system and AD, specially focusing on its functional expression in the microglia. Finally, we also summarize some agents that could interfere with the TAM signaling pathways and discuss potential difficulties and strategies for drug development.
Collapse
Affiliation(s)
- Shiqi Zhou
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Yanyan Li
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Zhao Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Yuhe Yuan
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
11
|
Zhou J, Wang Y, Huang G, Yang M, Zhu Y, Jin C, Jing D, Ji K, Shi Y. LilrB3 is a putative cell surface receptor of APOE4. Cell Res 2023; 33:116-130. [PMID: 36588123 PMCID: PMC9892561 DOI: 10.1038/s41422-022-00759-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 11/03/2022] [Indexed: 01/03/2023] Open
Abstract
The three isoforms of apolipoprotein E (APOE2, APOE3, and APOE4) only differ in two amino acid positions but exert quite different immunomodulatory effects. The underlying mechanism of such APOE isoform dependence remains enigmatic. Here we demonstrate that APOE4, but not APOE2, specifically interacts with the leukocyte immunoglobulin-like receptor B3 (LilrB3). Two discrete immunoglobin-like domains of the LilrB3 extracellular domain (ECD) recognize a positively charged surface patch on the N-terminal domain (NTD) of APOE4. The atomic structure reveals how two APOE4 molecules specifically engage two LilrB3 molecules, bringing their intracellular signaling motifs into close proximity through formation of a hetero-tetrameric complex. Consistent with our biochemical and structural analyses, APOE4, but not APOE2, activates human microglia cells (HMC3) into a pro-inflammatory state in a LilrB3-dependent manner. Together, our study identifies LilrB3 as a putative immune cell surface receptor for APOE4, but not APOE2, and may have implications for understanding the biological functions as well as disease relevance of the APOE isoforms.
Collapse
Affiliation(s)
- Jiayao Zhou
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China.
- Zhejiang Provincial Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China.
| | - Yumeng Wang
- Advanced Research Center for Biological Structure & Beijing Advanced Innovation Center for Structural Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Gaoxingyu Huang
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
- Zhejiang Provincial Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
| | - Min Yang
- Advanced Research Center for Biological Structure & Beijing Advanced Innovation Center for Structural Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Yumin Zhu
- Department of Maternal, Child & Adolescent Health, School of Public Health, Anhui Medical University, MOE Key Laboratory of Population Health Across Life Cycle, Anhui Provincial Key Laboratory of Population Health and Aristogenics, Hefei, Anhui, China
| | - Chen Jin
- Advanced Research Center for Biological Structure & Beijing Advanced Innovation Center for Structural Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Dan Jing
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
- Zhejiang Provincial Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
| | - Kai Ji
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
- Zhejiang Provincial Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
| | - Yigong Shi
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China.
- Zhejiang Provincial Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China.
- Advanced Research Center for Biological Structure & Beijing Advanced Innovation Center for Structural Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China.
| |
Collapse
|
12
|
Mhatre-Winters I, Eid A, Han Y, Tieu K, Richardson JR. Sex and APOE Genotype Alter the Basal and Induced Inflammatory States of Primary Astrocytes from Humanized Targeted Replacement Mice. ASN Neuro 2023; 15:17590914221144549. [PMID: 36604975 PMCID: PMC9982390 DOI: 10.1177/17590914221144549] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Apolipoprotein E4 (APOE4) genotype and sex are significant risk factors for Alzheimer's disease (AD), with females demonstrating increased risk modulated by APOE genotype. APOE is predominantly expressed in astrocytes, however, there is a lack of comprehensive assessments of sex differences in astrocytes stratified by APOE genotype. Here, we examined the response of mixed-sex and sex-specific neonatal APOE3 and APOE4 primary mouse astrocytes (PMA) to a cytokine mix of IL1b, TNFa, and IFNg. Pro-inflammatory and anti-inflammatory cytokine profiles were assessed by qRT-PCR and Meso Scale Discovery multiplex assay. Mixed-sex APOE4 PMA were found to have higher basal messenger RNA expression of several pro-inflammatory cytokines including Il6, Tnfa, Il1b, Mcp1, Mip1a, and Nos2 compared to APOE3 PMA, which was accompanied by increased levels of these secreted cytokines. In sex-specific cultures, basal expression of Il1b, Il6, and Nos2 was 1.5 to 2.5 fold higher in APOE4 female PMA compared to APOE4 males, with both being higher than APOE3 PMA. Similar results were found for secreted levels of these cytokines. Together, these findings indicate that APOE4 genotype and female sex, contribute to a greater inflammatory response in primary astrocytes and these data may provide a framework for investigating the mechanisms contributing to genotype and sex differences in AD-related neuroinflammation.
Collapse
Affiliation(s)
- Isha Mhatre-Winters
- Department of Environmental Health Sciences, Robert Stempel College
of Public Health and Social Work, Florida International
University, Miami, FL, USA,Department of Neurosciences, School of Biomedical Sciences, Kent
State University, Kent, OH, USA
| | - Aseel Eid
- Department of Environmental Health Sciences, Robert Stempel College
of Public Health and Social Work, Florida International
University, Miami, FL, USA
| | - Yoonhee Han
- Department of Environmental Health Sciences, Robert Stempel College
of Public Health and Social Work, Florida International
University, Miami, FL, USA
| | - Kim Tieu
- Department of Environmental Health Sciences, Robert Stempel College
of Public Health and Social Work, Florida International
University, Miami, FL, USA
| | - Jason R. Richardson
- Department of Environmental Health Sciences, Robert Stempel College
of Public Health and Social Work, Florida International
University, Miami, FL, USA,Jason R. Richardson, Department of
Environmental Health Sciences, Robert Stempel College of Public Health and
Social Work, Florida International University, Miami, FL 33199-2156, USA.
| |
Collapse
|
13
|
Sun Y, Islam S, Gao Y, Nakamura T, Zou K, Michikawa M. Apolipoprotein E4 inhibits γ-secretase activity via binding to the γ-secretase complex. J Neurochem 2022; 164:858-874. [PMID: 36582176 DOI: 10.1111/jnc.15750] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/07/2022] [Accepted: 12/16/2022] [Indexed: 12/31/2022]
Abstract
The mechanisms of amyloid accumulation in familial Alzheimer's disease (FAD) and sporadic AD (SAD) are controversial. In FAD, mutations in presenilin (PSEN) impair γ-secretase activity and lead to abnormal amyloid β-protein (Aβ) production, thereby increasing the Aβ42/40 ratio. SAD is postulated to be caused by decreased Aβ clearance of apolipoprotein E4 (APOE4), the strongest risk factor for SAD. However, whether intracellular APOE4 affects Aβ production is unclear. Using APOE3 and APOE4 knock-in (KI) mouse brain and primary cultured fibroblasts from these mice, in this study, we demonstrated that APOE3 and APOE4 bind to the γ-secretase complex and isoform-dependently regulate its activity and Aβ production. We found that Aβ40 levels and γ-secretase activity were higher in APOE knockout mouse brain than in wild-type mouse brain. APOE4-KI fibroblasts had significant lower Aβ levels and γ-secretase activity but higher Aβ42/40 ratio compared with APOE3-KI cells, indicating that APOE4-KI reduces Aβ production by inhibiting γ-secretase activity. Interestingly, the levels of γ-secretase complex bound to APOE4 are higher than those bound to APOE3, and the levels of γ-secretase complex in the brain and fibroblasts of APOE4-KI mice were higher than those of APOE3-KI mice. Taken together, our findings demonstrate that intracellular APOE4 inhibits Aβ production, more preferentially inhibits Aβ40 production, and thereby induces an increase in the Aβ42/40 ratio via binding to the γ-secretase complex. These results suggest a novel mechanism in which intracellular APOE4 contributes to the pathogenesis of SAD by inhibiting γ-secretase activity.
Collapse
Affiliation(s)
- Yang Sun
- Department of Biochemistry, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Sadequl Islam
- Department of Biochemistry, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Yuan Gao
- Department of Biochemistry, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Tomohisa Nakamura
- Department of Biochemistry, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Kun Zou
- Department of Biochemistry, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Makoto Michikawa
- Department of Biochemistry, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| |
Collapse
|
14
|
Asante I, Louie S, Yassine HN. Uncovering mechanisms of brain inflammation in Alzheimer's disease with APOE4: Application of single cell-type lipidomics. Ann N Y Acad Sci 2022; 1518:84-105. [PMID: 36200578 PMCID: PMC10092192 DOI: 10.1111/nyas.14907] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
A chronic state of unresolved inflammation in Alzheimer's disease (AD) is intrinsically involved with the remodeling of brain lipids. This review highlights the effect of carrying the apolipoprotein E ε4 allele (APOE4) on various brain cell types in promoting an unresolved inflammatory state. Among its pleotropic effects on brain lipids, we focus on APOE4's activation of Ca2+ -dependent phospholipase A2 (cPLA2) and its effects on arachidonic acid, eicosapentaenoic acid, and docosahexaenoic acid signaling cascades in the brain. During the process of neurodegeneration, various brain cell types, such as astrocytes, microglia, and neurons, together with the neurovascular unit, develop distinct inflammatory phenotypes that impact their functions and have characteristic lipidomic fingerprints. We propose that lipidomic phenotyping of single cell-types harvested from brains differing by age, sex, disease severity stage, and dietary and genetic backgrounds can be employed to probe mechanisms of neurodegeneration. A better understanding of the brain cellular inflammatory/lipidomic response promises to guide the development of nutritional and drug interventions for AD dementia.
Collapse
Affiliation(s)
- Isaac Asante
- Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Stan Louie
- School of Pharmacy, University of Southern California, Los Angeles, California, USA
| | - Hussein N Yassine
- Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
15
|
Ebright B, Assante I, Poblete RA, Wang S, Duro MV, Bennett DA, Arvanitakis Z, Louie SG, Yassine HN. Eicosanoid lipidome activation in post-mortem brain tissues of individuals with APOE4 and Alzheimer's dementia. Alzheimers Res Ther 2022; 14:152. [PMID: 36217192 PMCID: PMC9552454 DOI: 10.1186/s13195-022-01084-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 09/16/2022] [Indexed: 01/18/2023]
Abstract
BACKGROUND Chronic neuroinflammation is one of the hallmarks of late-onset Alzheimer's disease (AD) dementia pathogenesis. Carrying the apolipoprotein ε4 (APOE4) allele has been associated with an accentuated response to brain inflammation and increases the risk of AD dementia progression. Among inflammation signaling pathways, aberrant eicosanoid activation plays a prominent role in neurodegeneration. METHODS Using brains from the Religious Order Study (ROS), this study compared measures of brain eicosanoid lipidome in older persons with AD dementia to age-matched controls with no cognitive impairment (NCI), stratified by APOE genotype. RESULTS Lipidomic analysis of the dorsolateral prefrontal cortex demonstrated lower levels of omega-3 fatty acids eicosapentaenoic acid (EPA), docosapentaenoic acid (DPA), and DHA-derived neuroprotectin D1 (NPD-1) in persons with AD dementia, all of which associated with lower measures of cognitive function. A significant interaction was observed between carrying the APOE4 allele and higher levels of both pro-inflammatory lipids and pro-resolving eicosanoid lipids on measures of cognitive performance and on neuritic plaque burden. Furthermore, analysis of lipid metabolism pathways implicated activation of calcium-dependent phospholipase A2 (cPLA2), 5-lipoxygenase (5-LOX), and soluble epoxide hydrolase (sEH) enzymes. CONCLUSION These findings implicate activation of the eicosanoid lipidome in the chronic unresolved state of inflammation in AD dementia, which is increased in carriers of the APOE4 allele, and identify potential therapeutic targets for resolving this chronic inflammatory state.
Collapse
Affiliation(s)
- Brandon Ebright
- School of Pharmacy, University of Southern California, Los Angeles, USA
| | - Isaac Assante
- School of Pharmacy, University of Southern California, Los Angeles, USA
- Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Roy A Poblete
- Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Shaowei Wang
- Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Marlon V Duro
- Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Zoe Arvanitakis
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Stan G Louie
- School of Pharmacy, University of Southern California, Los Angeles, USA.
- Keck School of Medicine, University of Southern California, Los Angeles, USA.
| | - Hussein N Yassine
- Keck School of Medicine, University of Southern California, Los Angeles, USA.
| |
Collapse
|
16
|
Sex and APOE Genotype Alter the Basal and Induced Inflammatory States of Primary Microglia from APOE Targeted Replacement Mice. Int J Mol Sci 2022; 23:ijms23179829. [PMID: 36077227 PMCID: PMC9456163 DOI: 10.3390/ijms23179829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/17/2022] [Accepted: 08/19/2022] [Indexed: 11/16/2022] Open
Abstract
The sex and APOE4 genotype are significant risk factors for Alzheimer’s disease (AD); however, the mechanism(s) responsible for this interaction are still a matter of debate. Here, we assess the responses of mixed-sex and sex-specific APOE3 and APOE4 primary microglia (PMG) to lipopolysaccharide and interferon-gamma. In our investigation, inflammatory cytokine profiles were assessed by qPCR and multiplex ELISA assays. Mixed-sex APOE4 PMG exhibited higher basal mRNA expression and secreted levels of TNFa and IL1b. In sex-specific cultures, basal expression and secreted levels of IL1b, TNFa, IL6, and NOS2 were 2−3 fold higher in APOE4 female PMG compared to APOE4 males, with both higher than APOE3 cells. Following an inflammatory stimulus, the expression of pro-inflammatory cytokines and the secreted cytokine level were upregulated in the order E4 female > E4 male > E3 female > E3 male in sex-specific cultures. These data indicate that the APOE4 genotype and female sex together contribute to a greater inflammatory response in PMG isolated from targeted replacement humanized APOE mice. These data are consistent with clinical data and indicate that sex-specific PMG may provide a platform for exploring mechanisms of genotype and sex differences in AD related to neuroinflammation and neurodegeneration.
Collapse
|
17
|
Huo C, Chen MH, Hour TC, Huang LC, Fong YO, Kuo YY, Yang YH, Chuu CP. Application of Micro-Western Array for Identifying Different Serum Protein Expression Profile among Healthy Control, Alzheimer’s Disease Patients and Patients’ Adult Children. Brain Sci 2022; 12:brainsci12091134. [PMID: 36138870 PMCID: PMC9496696 DOI: 10.3390/brainsci12091134] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 08/17/2022] [Accepted: 08/23/2022] [Indexed: 12/03/2022] Open
Abstract
(1) Background: Alzheimer’s disease (AD) is the most common form of dementia. Increased levels of inflammatory proteins have been observed in brain and plasma samples of AD patients; however, it is not clear if other serum proteins correlate to the development or disease progression of AD. (2) Methods: Micro-Western Array (MWA) is a high-throughput antibody-based proteomics system which allows detection of the expression levels of 24–96 different proteins within 6–30 samples simultaneously. We applied MWA to explore potential serum protein biomarkers correlated to the development and progression of AD by examining the difference in serum protein profile of 31 healthy control (HC), 30 patients with AD and 30 patients’ adult children (ACS). (3) Results: Compared to HC, AD and ACS express similar pattern of serum proteins, including higher protein levels of ABCA1, ABCG1, SREBP1 and LXRβ but lower protein levels of ApoD, ApoE, ApoH, c_Myc, COX2 and Hippo-YAP signaling proteins. AD patients had higher serum levels of ABCG1, ApoD, ApoH, COX2, LXRα and YAP, but lower levels of ABCA1, ApoE, c_Myc, LATS1, MST1, MST2, Nanog, NFκB_p50, PPARγ and SREBP2, as compared to ACS. Pearson’s correlation analysis revealed that the protein expression level of ApoE, c_Myc, LATS1, MST2, NFκB p50, PPARγ and SREBP1 was negatively correlated to age, while that of ApoE, c_Myc, LATS1, MST1, MST2, Nanog, NFκB p50 and PPARγ was positively correlated to age. (4) Conclusions: We identified a group of serum proteins which may correlate to disease progression of AD and can be potential diagnostic serum protein biomarkers.
Collapse
Affiliation(s)
- Chieh Huo
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Ming-Hui Chen
- Neuroscience Research Center, Kaohsiung Medical University, Kaohsiung City 80708, Taiwan
| | - Tzyh-Chyuan Hour
- Neuroscience Research Center, Kaohsiung Medical University, Kaohsiung City 80708, Taiwan
- Department of Biochemistry, School of Medicine, Kaohsiung Medical University, Kaohsiung City 80708, Taiwan
| | - Ling-Chun Huang
- Department of Biochemistry, School of Medicine, Kaohsiung Medical University, Kaohsiung City 80708, Taiwan
- Department of Neurology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung City 80145, Taiwan
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung City 80756, Taiwan
| | - Yi-On Fong
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung City 80756, Taiwan
| | - Ying-Yu Kuo
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Yuan-Han Yang
- Neuroscience Research Center, Kaohsiung Medical University, Kaohsiung City 80708, Taiwan
- Department of Neurology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung City 80145, Taiwan
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung City 80756, Taiwan
- School of Post-Baccalaureate Medicine, Kaohsiung Medical University, Kaohsiung City 80708, Taiwan
- Correspondence: (Y.-H.Y.); (C.-P.C.); Tel.: +886-7-3162-158 (Y.-H.Y.); +886-37-206-166 (ext. 37300) (C.-P.C.)
| | - Chih-Pin Chuu
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli 35053, Taiwan
- Department of Life Sciences, National Central University, Taoyuan City 32031, Taiwan
- PhD Program for Aging, Graduate Institute of Basic Medical Science, China Medical University, Taichung City 40402, Taiwan
- Biotechnology Center, National Chung Hsing University, Taichung City 40227, Taiwan
- Correspondence: (Y.-H.Y.); (C.-P.C.); Tel.: +886-7-3162-158 (Y.-H.Y.); +886-37-206-166 (ext. 37300) (C.-P.C.)
| |
Collapse
|
18
|
APOE4 drives inflammation in human astrocytes via TAGLN3 repression and NF-κB activation. Cell Rep 2022; 40:111200. [PMID: 35977506 DOI: 10.1016/j.celrep.2022.111200] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 05/23/2022] [Accepted: 07/21/2022] [Indexed: 01/04/2023] Open
Abstract
Apolipoprotein E4 (APOEε4) is the major allelic risk factor for late-onset sporadic Alzheimer's disease (sAD). Inflammation is increasingly considered as critical in sAD initiation and progression. Identifying brain molecular mechanisms that could bridge these two risk factors remain unelucidated. Leveraging induced pluripotent stem cell (iPSC)-based strategies, we demonstrate that APOE controls inflammation in human astrocytes by regulating Transgelin 3 (TAGLN3) expression and, ultimately, nuclear factor κB (NF-κB) activation. We uncover that APOE4 specifically downregulates TAGLN3, involving histone deacetylases activity, which results in low-grade chronic inflammation and hyperactivated inflammatory responses. We show that APOE4 exerts a dominant negative effect to prime astrocytes toward a pro-inflammatory state that is pharmacologically reversible by TAGLN3 supplementation. We further confirm that TAGLN3 is downregulated in the brain of patients with sAD. Our findings highlight the APOE-TAGLN3-NF-κB axis regulating neuroinflammation in human astrocytes and reveal TAGLN3 as a molecular target to modulate neuroinflammation, as well as a potential biomarker for AD.
Collapse
|
19
|
Kang DW, Wang SM, Um YH, Kim NY, Lee CU, Lim HK. Associations Between Sub-Threshold Amyloid-β Deposition, Cortical Volume, and Cognitive Function Modulated by APOE ɛ4 Carrier Status in Cognitively Normal Older Adults. J Alzheimers Dis 2022; 89:1003-1016. [PMID: 35964194 PMCID: PMC9535581 DOI: 10.3233/jad-220427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Background: There has been renewed interest in the deteriorating effects of sub-threshold amyloid-β (Aβ) accumulation in Alzheimer’s disease (AD). Despite evidence suggesting a synergistic interaction between the APOE ɛ4 allele and Aβ deposition in neurodegeneration, few studies have investigated the modulatory role of this allele in sub-threshold Aβ deposition during the preclinical phase. Objective: We aimed to explore the differential effect of the APOE ɛ4 carrier status on the association between sub-threshold Aβ deposition, cortical volume, and cognitive performance in cognitively normal older adults (CN). Methods: A total of 112 CN with sub-threshold Aβ deposition was included in the study. Participants underwent structural magnetic resonance imaging, [18F] flutemetamol PET-CT, and a neuropsychological battery. Potential interactions between APOE ɛ4 carrier status, Aβ accumulation, and cognitive function for cortical volume were assessed with whole-brain voxel-wise analysis. Results: We found that greater cortical volume was observed with higher regional Aβ deposition in the APOE ɛ4 carriers, which could be attributed to an interaction between the APOE ɛ4 carrier status and regional Aβ deposition in the posterior cingulate cortex/precuneus. Finally, the APOE ɛ4 carrier status-neuropsychological test score interaction demonstrated a significant effect on the gray matter volume of the left middle occipital gyrus. Conclusion: There might be a compensatory response to initiating Aβ in APOE ɛ4 carriers during the earliest AD stage. Despite its exploratory nature, this study offers some insight into recent interests concerning probabilistic AD modeling, focusing on the modulating role of the APOE ɛ4 carrier status during the preclinical period.
Collapse
Affiliation(s)
- Dong Woo Kang
- Department of Psychiatry, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sheng-Min Wang
- Department of Psychiatry, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yoo Hyun Um
- Department of Psychiatry, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Nak Young Kim
- Department of Psychiatry, Keyo Hospital, Uiwang, Republic of Korea
| | - Chang Uk Lee
- Department of Psychiatry, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hyun Kook Lim
- Department of Psychiatry, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
20
|
Wang S, Li B, Solomon V, Fonteh A, Rapoport SI, Bennett DA, Arvanitakis Z, Chui HC, Sullivan PM, Yassine HN. Calcium-dependent cytosolic phospholipase A 2 activation is implicated in neuroinflammation and oxidative stress associated with ApoE4. Mol Neurodegener 2022; 17:42. [PMID: 35705959 PMCID: PMC9202185 DOI: 10.1186/s13024-022-00549-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 06/03/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Apolipoprotein E4 (APOE4) is associated with a greater response to neuroinflammation and the risk of developing late-onset Alzheimer's disease (AD), but the mechanisms for this association are not clear. The activation of calcium-dependent cytosolic phospholipase A2 (cPLA2) is involved in inflammatory signaling and is elevated within the plaques of AD brains. The relation between APOE4 genotype and cPLA2 activity is not known. METHODS Mouse primary astrocytes, mouse and human brain samples differing by APOE genotypes were collected for measuring cPLA2 expression, phosphorylation, and activity in relation to measures of inflammation and oxidative stress. RESULTS Greater cPLA2 phosphorylation, cPLA2 activity and leukotriene B4 (LTB4) levels were identified in ApoE4 compared to ApoE3 in primary astrocytes, brains of ApoE-targeted replacement (ApoE-TR) mice, and in human brain homogenates from the inferior frontal cortex of persons with AD dementia carrying APOE3/4 compared to APOE3/3. Higher phosphorylated p38 MAPK but not ERK1/2 was found in ApoE4 primary astrocytes and mouse brains than that in ApoE3. Greater cPLA2 translocation to cytosol was observed in human postmortem frontal cortical synaptosomes with recombinant ApoE4 than ApoE3 ex vivo. In ApoE4 astrocytes, the greater levels of LTB4, reactive oxygen species (ROS), and inducible nitric oxide synthase (iNOS) were reduced after cPLA2 inhibition. CONCLUSIONS Our findings implicate greater activation of cPLA2 signaling system with APOE4, which could represent a potential drug target for mitigating the increased neuroinflammation with APOE4 and AD.
Collapse
Affiliation(s)
- Shaowei Wang
- Keck School of Medicine, University of Southern California, Los Angeles, CA USA
| | - Boyang Li
- Keck School of Medicine, University of Southern California, Los Angeles, CA USA
| | - Victoria Solomon
- Keck School of Medicine, University of Southern California, Los Angeles, CA USA
| | - Alfred Fonteh
- Huntington Medical Research Institutes, Pasadena, CA USA
| | | | - David A. Bennett
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL USA
| | - Zoe Arvanitakis
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL USA
| | - Helena C. Chui
- Keck School of Medicine, University of Southern California, Los Angeles, CA USA
| | - Patrick M. Sullivan
- Department of Medicine, Duke University Medical Center, Durham Veterans Health Administration Medical Center’s Geriatric Research, Education and Clinical Center, Durham, NC USA
| | - Hussein N. Yassine
- Keck School of Medicine, University of Southern California, Los Angeles, CA USA
| |
Collapse
|
21
|
Spotorno N, Najac C, Stomrud E, Mattsson-Carlgren N, Palmqvist S, van Westen D, Ronen I, Hansson O. Astrocytic function is associated with both amyloid-β and tau pathology in non-demented APOE ε4 carriers. Brain Commun 2022; 4:fcac135. [PMID: 35702728 PMCID: PMC9185373 DOI: 10.1093/braincomms/fcac135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 02/17/2022] [Accepted: 05/19/2022] [Indexed: 11/13/2022] Open
Abstract
Abstract
A growing body of evidence suggests that astrocytes play a major role in the pathophysiology of Alzheimer’s disease. Given that APOE is primarily expressed in astrocytes, these cells might be an important link between the APOE ε4 allele and development of Alzheimer’s disease pathology. Here, we investigate this hypothesis in vivo by measuring myo-Inositol, a metabolite involved in astrocytic functions, with magnetic resonance spectroscopy. Currently, there are conflicting evidence regarding the relationship between APOE ε4 and myo-Inositol concentration. Furthermore, data supporting a relationship between APOE ε4, myo-Inositol and Alzheimer’s disease pathology (amyloid-beta and tau proteins) in the preclinical stage of Alzheimer’s disease are limited. A previous study revealed differences in myo-Inositol levels between APOE ε4 carriers and noncarriers already in preclinical Alzheimer’s disease participant. However, other reports showed no impact of APOE genotype on the association between myo-Inositol and rate of amyloid-beta accumulation. In the present study we determined the effect of APOE genotype on the association between myo-Inositol and both amyloid-β and tau deposition quantified by PET in 428 cognitively unimpaired elderly and patients with mild cognitive impairment from the Swedish BioFINDER-2 cohort.
APOE genotype impacted the associations between myo-Inositol and amyloid-β pathology as revealed by an interaction effect between APOE genotype and levels of myo-inositol (p < 0.001) such that higher myo-Inositol concentration was related to more amyloid-beta pathology in APOE ε4 carriers only. A similar interaction effect was also found when investigating the effect of APOE on the association between myo-inositol and tau pathology (p < 0.01). Focusing on the APOE ε4 subsample, myo-Inositol partially (17%) mediated the association between amyloid-beta and tau pathology (p < 0.05). Further, in a subgroup of participants with available plasma levels of glial fibrillary acidic protein, a marker of astroglial activation and astrocytosis, we found that glial fibrillary acidic protein correlated with myo-inositol only in APOE e4 carriers (APOE ε4 carriers: p < 0.01; APOE ε4 non carriers: p > 0.8), suggesting that myo-Inosotol might reflect an aspect of the astrocytic involvement in Alzheimer’s pathology which is specific to the impact of APOE ε4. Therefore, we suggest that myo-Inositol is a candidate in vivo marker to study the impact of APOE ε4 on the interplay between astrocytes and the pathophysiology of Alzheimer’s disease.
Collapse
Affiliation(s)
- Nicola Spotorno
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
| | - Chloé Najac
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Erik Stomrud
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Niklas Mattsson-Carlgren
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Department of Neurology, Skåne University Hospital, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Danielle van Westen
- Image and Function, Skane University Hospital, Lund, Sweden
- Diagnostic Radiology, Institution for Clinical Sciences, Lund University, Lund, Sweden
| | - Itamar Ronen
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Malmö, Lund University, Lund, Sweden
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
22
|
Moon HJ, Haroutunian V, Zhao L. Human apolipoprotein E isoforms are differentially sialylated and the sialic acid moiety in ApoE2 attenuates ApoE2-Aβ interaction and Aβ fibrillation. Neurobiol Dis 2022; 164:105631. [PMID: 35041991 PMCID: PMC9809161 DOI: 10.1016/j.nbd.2022.105631] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 01/09/2022] [Accepted: 01/11/2022] [Indexed: 01/05/2023] Open
Abstract
The APOE genotype is the most prominent genetic risk factor for the development of late-onset Alzheimer''s disease (LOAD); however, the underlying mechanisms remain unclear. In the present study, we found that the sialylation profiles of ApoE protein in the human brain are significantly different among the three isoforms, with ApoE2 exhibiting the most abundant sialic acid modification whereas ApoE4 had the least. We further observed that the sialic acid moiety in ApoE2 significantly affected the interaction between ApoE2 and Aβ peptides. The removal of sialic acid in ApoE2 increased the ApoE2 binding affinity for the Aβ17-24 region of Aβ and promoted Aβ fibrillation. These findings provide a plausible explanation for the well-documented differential roles of ApoE isoforms in Aβ pathogenesis. Specifically, compared to the other two isotypes, the higher expression of sialic acid in ApoE2 may contribute to the less potent interaction between ApoE2 and Aβ and ultimately the slower rate of brain Aβ deposition, a mechanism thought to underlie ApoE2-mediated decreased risk for AD. Future studies are warranted to determine whether the differential sialylation in ApoE isoforms may also contribute to some of their other distinct properties, such as their divergent preferences in associations with lipids and lipoproteins, as well as their potential impact on neuroinflammation through modulation of microglial Siglec activity. Overall, our findings lead to the insight that the sialic acid structure is an important posttranslational modification (PTM) that alters ApoE protein functions with relevance for AD.
Collapse
Affiliation(s)
- Hee-Jung Moon
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA
| | - Vahram Haroutunian
- The Alzheimer's Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 100029, USA
| | - Liqin Zhao
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA; Neuroscience Graduate Program, University of Kansas, Lawrence, KS 66045, USA.
| |
Collapse
|
23
|
Mishra A, Wang Y, Yin F, Vitali F, Rodgers KE, Soto M, Mosconi L, Wang T, Brinton RD. A tale of two systems: Lessons learned from female mid-life aging with implications for Alzheimer's prevention & treatment. Ageing Res Rev 2022; 74:101542. [PMID: 34929348 PMCID: PMC8884386 DOI: 10.1016/j.arr.2021.101542] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 12/05/2021] [Accepted: 12/13/2021] [Indexed: 02/03/2023]
Abstract
Neurological aging is frequently viewed as a linear process of decline, whereas in reality, it is a dynamic non-linear process. The dynamic nature of neurological aging is exemplified during midlife in the female brain. To investigate fundamental mechanisms of midlife aging that underlie risk for development of Alzheimer's disease (AD) in late life, we investigated the brain at greatest risk for the disease, the aging female brain. Outcomes of our research indicate that mid-life aging in the female is characterized by the emergence of three phases: early chronological (pre-menopause), endocrinological (peri-menopause) and late chronological (post-menopause) aging. The endocrinological aging program is sandwiched between early and late chronological aging. Throughout the three stages of midlife aging, two systems of biology, metabolic and immune, are tightly integrated through a network of signaling cascades. The network of signaling between these two systems of biology underlie an orchestrated sequence of adaptative starvation responses that shift the brain from near exclusive dependence on a single fuel, glucose, to utilization of an auxiliary fuel derived from lipids, ketone bodies. The dismantling of the estrogen control of glucose metabolism during mid-life aging is a critical contributor to the shift in fuel systems and emergence of dynamic neuroimmune phenotype. The shift in fuel reliance, puts the largest reservoir of local fatty acids, white matter, at risk for catabolism as a source of lipids to generate ketone bodies through astrocytic beta oxidation. APOE4 genotype accelerates the tipping point for emergence of the bioenergetic crisis. While outcomes derived from research conducted in the female brain are not directly translatable to the male brain, the questions addressed in a female centric program of research are directly applicable to investigation of the male brain. Like females, males with AD exhibit deficits in the bioenergetic system of the brain, activation of the immune system and hallmark Alzheimer's pathologies. The drivers and trajectory of mechanisms underlying neurodegeneration in the male brain will undoubtedly share common aspects with the female in addition to factors unique to the male. Preclinical and clinical evidence indicate that midlife endocrine aging can also be a transitional bridge to autoimmune disorders. Collectively, the data indicate that endocrinological aging is a critical period "tipping point" in midlife which can initiate emergence of the prodromal stage of late-onset-Alzheimer's disease. Interventions that target both immune and metabolic shifts that occur during midlife aging have the potential to alter the trajectory of Alzheimer's risk in late life. Further, to achieve precision medicine for AD, chromosomal sex is a critical variable to consider along with APOE genotype, other genetic risk factors and stage of disease.
Collapse
Affiliation(s)
- Aarti Mishra
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ 85719, USA
| | - Yiwei Wang
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ 85719, USA
| | - Fei Yin
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ 85719, USA
| | - Francesca Vitali
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ 85719, USA
| | - Kathleen E Rodgers
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ 85719, USA
| | - Maira Soto
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ 85719, USA
| | - Lisa Mosconi
- Department of Neurology, Weill Cornell Medicine, New York, NY 10021, USA
| | - Tian Wang
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ 85719, USA
| | - Roberta D Brinton
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ 85719, USA.
| |
Collapse
|
24
|
Coronado BNL, da Cunha FBS, de Oliveira RM, Nóbrega ODT, Ricart CAO, Fontes W, de Sousa MV, de Ávila MP, Martins AMA. Novel Possible Protein Targets in Neovascular Age-Related Macular Degeneration: A Pilot Study Experiment. Front Med (Lausanne) 2022; 8:692272. [PMID: 35155457 PMCID: PMC8828634 DOI: 10.3389/fmed.2021.692272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 12/14/2021] [Indexed: 11/29/2022] Open
Abstract
Age-related macular degeneration (AMD) is among the world's leading causes of blindness. In its neovascular form (nAMD), around 25% of patients present further anatomical and visual deterioration due to persistence of neovascular activity, despite gold-standard treatment protocols using intravitreal anti-VEGF medications. Thus, to comprehend, the molecular pathways that drive choroidal neoangiogenesis, associated with the vascular endothelial growth factor (VEGF), are important steps to elucidate the mechanistic events underneath the disease development. This is a pilot study, a prospective, translational experiment, in a real-life context aiming to evaluate the protein profiles of the aqueous humor of 15 patients divided into three groups: group 1, composed of patients with nAMD, who demonstrated a good response to anti-VEGF intravitreal injections during follow-up (good responsive); group 2, composed of patients with anti-VEGF-resistant nAMD, who demonstrated choroidal neovascularization activity during follow-up (poor/non-responsive); and group 3, composed of control patients without systemic diseases or signs of retinopathy. For proteomic characterization of the groups, mass spectrometry (label-free LC-MS/MS) was used. A total of 2,336 proteins were identified, of which 185 were distinctly regulated and allowed the differentiation of the clinical conditions analyzed. Among those, 39 proteins, including some novel ones, were analyzed as potential disease effectors through their pathophysiological implications in lipid metabolism, oxidative stress, complement system, inflammatory pathways, and angiogenesis. So, this study suggests the participation of other promising biomarkers in neovascular AMD, in addition to the known VEGF.
Collapse
Affiliation(s)
- Bruno Nobre Lins Coronado
- Department of Medical Science, Faculty of Medicine, University of Brasilia, Brasilia, Brazil
- Faculty of Medicine, CESMAC University Center, Maceio, Brazil
- *Correspondence: Bruno Nobre Lins Coronado
| | | | - Raphaela Menezes de Oliveira
- Laboratory of Protein Chemistry and Biochemistry, Department of Cell Biology, Institute of Biological Sciences, University of Brasilia, Brasilia, Brazil
| | | | - Carlos André Ornelas Ricart
- Laboratory of Protein Chemistry and Biochemistry, Department of Cell Biology, Institute of Biological Sciences, University of Brasilia, Brasilia, Brazil
| | - Wagner Fontes
- Laboratory of Protein Chemistry and Biochemistry, Department of Cell Biology, Institute of Biological Sciences, University of Brasilia, Brasilia, Brazil
| | - Marcelo Valle de Sousa
- Laboratory of Protein Chemistry and Biochemistry, Department of Cell Biology, Institute of Biological Sciences, University of Brasilia, Brasilia, Brazil
| | | | - Aline Maria Araújo Martins
- Department of Medical Science, Faculty of Medicine, University of Brasilia, Brasilia, Brazil
- Department of Health Science, School of Medicine, University Center of Brasilia (UniCEUB), Brasilia, Brazil
- Aline Maria Araújo Martins
| |
Collapse
|
25
|
Platelet-derived growth factor-BB and white matter hyperintensity burden in APOE4 carriers. CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2022; 3. [PMID: 35844252 PMCID: PMC9286493 DOI: 10.1016/j.cccb.2022.100131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Background The apolipoprotein-e4 (APOE4) gene increases risk for developing late-onset Alzheimer's disease (AD) and has been linked to increased microvascular dysfunction, including pericyte degeneration and blood-brain barrier breakdown. Platelet-derived growth factor-BB (PDGF-BB) is a glycoprotein involved in blood-brain barrier and pericyte maintenance. Increased PDGF-BB levels have been reported in white matter in AD brain tissue. However, the association between circulating levels of PDGF-BB and cerebral white matter damage in older adults remains unknown. Methods Participants included community-dwelling older adults (age range 55–90 years, M = 73.1 years; SD = 7.5; 61.0% male) from the Alzheimer's Disease Neuroimaging Initiative who underwent venipuncture and blood plasma immunoassay for PDGF-BB, brain MRI scanning with T2-FLAIR for volumetric quantification of white matter hyperintensities (WMH) and APOE4 genotyping (N = 64). Linear regression analyses examined the relationship between plasma PDGF-BB levels and WMH volume, adjusting for age, sex, intracranial volume (ICV) and stratifying by APOE4 status. Results Greater levels of circulating PDGF-BB were related to greater WMH volume, even after accounting for age, sex, ICV and APOE4 carrier status (p = 0.040). Nineteen (29.2%) were APOE4 carriers. When stratified by APOE4 status, the relationship between PDGF-BB and WMH volume was only significant for APOE4 carriers (p = 0.007), but not non-carriers (p = 0.448), after adjusting for age, sex and ICV. Discussion These findings reveal a differential relationship between PDGF-BB and WMH volume for APOE4 carriers versus non-carriers. The APOE4 variant leads to accelerated cerebrovascular injury and cognitive decline. Elevated levels of PDGF-BB in carriers may suggest a role for pericytes and blood-brain barrier dysfunction in white matter damage, vascular cognitive impairment and AD. Additional studies will elucidate the role of PDGF ligands and receptors in these conditions.
Collapse
|
26
|
Abstract
Apolipoprotein E (APOE) has three different isoforms, with APOE4 carriers representing a major risk factor for the development of Alzheimer’s disease (AD). AD is the most common form of dementia, and is a relentlessly progressive disorder that afflicts the aged, characterized by severe memory loss. Presently, AD does not have a cure, increasing the urgency for the development of novel therapeutics for the prevention/treatment of AD. The APOE4 isoform is associated with many pathological mechanisms, such as increased neuroinflammation and a reduction in β-amyloid (Aβ) clearance. The accumulation of Aβ plaques in the brain is a hallmark of AD. The presence of APOE4 can increase neuroinflammation via overactivation of the nuclear factor kappa B (NF-κB) pathway. The NF-κB pathway is a family of transcription factors involved with regulating over 400 genes involved with inflammation. AD is associated with sustained inflammation and an overactivation of the NF-κB pathway. Therefore, targeting the APOE4 isoform and suppressing the NF-κB pathway using anti-inflammatory compounds may result in the development of novel therapeutics for the prevention/treatment of AD.
Collapse
|
27
|
Wang S, Li B, Solomon V, Fonteh A, Rapoport SI, Bennett DA, Arvanitakis Z, Chui HC, Miller C, Sullivan PM, Wang HY, Yassine HN. Calcium-dependent cytosolic phospholipase A 2 activation is implicated in neuroinflammation and oxidative stress associated with ApoE4. Mol Neurodegener 2021; 16:26. [PMID: 33863362 PMCID: PMC8052701 DOI: 10.1186/s13024-021-00438-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 03/03/2021] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Apolipoprotein E4 (APOE4) is associated with a greater response to neuroinflammation and the risk of developing late-onset Alzheimer's disease (AD), but the mechanisms for this association are not clear. The activation of calcium-dependent cytosolic phospholipase A2 (cPLA2) is involved in inflammatory signaling and is elevated within the plaques of AD brains. The relation between APOE4 genotype and cPLA2 activity is not known. METHODS Mouse primary astrocytes, mouse and human brain samples differing by APOE genotypes were collected for measuring cPLA2 expression, phosphorylation, and activity in relation to measures of inflammation and oxidative stress. RESULTS Greater cPLA2 phosphorylation, cPLA2 activity and leukotriene B4 (LTB4) levels were identified in ApoE4 compared to ApoE3 in primary astrocytes, brains of ApoE-targeted replacement (ApoE-TR) mice, and in human brain homogenates from the inferior frontal cortex of patients with AD carrying APOE3/E4 compared to APOE3/E3. Greater cPLA2 phosphorylation was also observed in human postmortem frontal cortical synaptosomes and primary astrocytes after treatment with recombinant ApoE4 ex vivo. In ApoE4 astrocytes, the greater levels of LTB4, reactive oxygen species (ROS), and inducible nitric oxide synthase (iNOS) were reduced after cPLA2 inhibition. CONCLUSIONS Our findings implicate greater activation of cPLA2 signaling system with APOE4, which could represent a potential drug target for mitigating the increased neuroinflammation with APOE4 and AD.
Collapse
Affiliation(s)
- Shaowei Wang
- Departments of Medicine and Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA USA
| | - Boyang Li
- Departments of Medicine and Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA USA
| | - Victoria Solomon
- Departments of Medicine and Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA USA
| | - Alfred Fonteh
- Huntington Medical Research Institutes, Pasadena, CA USA
| | | | - David A. Bennett
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL USA
| | - Zoe Arvanitakis
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL USA
| | - Helena C. Chui
- Departments of Medicine and Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA USA
| | - Carol Miller
- Departments of Medicine and Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA USA
| | - Patrick M. Sullivan
- Department of Medicine, Duke University Medical Center, Durham Veterans Health Administration Medical Center’s Geriatric Research, Education and Clinical Center, Durham, NC USA
| | - Hoau-Yan Wang
- The City University of New York School of Medicine, New York, NY USA
- Graduate School of The City University of New York, New York, USA
| | - Hussein N. Yassine
- Departments of Medicine and Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA USA
| |
Collapse
|
28
|
Martens N, Schepers M, Zhan N, Leijten F, Voortman G, Tiane A, Rombaut B, Poisquet J, Sande NVD, Kerksiek A, Kuipers F, Jonker JW, Liu H, Lütjohann D, Vanmierlo T, Mulder MT. 24(S)-Saringosterol Prevents Cognitive Decline in a Mouse Model for Alzheimer's Disease. Mar Drugs 2021; 19:190. [PMID: 33801706 PMCID: PMC8065937 DOI: 10.3390/md19040190] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/16/2021] [Accepted: 03/25/2021] [Indexed: 12/13/2022] Open
Abstract
We recently found that dietary supplementation with the seaweed Sargassum fusiforme, containing the preferential LXRβ-agonist 24(S)-saringosterol, prevented memory decline and reduced amyloid-β (Aβ) deposition in an Alzheimer's disease (AD) mouse model without inducing hepatic steatosis. Here, we examined the effects of 24(S)-saringosterol as a food additive on cognition and neuropathology in AD mice. Six-month-old male APPswePS1ΔE9 mice and wildtype C57BL/6J littermates received 24(S)-saringosterol (0.5 mg/25 g body weight/day) (APPswePS1ΔE9 n = 20; C57BL/6J n = 19) or vehicle (APPswePS1ΔE9 n = 17; C57BL/6J n = 19) for 10 weeks. Cognition was assessed using object recognition and object location tasks. Sterols were analyzed by gas chromatography/mass spectrometry, Aβ and inflammatory markers by immunohistochemistry, and gene expression by quantitative real-time PCR. Hepatic lipids were quantified after Oil-Red-O staining. Administration of 24(S)-saringosterol prevented cognitive decline in APPswePS1ΔE9 mice without affecting the Aβ plaque load. Moreover, 24(S)-saringosterol prevented the increase in the inflammatory marker Iba1 in the cortex of APPswePS1ΔE9 mice (p < 0.001). Furthermore, 24(S)-saringosterol did not affect the expression of lipid metabolism-related LXR-response genes in the hippocampus nor the hepatic neutral lipid content. Thus, administration of 24(S)-saringosterol prevented cognitive decline in APPswePS1ΔE9 mice independent of effects on Aβ load and without adverse effects on liver fat content. The anti-inflammatory effects of 24(S)-saringosterol may contribute to the prevention of cognitive decline.
Collapse
Affiliation(s)
- Nikita Martens
- Department of Internal Medicine, Section Pharmacology and Vascular Medicine, Erasmus University Medical Center, 3015 Rotterdam, The Netherlands
- Department of Neuroscience, Biomedical Research Institute, European Graduate School of Neuroscience, Hasselt University, BE 3590 Hasselt, Belgium
| | - Melissa Schepers
- Department of Neuroscience, Biomedical Research Institute, European Graduate School of Neuroscience, Hasselt University, BE 3590 Hasselt, Belgium
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neurosciences, Division Translational Neuroscience, Maastricht University, 6200 Maastricht, The Netherlands
| | - Na Zhan
- Department of Internal Medicine, Section Pharmacology and Vascular Medicine, Erasmus University Medical Center, 3015 Rotterdam, The Netherlands
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266100, China
| | - Frank Leijten
- Department of Internal Medicine, Section Pharmacology and Vascular Medicine, Erasmus University Medical Center, 3015 Rotterdam, The Netherlands
| | - Gardi Voortman
- Department of Internal Medicine, Section Pharmacology and Vascular Medicine, Erasmus University Medical Center, 3015 Rotterdam, The Netherlands
| | - Assia Tiane
- Department of Neuroscience, Biomedical Research Institute, European Graduate School of Neuroscience, Hasselt University, BE 3590 Hasselt, Belgium
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neurosciences, Division Translational Neuroscience, Maastricht University, 6200 Maastricht, The Netherlands
| | - Ben Rombaut
- Department of Neuroscience, Biomedical Research Institute, European Graduate School of Neuroscience, Hasselt University, BE 3590 Hasselt, Belgium
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neurosciences, Division Translational Neuroscience, Maastricht University, 6200 Maastricht, The Netherlands
| | - Janne Poisquet
- Department of Neuroscience, Biomedical Research Institute, European Graduate School of Neuroscience, Hasselt University, BE 3590 Hasselt, Belgium
| | - Nienke van de Sande
- Department of Internal Medicine, Section Pharmacology and Vascular Medicine, Erasmus University Medical Center, 3015 Rotterdam, The Netherlands
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neurosciences, Division Translational Neuroscience, Maastricht University, 6200 Maastricht, The Netherlands
| | - Anja Kerksiek
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53105 Bonn, Germany
| | - Folkert Kuipers
- Department of Pediatrics, Section of Molecular Metabolism and Nutrition, University Medical Center Groningen, 9713 Groningen, The Netherlands
| | - Johan W Jonker
- Department of Pediatrics, Section of Molecular Metabolism and Nutrition, University Medical Center Groningen, 9713 Groningen, The Netherlands
| | - Hongbing Liu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266100, China
| | - Dieter Lütjohann
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53105 Bonn, Germany
| | - Tim Vanmierlo
- Department of Internal Medicine, Section Pharmacology and Vascular Medicine, Erasmus University Medical Center, 3015 Rotterdam, The Netherlands
- Department of Neuroscience, Biomedical Research Institute, European Graduate School of Neuroscience, Hasselt University, BE 3590 Hasselt, Belgium
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neurosciences, Division Translational Neuroscience, Maastricht University, 6200 Maastricht, The Netherlands
| | - Monique T Mulder
- Department of Internal Medicine, Section Pharmacology and Vascular Medicine, Erasmus University Medical Center, 3015 Rotterdam, The Netherlands
| |
Collapse
|
29
|
Scheinman SB, Zaldua S, Dada A, Krochmaliuk K, Dye K, Marottoli FM, Thatcher GRJ, Tai LM. Systemic Candesartan Treatment Modulates Behavior, Synaptic Protein Levels, and Neuroinflammation in Female Mice That Express Human APOE4. Front Neurosci 2021; 15:628403. [PMID: 33642985 PMCID: PMC7902885 DOI: 10.3389/fnins.2021.628403] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 01/20/2021] [Indexed: 11/26/2022] Open
Abstract
Evidence suggests that angiotensin receptor blockers (ARBs) could be beneficial for Alzheimer’s disease (AD) patients independent of any effects on hypertension. However, studies in rodent models directly testing the activity of ARB treatment on behavior and AD-relevent pathology including neuroinflammation, Aβ levels, and cerebrovascular function, have produced mixed results. APOE4 is a major genetic risk factor for AD and has been linked to many of the same functions as those purported to be modulated by ARB treatment. Therefore, evaluating the effects of ARB treatment on behavior and AD-relevant pathology in mice that express human APOE4 could provide important information on whether to further develop ARBs for AD therapy. In this study, we treated female and male mice that express the human APOE4 gene in the absence (E4FAD−) or presence (E4FAD+) of high Aβ levels with the ARB prodrug candesartan cilexetil for a duration of 4 months. Compared to vehicle, candesartan treatment resulted in greater memory-relevant behavior and higher hippocampal presynaptic protein levels in female, but not male, E4FAD− and E4FAD+ mice. The beneficial effects of candesartan in female E4FAD− and E4FAD+ mice occurred in tandem with lower GFAP and Iba1 levels in the hippocampus, whereas there were no effects on markers of cerebrovascular function and Aβ levels. Collectively, these data imply that the effects of ARBs on AD-relevant pathology may be modulated in part by the interaction between APOE genotype and biological sex. Thus, the further development of ARBs could provide therapeutic options for targeting neuroinflammation in female APOE4 carriers.
Collapse
Affiliation(s)
- Sarah B Scheinman
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Steve Zaldua
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Adedoyin Dada
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Kateryna Krochmaliuk
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Katherine Dye
- UICentre, University of Illinois at Chicago, Chicago, IL, United States
| | - Felecia M Marottoli
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Gregory R J Thatcher
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, United States
| | - Leon M Tai
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
30
|
APOE4 genetic polymorphism results in impaired recovery in a repeated mild traumatic brain injury model and treatment with Bryostatin-1 improves outcomes. Sci Rep 2020; 10:19919. [PMID: 33199792 PMCID: PMC7670450 DOI: 10.1038/s41598-020-76849-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Accepted: 10/08/2020] [Indexed: 11/28/2022] Open
Abstract
After traumatic brain injury (TBI), some people have worse recovery than others. Single nucleotide polymorphisms (SNPs) in Apolipoprotein E (APOE) are known to increase risk for developing Alzheimer’s disease, however there is controversy from human and rodent studies as to whether ApoE4 is a risk factor for worse outcomes after brain trauma. To resolve these conflicting studies we have explored the effect of the human APOE4 gene in a reproducible mouse model that mimics common human injuries. We have investigated cellular and behavioral outcomes in genetically engineered human APOE targeted replacement (TR) mice following repeated mild TBI (rmTBI) using a lateral fluid percussion injury model. Relative to injured APOE3 TR mice, injured APOE4 TR mice had more inflammation, neurodegeneration, apoptosis, p-tau, and activated microglia and less total brain-derived neurotrophic factor (BDNF) in the cortex and/or hippocampus at 1 and/or 21 days post-injury. We utilized a novel personalized approach to treating APOE4 susceptible mice by administering Bryostatin-1, which improved cellular as well as motor and cognitive behavior outcomes at 1 DPI in the APOE4 injured mice. This study demonstrates that APOE4 is a risk factor for poor outcomes after rmTBI and highlights how personalized therapeutics can be a powerful treatment option.
Collapse
|
31
|
Emam S, Nasrollahpour M, Colarusso B, Cai X, Grant S, Kulkarni P, Ekenseair A, Gharagouzloo C, Ferris CF, Sun NX. Detection of presymptomatic Alzheimer's disease through breath biomarkers. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2020; 12:e12088. [PMID: 33088894 PMCID: PMC7560498 DOI: 10.1002/dad2.12088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 07/02/2020] [Accepted: 07/13/2020] [Indexed: 12/26/2022]
Abstract
Introduction Novel sensors were developed to detect exhaled volatile organic compounds to aid in the diagnosis of mild cognitive impairment associated with early stage Alzheimer's disease (AD). The sensors were sensitive to a rat model that combined the human apolipoprotein E (APOE)4 gene with aging and the Western diet. Methods Gas sensors fabricated from molecularly imprinted polymer-graphene were engineered to react with alkanes and small fatty acids associated with lipid peroxidation. With a detection sensitivity in parts per trillion the sensors were tested against the breath of wild-type and APOE4 male rats. Resting state BOLD functional connectivity was used to assess hippocampal function. Results Only APOE4 rats, and not wild-type controls, tested positive to several small hydrocarbons and presented with reduced functional coupling in hippocampal circuitry. Discussion These results are proof-of-concept toward the development of sensors that can be used as breath detectors in the diagnosis, prognosis, and treatment of presymptomatic AD.
Collapse
Affiliation(s)
- Shadi Emam
- Department of Electrical and Computer Engineering Advanced Materials and Microsystems Laboratory Northeastern University Boston Massachusetts USA
| | - Mehdi Nasrollahpour
- Department of Electrical and Computer Engineering Advanced Materials and Microsystems Laboratory Northeastern University Boston Massachusetts USA
| | - Bradley Colarusso
- Department of Psychology Center for Translational NeuroImaging Northeastern University Boston Massachusetts USA
| | - Xuezhu Cai
- Department of Psychology Center for Translational NeuroImaging Northeastern University Boston Massachusetts USA
| | - Simone Grant
- Department of Chemical Engineering Northeastern University Boston Massachusetts USA
| | - Praveen Kulkarni
- Department of Psychology Center for Translational NeuroImaging Northeastern University Boston Massachusetts USA
| | - Adam Ekenseair
- Department of Chemical Engineering Northeastern University Boston Massachusetts USA
| | - Codi Gharagouzloo
- Imaginostics Inc. Northeastern University Cambridge Massachusetts USA
| | - Craig F Ferris
- Department of Psychology Center for Translational NeuroImaging Northeastern University Boston Massachusetts USA
| | - Nian-Xiang Sun
- Department of Electrical and Computer Engineering Advanced Materials and Microsystems Laboratory Northeastern University Boston Massachusetts USA
| |
Collapse
|
32
|
Apolipoprotein E4 exhibits intermediates with domain interaction. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2020; 1868:140535. [PMID: 32882410 DOI: 10.1016/j.bbapap.2020.140535] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 08/18/2020] [Accepted: 08/27/2020] [Indexed: 11/23/2022]
Abstract
ApoE4(C112R) is the strongest risk factor for Alzheimer's disease, while apoE3(C112) is considered normal. The C112R substitution is believed to alter the interactions between the N-terminal (NTD) and the C-terminal domain (CTD) leading to major functional differences. Here we investigate how the molecular property of the residue at position 112 affects domain interaction using an array of C112X substitutions with arginine, alanine, threonine, valine, leucine and isoleucine as 'X'. We attempt to determine the free energy of domain interaction (∆GINT) from stabilities of the NTD (∆GNTD) and CTD (∆GCTD) in the full-length apoE, and the stabilities of fragments of the NTD (∆GNTF) and CTD (∆GCTF), using the relationship, ∆GINT = ∆GNTD + ∆GCTD - ∆GNTF - ∆GCTF. We find that although ∆GNTD is strongly dependent on the C112X substitutions, ∆GNTD - ∆GNTF is small. Furthermore, ∆GCTD remains nearly the same as ∆GCTF. Therefore, ∆GINT is estimated to be small and similar for the apoE isoforms. However, stability of domain interaction monitored by urea dependent changes in interdomain Forster Resonance Energy Transfer (FRET) is found to be strongly dependent on C112X substitutions. ApoE4 exhibits the highest mid-point of denaturation of interdomain FRET. To resolve the apparently contradictory observations, we hypothesize that higher interdomain FRET in apoE4 in urea may involve 'intermediate' states. Enhanced fluorescence of bis-ANS and susceptibility to proteolytic cleavage support that apoE4, specifically, the NTD of apoE4 harbor 'intermediates' in both native and mildly denaturing conditions. The intermediates could hold key to the pathological functions of apoE4.
Collapse
|
33
|
Johnson LW, Lundgren K, Henrich V, Phillips S. Factors influencing recovery from mild traumatic brain injury. Brain Inj 2020; 34:1202-1212. [PMID: 32705914 DOI: 10.1080/02699052.2020.1795719] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
PRIMARY OBJECTIVE This study determined whether initial GCS score, head CT results, cognitive performance on IMPACT testing, or APOE genotype most effectively predicted 1-month functional outcome after mild traumatic brain injury (mTBI). This study tested the hypotheses that participants with poor performance on initial cognitive testing and those with an APOEe4 genotype would exhibit a poorer 1-month recovery after mTBI. RESEARCH DESIGN Regression analysis determined which independent variables were most effective in predicting 1-month GOS-E or DRS score. Independent t-test procedures determined whether cognitive recovery varied across APOEe4 carriers. METHODS AND PROCEDURES 49 participants admitted to the hospital with mTBI received cognitive evaluation within 48 hours after injury and again one month later. DNA analysis provided participant APOE genotype. MAIN OUTCOMES AND RESULTS Results showed that no study variables significantly predicted GOS-E or DRS scores, however, differences were identified when APOE groups were compared. Participants who were noncarriers of APOEe4 had significantly slower reaction times compared to APOEe4 carriers. Participants who were homozygous APOEe4 carriers had significantly lower instances of impulsivity than noncarriers. CONCLUSIONS Further research is needed to understand how APOE allele status and performance on initial cognitive testing may influence short-term recovery after mTBI.
Collapse
Affiliation(s)
- Leslie Weaver Johnson
- Communications Disorders Program, North Carolina Central University , Durham, North Carolina, USA
| | - Kristine Lundgren
- Department of Communication and Sciences Disorders, University of North Carolina - Greensboro , Greensboro, North Carolina, USA
| | - Vincent Henrich
- Department of Biology, University of North Carolina - Greensboro , Greensboro, North Carolina, USA
| | - Susan Phillips
- Department of Communication and Sciences Disorders, University of North Carolina - Greensboro , Greensboro, North Carolina, USA
| |
Collapse
|
34
|
Wong MY, Lewis M, Doherty JJ, Shi Y, Cashikar AG, Amelianchik A, Tymchuk S, Sullivan PM, Qian M, Covey DF, Petsko GA, Holtzman DM, Paul SM, Luo W. 25-Hydroxycholesterol amplifies microglial IL-1β production in an apoE isoform-dependent manner. J Neuroinflammation 2020; 17:192. [PMID: 32552741 PMCID: PMC7298825 DOI: 10.1186/s12974-020-01869-3] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 06/08/2020] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Genome-wide association studies of Alzheimer's disease (AD) have implicated pathways related to lipid homeostasis and innate immunity in AD pathophysiology. However, the exact cellular and chemical mediators of neuroinflammation in AD remain poorly understood. The oxysterol 25-hydroxycholesterol (25-HC) is an important immunomodulator produced by peripheral macrophages with wide-ranging effects on cell signaling and innate immunity. Cholesterol 25-hydroxylase (CH25H), the enzyme responsible for 25-HC production, has also been found to be one of the disease-associated microglial (DAM) genes that are upregulated in the brain of AD and AD transgenic mouse models. METHODS We used real-time PCR and immunoblotting to examine CH25H expression in human AD brain tissue and in transgenic mouse brain tissue-bearing amyloid-β plaques or tau pathology. The innate immune response of primary mouse microglia under different treatment conditions or bearing different genetic backgrounds was analyzed using ELISA, western blotting, or immunocytochemistry. RESULTS We found that CH25H expression is upregulated in human AD brain tissue and in transgenic mouse brain tissue-bearing amyloid-β plaques or tau pathology. Treatment with the toll-like receptor 4 (TLR4) agonist lipopolysaccharide (LPS) markedly upregulates CH25H expression in the mouse brain and stimulates CH25H expression and 25-HC secretion in mouse primary microglia. We found that LPS-induced microglial production of the pro-inflammatory cytokine IL-1β is markedly potentiated by 25-HC and attenuated by the deletion of CH25H. Microglia expressing apolipoprotein E4 (apoE4), a genetic risk factor for AD, produce greater amounts of 25-HC than apoE3-expressing microglia following treatment with LPS. Remarkably, 25-HC treatment results in a greater level of IL-1β secretion in LPS-activated apoE4-expressing microglia than in apoE2- or apoE3-expressing microglia. Blocking potassium efflux or inhibiting caspase-1 prevents 25-HC-potentiated IL-1β release in apoE4-expressing microglia, indicating the involvement of caspase-1 inflammasome activity. CONCLUSION 25-HC may function as a microglial-secreted inflammatory mediator in the brain, promoting IL-1β-mediated neuroinflammation in an apoE isoform-dependent manner (E4>>E2/E3) and thus may be an important mediator of neuroinflammation in AD.
Collapse
Affiliation(s)
- Man Ying Wong
- grid.5386.8000000041936877XAppel Alzheimer’s Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY USA
| | - Michael Lewis
- grid.476678.c0000 0004 5913 664XSage Therapeutics, Cambridge, Massachusetts USA
| | - James J. Doherty
- grid.476678.c0000 0004 5913 664XSage Therapeutics, Cambridge, Massachusetts USA
| | - Yang Shi
- grid.4367.60000 0001 2355 7002Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO USA
| | - Anil G. Cashikar
- grid.4367.60000 0001 2355 7002Departments of Neurology and Psychiatry, Hope Center for Neurological Disorders, Taylor Family Institute, Washington University School of Medicine, St. Louis, MO USA
| | - Anna Amelianchik
- grid.5386.8000000041936877XAppel Alzheimer’s Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY USA
| | - Svitlana Tymchuk
- grid.5386.8000000041936877XAppel Alzheimer’s Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY USA
| | - Patrick M. Sullivan
- grid.281208.10000 0004 0419 3073Department of Medicine, Duke University Medical Center, Durham Veterans Health Administration Medical Center’s Geriatric Research, Education and Clinical Center, Durham, NC USA
| | - Mingxing Qian
- grid.4367.60000 0001 2355 7002Departments of Developmental Biology, Anesthesiology, Taylor Family Institute for Innovative Psychiatric Research, Washington University in St. Louis, 660 South Euclid Avenue, St. Louis, MO 63110 USA
| | - Douglas F. Covey
- grid.4367.60000 0001 2355 7002Departments of Developmental Biology, Anesthesiology, Taylor Family Institute for Innovative Psychiatric Research, Washington University in St. Louis, 660 South Euclid Avenue, St. Louis, MO 63110 USA
| | - Gregory A. Petsko
- grid.5386.8000000041936877XAppel Alzheimer’s Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY USA
| | - David M. Holtzman
- grid.4367.60000 0001 2355 7002Department of Neurology, Hope Center for Neurological Disorders, Charles F. and Joanne Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO USA
| | - Steven M. Paul
- grid.476678.c0000 0004 5913 664XSage Therapeutics, Cambridge, Massachusetts USA ,grid.4367.60000 0001 2355 7002Departments of Neurology and Psychiatry, Hope Center for Neurological Disorders, Taylor Family Institute, Washington University School of Medicine, St. Louis, MO USA
| | - Wenjie Luo
- Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
35
|
Raman S, Brookhouser N, Brafman DA. Using human induced pluripotent stem cells (hiPSCs) to investigate the mechanisms by which Apolipoprotein E (APOE) contributes to Alzheimer's disease (AD) risk. Neurobiol Dis 2020; 138:104788. [PMID: 32032733 PMCID: PMC7098264 DOI: 10.1016/j.nbd.2020.104788] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 01/14/2020] [Accepted: 01/31/2020] [Indexed: 01/02/2023] Open
Abstract
Although the biochemical and pathological hallmarks of Alzheimer's disease (AD), such as axonal transport defects, synaptic loss, and selective neuronal death, are well characterized, the underlying mechanisms that cause AD are largely unknown, thereby making it difficult to design effective therapeutic interventions. Genome-wide association studies (GWAS) studies have identified several factors associated with increased AD risk. Of these genetic factors, polymorphisms in the Apolipoprotein E (APOE) gene are the strongest and most prevalent. While it has been established that the ApoE protein modulates the formation of amyloid plaques and neurofibrillary tangles, the precise molecular mechanisms by which various ApoE isoforms enhance or mitigate AD onset and progression in aging adults are yet to be elucidated. Advances in cellular reprogramming to generate disease-in-a-dish models now provide a simplified and accessible system that complements animal and primary cell models to study ApoE in the context of AD. In this review, we will describe the use and manipulation of human induced pluripotent stem cells (hiPSCs) in dissecting the interaction between ApoE and AD. First, we will provide an overview of the proposed roles that ApoE plays in modulating pathophysiology of AD. Next, we will summarize the recent studies that have employed hiPSCs to model familial and sporadic AD. Lastly, we will speculate on how current advances in genome editing technologies and organoid culture systems can be used to improve hiPSC-based tools to investigate ApoE-dependent modulation of AD onset and progression.
Collapse
Affiliation(s)
- Sreedevi Raman
- School of Biological and Health Systems Engineering, Arizona State University, United States of America
| | - Nicholas Brookhouser
- School of Biological and Health Systems Engineering, Arizona State University, United States of America; Graduate Program in Clinical Translational Sciences, University of Arizona College of Medicine-Phoenix, United States of America
| | - David A Brafman
- School of Biological and Health Systems Engineering, Arizona State University, United States of America.
| |
Collapse
|
36
|
Contreras JA, Aslanyan V, Sweeney MD, Sanders LMJ, Sagare AP, Zlokovic BV, Toga AW, Han SD, Morris JC, Fagan A, Massoumzadeh P, Benzinger TL, Pa J. Functional connectivity among brain regions affected in Alzheimer's disease is associated with CSF TNF-α in APOE4 carriers. Neurobiol Aging 2020; 86:112-122. [PMID: 31870643 PMCID: PMC7205323 DOI: 10.1016/j.neurobiolaging.2019.10.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 09/20/2019] [Accepted: 10/22/2019] [Indexed: 01/01/2023]
Abstract
It is now recognized that understanding how neuroinflammation affects brain function may provide new insights into Alzheimer's pathophysiology. Tumor necrosis factor (TNF)-α, an inflammatory cytokine marker, has been implicated in Alzheimer's disease (AD), as it can impair neuronal function through suppression of long-term potentiation. Our study investigated the relationship between cerebrospinal fluid TNF-α and functional connectivity (FC) in a cohort of 64 older adults (μ age = 69.76 years; 30 cognitively normal, 34 mild AD). Higher cerebrospinal fluid TNF-α levels were associated with lower FC among brain regions important for high-level decision-making, inhibitory control, and memory. This effect was moderated by apolipoprotein E-ε4 (APOE4) status. Graph theory metrics revealed there were significant differences between APOE4 carriers at the node level, and by diagnosis at the network level suggesting global brain network dysfunction in participants with AD. These findings suggest proinflammatory mechanisms may contribute to reduced FC in regions important for high-level cognition. Future studies are needed to understand the role of inflammation on brain function and clinical progression, especially in APOE4 carriers.
Collapse
Affiliation(s)
- Joey Annette Contreras
- Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA
| | - Vahan Aslanyan
- Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA
| | - Melanie D Sweeney
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, USA
| | - Lianne M J Sanders
- Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA; Department of Human Movement Sciences, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Abhay P Sagare
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, USA
| | - Berislav V Zlokovic
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, USA
| | - Arthur W Toga
- Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA
| | - S Duke Han
- Family Medicine, Neurology, Psychology, and Gerontology, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - John C Morris
- Knight Alzheimer's Disease Research Center, Washington University, St. Louis, MO, USA; Department of Neurology, Washington University, St Louis, MO, USA
| | - Anne Fagan
- Knight Alzheimer's Disease Research Center, Washington University, St. Louis, MO, USA; Department of Neurology, Washington University, St Louis, MO, USA
| | - Parinaz Massoumzadeh
- Knight Alzheimer's Disease Research Center, Washington University, St. Louis, MO, USA
| | - Tammie L Benzinger
- Knight Alzheimer's Disease Research Center, Washington University, St. Louis, MO, USA; Department of Radiology and Neurological Surgery, Washington University, St. Louis, MO, USA
| | - Judy Pa
- Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
37
|
Williams T, Borchelt DR, Chakrabarty P. Therapeutic approaches targeting Apolipoprotein E function in Alzheimer's disease. Mol Neurodegener 2020; 15:8. [PMID: 32005122 PMCID: PMC6995170 DOI: 10.1186/s13024-020-0358-9] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 01/14/2020] [Indexed: 12/12/2022] Open
Abstract
One of the primary genetic risk factors for Alzheimer’s disease (AD) is the presence of the Ɛ4 allele of apolipoprotein E (APOE). APOE is a polymorphic lipoprotein that is a major cholesterol carrier in the brain. It is also involved in various cellular functions such as neuronal signaling, neuroinflammation and glucose metabolism. Humans predominantly possess three different allelic variants of APOE, termed E2, E3, and E4, with the E3 allele being the most common. The presence of the E4 allele is associated with increased risk of AD whereas E2 reduces the risk. To understand the molecular mechanisms that underlie APOE-related genetic risk, considerable effort has been devoted towards developing cellular and animal models. Data from these models indicate that APOE4 exacerbates amyloid β plaque burden in a dose-dependent manner. and may also enhance tau pathogenesis in an isoform-dependent manner. Other studies have suggested APOE4 increases the risk of AD by mechanisms that are distinct from modulation of Aβ or tau pathology. Further, whether plasma APOE, by influencing systemic metabolic pathways, can also possibly alter CNS function indirectly is not complete;y understood. Collectively, the available studies suggest that APOE may impact multiple signaling pathways and thus investigators have sought therapeutics that would disrupt pathological functions of APOE while preserving or enhancing beneficial functions. This review will highlight some of the therapeutic strategies that are currently being pursued to target APOE4 towards preventing or treating AD and we will discuss additional strategies that holds promise for the future.
Collapse
Affiliation(s)
- Tosha Williams
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA.,Department of Neuroscience, University of Florida, Gainesville, FL, 32610, USA
| | - David R Borchelt
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA.,Department of Neuroscience, University of Florida, Gainesville, FL, 32610, USA.,McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA
| | - Paramita Chakrabarty
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, 32610, USA. .,Department of Neuroscience, University of Florida, Gainesville, FL, 32610, USA. .,McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
38
|
Wang Y, Mishra A, Brinton RD. Transitions in metabolic and immune systems from pre-menopause to post-menopause: implications for age-associated neurodegenerative diseases. F1000Res 2020; 9. [PMID: 32047612 PMCID: PMC6993821 DOI: 10.12688/f1000research.21599.1] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/22/2020] [Indexed: 12/13/2022] Open
Abstract
The brain undergoes two aging programs: chronological and endocrinological. This is particularly evident in the female brain, which undergoes programs of aging associated with reproductive competency. Comprehensive understanding of the dynamic metabolic and neuroinflammatory aging process in the female brain can illuminate windows of opportunities to promote healthy brain aging. Bioenergetic crisis and chronic low-grade inflammation are hallmarks of brain aging and menopause and have been implicated as a unifying factor causally connecting genetic risk factors for Alzheimer's disease and other neurodegenerative diseases. In this review, we discuss metabolic phenotypes of pre-menopausal, peri-menopausal, and post-menopausal aging and their consequent impact on the neuroinflammatory profile during each transition state. A critical aspect of the aging process is the dynamic metabolic neuro-inflammatory profiles that emerge during chronological and endocrinological aging. These dynamic systems of biology are relevant to multiple age-associated neurodegenerative diseases and provide a therapeutic framework for prevention and delay of neurodegenerative diseases of aging. While these findings are based on investigations of the female brain, they have a broader fundamental systems of biology strategy for investigating the aging male brain. Molecular characterization of alterations in fuel utilization and neuroinflammatory mechanisms during these neuro-endocrine transition states can inform therapeutic strategies to mitigate the risk of Alzheimer's disease in women. We further discuss a precision hormone replacement therapy approach to target symptom profiles during endocrine and chronological aging to reduce risk for age-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Yiwei Wang
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ, 85721, USA
| | - Aarti Mishra
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ, 85721, USA
| | - Roberta Diaz Brinton
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ, 85721, USA
| |
Collapse
|
39
|
Lee EG, Tulloch J, Chen S, Leong L, Saxton AD, Kraemer B, Darvas M, Keene CD, Shutes-David A, Todd K, Millard S, Yu CE. Redefining transcriptional regulation of the APOE gene and its association with Alzheimer's disease. PLoS One 2020; 15:e0227667. [PMID: 31978088 PMCID: PMC6980611 DOI: 10.1371/journal.pone.0227667] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 12/24/2019] [Indexed: 01/09/2023] Open
Abstract
The apolipoprotein E gene (APOE) is the strongest genetic risk factor for late-onset Alzheimer's disease (AD), yet the expression of APOE is not clearly understood. For example, it is unclear whether AD patients have elevated or decreased APOE expression or why the correlation levels of APOE RNA and the ApoE protein differ across studies. Likewise, APOE has a single CpG island (CGI) that overlaps with its 3'-exon, and this CGI's effect is unknown. We previously reported that the APOE CGI is highly methylated in human postmortem brain (PMB) and that this methylation is altered in AD frontal lobe. In this study, we comprehensively characterized APOE RNA transcripts and correlated levels of RNA expression with DNA methylation levels across the APOE CGI. We discovered the presence of APOE circular RNA (circRNA) and found that circRNA and full-length mRNA each constitute approximately one third of the total APOE RNA, with truncated mRNAs likely constituting some of the missing fraction. All APOE RNA species demonstrated significantly higher expression in AD frontal lobe than in control frontal lobe. Furthermore, we observed a negative correlation between the levels of total APOE RNA and DNA methylation at the APOE CGI in the frontal lobe. When stratified by disease status, this correlation was strengthened in controls but not in AD. Our findings suggest a possible modified mechanism of gene action for APOE in AD that involves not only the protein isoforms but also an epigenetically regulated transcriptional program driven by DNA methylation in the APOE CGI.
Collapse
Affiliation(s)
- Eun-Gyung Lee
- Geriatric Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, United States of America
| | - Jessica Tulloch
- Geriatric Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, United States of America
| | - Sunny Chen
- Geriatric Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, United States of America
| | - Lesley Leong
- Geriatric Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, United States of America
| | - Aleen D. Saxton
- Geriatric Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, United States of America
| | - Brian Kraemer
- Geriatric Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, United States of America
- Department of Medicine, University of Washington, Seattle, WA, United States of America
| | - Martin Darvas
- Department of Pathology, University of Washington, Seattle, WA, United States of America
| | - C. Dirk Keene
- Department of Pathology, University of Washington, Seattle, WA, United States of America
| | - Andrew Shutes-David
- Geriatric Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, United States of America
| | - Kaitlin Todd
- Geriatric Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, United States of America
| | - Steve Millard
- Geriatric Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, United States of America
| | - Chang-En Yu
- Geriatric Research, Education, and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, United States of America
- Department of Medicine, University of Washington, Seattle, WA, United States of America
| |
Collapse
|
40
|
Russ TC, Kivimäki M, Batty GD. Respiratory Disease and Lower Pulmonary Function as Risk Factors for Dementia: A Systematic Review With Meta-analysis. Chest 2020; 157:1538-1558. [PMID: 31952950 DOI: 10.1016/j.chest.2019.12.012] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 11/13/2019] [Accepted: 12/12/2019] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND In addition to affecting the oxygen supply to the brain, pulmonary function is a marker of multiple insults throughout life (including smoking, illness, and socioeconomic deprivation). In this meta-analysis of existing longitudinal studies, the hypothesis that lower pulmonary function and respiratory illness are linked to an elevated risk of dementia was tested. METHODS A systematic review was conducted of longitudinal studies using PubMed until April 1, 2019, and, where possible, results were pooled in random effects meta-analyses. RESULTS Ten studies relating pulmonary function to later dementia risk and 11 studies of respiratory illness and dementia (including one that assessed both factors) were identified. The lowest quartile of FEV1 compared with the highest was associated with a 1.4-fold (hazard ratio [HR], 1.46; 95% CI, 0.77-2.75) increased dementia risk (Ntotal = 62,209; two studies). A decrease of 1 SD in FEV1 was associated with a 28% increase in dementia risk (HR, 1.28; 95% CI, 1.03-1.60; Ntotal = 67,505; six studies). Respiratory illness was also associated with increased dementia risk to a similar degree (pooled HR, 1.54; 95% CI, 1.30-1.81; Ntotal = 288,641; 11 studies). CONCLUSIONS Individuals with poor pulmonary function experience an increased risk of dementia. The extent to which the association between poor pulmonary function and dementia is causal remains unclear and requires examination.
Collapse
Affiliation(s)
- Tom C Russ
- Alzheimer Scotland Dementia Research Centre, University of Edinburgh, Edinburgh, UK; Centre for Cognitive Ageing & Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK; School of Philosophy, Psychology, and Language Sciences; and Edinburgh Dementia Prevention, University of Edinburgh, Edinburgh, UK; Division of Psychiatry, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK.
| | - Mika Kivimäki
- Department of Epidemiology and Public Health, University College, London, United Kingdom
| | - G David Batty
- Alzheimer Scotland Dementia Research Centre, University of Edinburgh, Edinburgh, UK; Centre for Cognitive Ageing & Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK; Department of Epidemiology and Public Health, University College, London, United Kingdom; School of Biological & Population Health Sciences, Oregon State University, Corvallis, OR
| |
Collapse
|
41
|
Boiko AS, Mednova IA, Kornetova EG, Bokhan NA, Semke AV, Loonen AJM, Ivanova SA. Cortisol and DHEAS Related to Metabolic Syndrome in Patients with Schizophrenia. Neuropsychiatr Dis Treat 2020; 16:1051-1058. [PMID: 32368067 PMCID: PMC7184116 DOI: 10.2147/ndt.s247161] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 03/28/2020] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Both dehydroepiandrosterone (DHEAS) and cortisol are secreted by the adrenal glands and may modulate metabolic syndrome (MetS), which often affects the health of patients with schizophrenia. The relationship between the serum levels of these hormones and MetS has not been established. PURPOSE In this pilot study, we investigated the serum levels in schizophrenia patients with and without MetS and compared them with those in healthy volunteers. PATIENTS AND METHODS After obtaining informed consent, 110 patients with acute paranoid schizophrenia were recruited directly after admission to the Mental Health Research Institute. The control group consisted of 51 persons reported on questioning to be mentally and somatically healthy. Blood samples to prepare serum were drawn after an 8-h overnight fast during one of the first days of admission. Serum cortisol and DHEAS concentrations were quantified by enzyme-linked immunosorbent assay. RESULTS A total of 42 patients had MetS and 68 patients were without MetS. The cortisol blood level was significantly (p = 0.012) higher in schizophrenia patients without MetS in comparison to healthy controls, while patients with schizophrenia and a MetS have significantly (p = 0.014) lower DHEAS levels than healthy volunteers. These differences could, however, exclusively be attributed to female participants. Analysis of covariance adjusted for gender and age demonstrated a significant relationship between age and DHEAS levels (F = 9.512, р = 0.003). CONCLUSION Lower DHEAS serum levels in relationship to MetS become evident in women, but not in men, and have age differences as a confounding factor.
Collapse
Affiliation(s)
- Anastasiia S Boiko
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russian Federation
| | - Irina A Mednova
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russian Federation
| | - Elena G Kornetova
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russian Federation.,Hospital, Siberian State Medical University, Tomsk, Russian Federation
| | - Nikolay A Bokhan
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russian Federation.,Psychiatry, Addictology and Psychotherapy, Siberian State Medical University, Russian Federation
| | - Arkadiy V Semke
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russian Federation
| | - Anton J M Loonen
- PharmacoTherapy, -Epidemiology and -Economics, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Svetlana A Ivanova
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russian Federation.,Psychiatry, Addictology and Psychotherapy, Siberian State Medical University, Russian Federation
| |
Collapse
|
42
|
Hemonnot AL, Hua J, Ulmann L, Hirbec H. Microglia in Alzheimer Disease: Well-Known Targets and New Opportunities. Front Aging Neurosci 2019; 11:233. [PMID: 31543810 PMCID: PMC6730262 DOI: 10.3389/fnagi.2019.00233] [Citation(s) in RCA: 187] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 08/14/2019] [Indexed: 12/21/2022] Open
Abstract
Microglia are the resident macrophages of the central nervous system. They play key roles in brain development, and physiology during life and aging. Equipped with a variety of molecular sensors and through the various functions they can fulfill, they are critically involved in maintaining the brain’s homeostasis. In Alzheimer disease (AD), microglia reaction was initially thought to be incidental and triggered by amyloid deposits and dystrophic neurites. However, recent genome-wide association studies have established that the majority of AD risk loci are found in or near genes that are highly and sometimes uniquely expressed in microglia. This leads to the concept of microglia being critically involved in the early steps of the disease and identified them as important potential therapeutic targets. Whether microglia reaction is beneficial, detrimental or both to AD progression is still unclear and the subject of intense debate. In this review, we are presenting a state-of-knowledge report intended to highlight the variety of microglial functions and pathways shown to be critically involved in AD progression. We first address both the acquisition of new functions and the alteration of their homeostatic roles by reactive microglia. Second, we propose a summary of new important parameters currently emerging in the field that need to be considered to identify relevant microglial targets. Finally, we discuss the many obstacles in designing efficient therapeutic strategies for AD and present innovative technologies that may foster our understanding of microglia roles in the pathology. Ultimately, this work aims to fly over various microglial functions to make a general and reliable report of the current knowledge regarding microglia’s involvement in AD and of the new research opportunities in the field.
Collapse
Affiliation(s)
- Anne-Laure Hemonnot
- Institute for Functional Genomics (IGF), University of Montpellier, Centre National de la Recherche Scientififique, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
| | - Jennifer Hua
- Institute for Functional Genomics (IGF), University of Montpellier, Centre National de la Recherche Scientififique, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
| | - Lauriane Ulmann
- Institute for Functional Genomics (IGF), University of Montpellier, Centre National de la Recherche Scientififique, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
| | - Hélène Hirbec
- Institute for Functional Genomics (IGF), University of Montpellier, Centre National de la Recherche Scientififique, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
| |
Collapse
|
43
|
Boiko AS, Mednova IA, Kornetova EG, Semke AV, Bokhan NA, Loonen AJ, Ivanova SA. Apolipoprotein serum levels related to metabolic syndrome in patients with schizophrenia. Heliyon 2019; 5:e02033. [PMID: 31317083 PMCID: PMC6611937 DOI: 10.1016/j.heliyon.2019.e02033] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 05/05/2019] [Accepted: 06/28/2019] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Schizophrenia is associated with a lowered life expectancy due to cardiovascular disease. This is, at least in part, related to an increased vulnerability to the development of metabolic syndrome (MetS) in patients with schizophrenia. The dysregulation of apolipoproteins (Apos) may also play a role in the pathogenesis of schizophrenia via their effect on cerebral cholesterol processing. AIM The aim of this study was to investigate serum Apos A1, C3, E, A2 and C2 concentration in schizophrenia patients with or without MetS in comparison to healthy donors. METHODS After obtaining informed consent, 53 patients with a diagnosis of paranoid schizophrenia according to ICD-10 criteria (F20) were included. Patients were divided into two groups with (N = 26) and without (N = 27) MetS according to the criteria of the International Diabetes Federation. The control group included 20 mentally and physically healthy subjects. Serum Apos A1, A2, C2, C3 and E were measured using xMAP technology (Luminex). RESULTS Serum ApoA1 was significantly decreased in patients with schizophrenia compared to healthy subjects (p = 0.002); ApoA2 was lower in patients without MetS in comparison to patients with MetS (p = 0.017) and the levels of ApoC3 and ApoC2 were increased in patients with schizophrenia with MetS in comparison with the control group and also with patients without MetS. No other significant differences were established concerning the other assayed apolipoproteins. CONCLUSIONS In line with literature data the results of our study suggest that while disturbances in ApoA1 level may play a role in the pathogenesis of schizophrenia, ApoA2, ApoC2, ApoC3 and ApoE may be primarily related to metabolic imbalance.
Collapse
Affiliation(s)
- Anastasiia S. Boiko
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Aleutskaya str., 4, Tomsk, Russian Federation
| | - Irina A. Mednova
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Aleutskaya str., 4, Tomsk, Russian Federation
| | - Elena G. Kornetova
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Aleutskaya str., 4, Tomsk, Russian Federation
- Siberian State Medical University, Moscowsky Trakt, 2, Tomsk, Russian Federation
| | - Arkadiy V. Semke
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Aleutskaya str., 4, Tomsk, Russian Federation
| | - Nikolay A. Bokhan
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Aleutskaya str., 4, Tomsk, Russian Federation
- National Research Tomsk State University, Lenin Avenue, 36, Tomsk, Russian Federation
- Siberian State Medical University, Moscowsky Trakt, 2, Tomsk, Russian Federation
| | - Anton J.M. Loonen
- University of Groningen, Groningen Research Institute of Pharmacy, PharmacoTherapy, Epidemiology &Economics, Antonius Deusinglaan 1, 9713 AV, Groningen, the Netherlands
- GGZ Westelijk Noord-Brabant, Hoofdlaan 8, 4661 AA, Halsteren, the Netherlands
| | - Svetlana A. Ivanova
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Aleutskaya str., 4, Tomsk, Russian Federation
- National Research Tomsk Polytechnic University, Lenin Avenue, 30, Tomsk, Russian Federation
- Siberian State Medical University, Moscowsky Trakt, 2, Tomsk, Russian Federation
| |
Collapse
|
44
|
Tzioras M, Davies C, Newman A, Jackson R, Spires‐Jones T. Invited Review: APOE at the interface of inflammation, neurodegeneration and pathological protein spread in Alzheimer's disease. Neuropathol Appl Neurobiol 2019; 45:327-346. [PMID: 30394574 PMCID: PMC6563457 DOI: 10.1111/nan.12529] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 10/27/2018] [Indexed: 12/13/2022]
Abstract
Despite more than a century of research, the aetiology of sporadic Alzheimer's disease (AD) remains unclear and finding disease modifying treatments for AD presents one of the biggest medical challenges of our time. AD pathology is characterized by deposits of aggregated amyloid beta (Aβ) in amyloid plaques and aggregated tau in neurofibrillary tangles. These aggregates begin in distinct brain regions and spread throughout the brain in stereotypical patterns. Neurodegeneration, comprising loss of synapses and neurons, occurs in brain regions with high tangle pathology, and an inflammatory response of glial cells appears in brain regions with pathological aggregates. Inheriting an apolipoprotein E ε4 (APOE4) allele strongly increases the risk of developing AD for reasons that are not yet entirely clear. Substantial amounts of evidence support a role for APOE in modulating the aggregation and clearance of Aβ, and data have been accumulating recently implicating APOE4 in exacerbating neurodegeneration, tau pathology and inflammation. We hypothesize that APOE4 influences all the pathological hallmarks of AD and may sit at the interface between neurodegeneration, inflammation and the spread of pathologies through the brain. Here, we conducted a systematic search of the literature and review evidence supporting a role for APOE4 in neurodegeneration and inflammation. While there is no direct evidence yet for APOE4 influencing the spread of pathology, we postulate that this may be found in future based on the literature reviewed here. In conclusion, this review highlights the importance of understanding the role of APOE in multiple important pathological mechanisms in AD.
Collapse
Affiliation(s)
- M. Tzioras
- UK Dementia Research Institute and Centre for Discovery Brain SciencesThe University of EdinburghEdinburghUK
| | - C. Davies
- UK Dementia Research Institute and Centre for Discovery Brain SciencesThe University of EdinburghEdinburghUK
| | - A. Newman
- UK Dementia Research Institute and Centre for Discovery Brain SciencesThe University of EdinburghEdinburghUK
| | - R. Jackson
- UK Dementia Research Institute and Centre for Discovery Brain SciencesThe University of EdinburghEdinburghUK
- Massachusetts General Hospital and Harvard Medical SchoolCharlestownMAUSA
| | - T. Spires‐Jones
- UK Dementia Research Institute and Centre for Discovery Brain SciencesThe University of EdinburghEdinburghUK
| |
Collapse
|
45
|
Finneran DJ, Nash KR. Neuroinflammation and fractalkine signaling in Alzheimer's disease. J Neuroinflammation 2019; 16:30. [PMID: 30744705 PMCID: PMC6371521 DOI: 10.1186/s12974-019-1412-9] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 01/23/2019] [Indexed: 02/07/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive, neurodegenerative disorder, and the most common form of dementia. As the understanding of AD has progressed, it is now believed that AD is an amyloid-initiated tauopathy with neuroinflammation serving as the link between amyloid deposition, tau pathology, and neurodegeneration. As microglia are the main immune effectors in the central nervous system, they have been the focus of attention in studies investigating the neuroinflammatory component of AD. Therefore, recent work has focused on immunomodulators, which can alter microglial activation without suppressing activity, as potential therapeutics for AD. Fractalkine (CX3CL1; FKN), a unique chemokine with a one-to-one relationship with its receptor, signals through its cognate receptor (CX3CR1) to reduce expression of pro-inflammatory genes in activated microglia. Disrupting FKN signaling has opposing effects on the two hallmark pathologies of AD, but over-expressing a soluble FKN has been shown to reduce tau pathology while not altering amyloid pathology. Recently, differential signaling has been reported when comparing two cleavage variants of soluble FKN. These differential effects may explain recent studies reporting seemingly conflicting results regarding the effect of FKN over expression on AD pathologies.
Collapse
Affiliation(s)
- Dylan J Finneran
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Bvld, Tampa, FL, 33612, USA.
| | - Kevin R Nash
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Bvld, Tampa, FL, 33612, USA.
| |
Collapse
|
46
|
Fernandez CG, Hamby ME, McReynolds ML, Ray WJ. The Role of APOE4 in Disrupting the Homeostatic Functions of Astrocytes and Microglia in Aging and Alzheimer's Disease. Front Aging Neurosci 2019; 11:14. [PMID: 30804776 PMCID: PMC6378415 DOI: 10.3389/fnagi.2019.00014] [Citation(s) in RCA: 154] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 01/16/2019] [Indexed: 12/20/2022] Open
Abstract
APOE4 is the greatest genetic risk factor for late-onset Alzheimer’s disease (AD), increasing the risk of developing the disease by 3-fold in the 14% of the population that are carriers. Despite 25 years of research, the exact mechanisms underlying how APOE4 contributes to AD pathogenesis remain incompletely defined. APOE in the brain is primarily expressed by astrocytes and microglia, cell types that are now widely appreciated to play key roles in the pathogenesis of AD; thus, a picture is emerging wherein APOE4 disrupts normal glial cell biology, intersecting with changes that occur during normal aging to ultimately cause neurodegeneration and cognitive dysfunction. This review article will summarize how APOE4 alters specific pathways in astrocytes and microglia in the context of AD and the aging brain. APOE itself, as a secreted lipoprotein without enzymatic activity, may prove challenging to directly target therapeutically in the classical sense. Therefore, a deeper understanding of the underlying pathways responsible for APOE4 toxicity is needed so that more tractable pathways and drug targets can be identified to reduce APOE4-mediated disease risk.
Collapse
Affiliation(s)
- Celia G Fernandez
- The Neurodegeneration Consortium, Institute of Applied Cancer Science (IACS), The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Mary E Hamby
- The Neurodegeneration Consortium, Institute of Applied Cancer Science (IACS), The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Morgan L McReynolds
- The Neurodegeneration Consortium, Institute of Applied Cancer Science (IACS), The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - William J Ray
- The Neurodegeneration Consortium, Institute of Applied Cancer Science (IACS), The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
47
|
Mishra A, Brinton RD. Inflammation: Bridging Age, Menopause and APOEε4 Genotype to Alzheimer's Disease. Front Aging Neurosci 2018; 10:312. [PMID: 30356809 PMCID: PMC6189518 DOI: 10.3389/fnagi.2018.00312] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 09/18/2018] [Indexed: 12/19/2022] Open
Abstract
Neuro-inflammatory processes that contribute to development of Alzheimer’s are evident early in the latent prodromal phase and worsen during the course of the disease. Despite substantial mechanistic and clinical evidence of inflammation, therapeutic approaches targeting inflammation have failed to alter the course of the disease. Disparate results from epidemiological and clinical trials targeting inflammation, highlight the complexity of the inflammatory process. Herein we review the dynamics of the inflammatory process across aging, midlife endocrine transitions, and the APOEε4 genotype and their contribution to progression of Alzheimer’s disease (AD). We discuss the chronic inflammatory processes that are activated during midlife chronological and endocrine aging, which ultimately limit the clearance capacity of microglia and lead to immune senescence. Aging, menopause, and APOEε4 combine the three hits of a compromised bioenergetic system of menopause with the chronic low grade innate inflammation of aging with the APOEε4 dyslipidemia and adaptive immune response. The inflammatory immune response is the unifying factor that bridges across each of the risk factors for AD. Immune system regulators that are specific to stage of disease and inflammatory phenotype would provide a therapeutic strategy to disconnect the bridge that drives disease. Outcomes of this analysis provide plausible mechanisms underlying failed clinical trials of anti-inflammatory agents in Alzheimer’s patients. Further, they highlight the need for stratifying AD clinical trial cohorts based on inflammatory phenotype. Combination therapies that include targeted use of anti-inflammatory agent’s specific to the immune phenotype are considered.
Collapse
Affiliation(s)
- Aarti Mishra
- Titus Family Department of Clinical Pharmacy, School of Pharmacy, University of Southern California, Los Angeles, CA, United States.,Center for Innovation in Brain Science, University of Arizona, Tucson, AZ, United States
| | - Roberta D Brinton
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ, United States.,Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, United States.,Department of Neurology, College of Medicine, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
48
|
Newcombe EA, Camats-Perna J, Silva ML, Valmas N, Huat TJ, Medeiros R. Inflammation: the link between comorbidities, genetics, and Alzheimer's disease. J Neuroinflammation 2018; 15:276. [PMID: 30249283 PMCID: PMC6154824 DOI: 10.1186/s12974-018-1313-3] [Citation(s) in RCA: 336] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 09/11/2018] [Indexed: 12/21/2022] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disorder, most cases of which lack a clear causative event. This has made the disease difficult to characterize and, thus, diagnose. Although some cases are genetically linked, there are many diseases and lifestyle factors that can lead to an increased risk of developing AD, including traumatic brain injury, diabetes, hypertension, obesity, and other metabolic syndromes, in addition to aging. Identifying common factors and trends between these conditions could enhance our understanding of AD and lead to the development of more effective treatments. Although the immune system is one of the body’s key defense mechanisms, chronic inflammation has been increasingly linked with several age-related diseases. Moreover, it is now well accepted that chronic inflammation has an important role in the onset and progression of AD. In this review, the different inflammatory signals associated with AD and its risk factors will be outlined to demonstrate how chronic inflammation may be influencing individual susceptibility to AD. Our goal is to bring attention to potential shared signals presented by the immune system during different conditions that could lead to the development of successful treatments.
Collapse
Affiliation(s)
- Estella A Newcombe
- Neurula Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Building 79, Brisbane, 4072, QLD, Australia.
| | - Judith Camats-Perna
- Neurula Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Building 79, Brisbane, 4072, QLD, Australia
| | - Mallone L Silva
- Neurula Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Building 79, Brisbane, 4072, QLD, Australia
| | - Nicholas Valmas
- Queensland Brain Institute, The University of Queensland, Brisbane, 4072, QLD, Australia
| | - Tee Jong Huat
- Neurula Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Building 79, Brisbane, 4072, QLD, Australia.,Centre for Stem Cell Ageing and Regenerative Engineering, The University of Queensland, Brisbane, 4072, QLD, Australia
| | - Rodrigo Medeiros
- Neurula Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Building 79, Brisbane, 4072, QLD, Australia.
| |
Collapse
|
49
|
Martiskainen H, Takalo M, Solomon A, Stančáková A, Marttinen M, Natunen T, Haapasalo A, Herukka SK, Kuusisto J, Soininen H, Kivipelto M, Laakso M, Hiltunen M. Decreased plasma C-reactive protein levels in APOE ε4 allele carriers. Ann Clin Transl Neurol 2018; 5:1229-1240. [PMID: 30349858 PMCID: PMC6186931 DOI: 10.1002/acn3.639] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 08/07/2018] [Accepted: 08/10/2018] [Indexed: 02/06/2023] Open
Abstract
Objective Apolipoprotein E (APOE) ε4 allele is a well‐established risk factor in Alzheimer's disease (AD). Here, we assessed the effects of APOE polymorphism on cardiovascular, metabolic, and inflammation‐related parameters in population‐based cohorts. Methods Association of cardiovascular, metabolic, and inflammation‐related parameters with the APOE polymorphism in a large Finnish Metabolic Syndrome in Men (METSIM) cohort and Finnish Geriatric Intervention study to prevent cognitive impairment and disability (FINGER) were investigated. Brain‐specific effects were addressed in postmortem brain samples. Results Individuals carrying the APOE ε4 allele displayed significantly elevated serum/plasma LDL cholesterol and apolipoprotein B levels. APOE ε3ε4 and ε4ε4 significantly associated with lower levels of plasma high‐sensitivity C‐reactive protein (hs‐CRP). Plasma amyloid‐β 42 (Aβ42) and reduced hs‐CRP levels showed an association independently of the APOE status. Interpretation These data suggest that the APOE ε4 allele associates with lower levels of hs‐CRP in individuals without dementia. Moreover, Aβ42 may encompass anti‐inflammatory effects reflected by reduced hs‐CRP levels.
Collapse
Affiliation(s)
- Henna Martiskainen
- Institute of Clinical Medicine Internal Medicine University of Eastern Finland P.O. Box 1627 Kuopio 70211 Finland
| | - Mari Takalo
- Institute of Biomedicine University of Eastern Finland Yliopistonranta 1 E, P.O. Box 1627 Kuopio 70211 Finland
| | - Alina Solomon
- Department of Neurology Institute of Clinical Medicine University of Eastern Finland P.O. Box 1627 Kuopio 70211 Finland.,Division of Clinical Geriatrics Center for Alzheimer Research NVS, Karolinska Institutet Novum 5th floor Huddinge 14157 Sweden
| | - Alena Stančáková
- Institute of Clinical Medicine Internal Medicine University of Eastern Finland P.O. Box 1627 Kuopio 70211 Finland
| | - Mikael Marttinen
- Institute of Biomedicine University of Eastern Finland Yliopistonranta 1 E, P.O. Box 1627 Kuopio 70211 Finland
| | - Teemu Natunen
- Institute of Biomedicine University of Eastern Finland Yliopistonranta 1 E, P.O. Box 1627 Kuopio 70211 Finland
| | - Annakaisa Haapasalo
- A.I Virtanen Institute for Molecular Sciences University of Eastern Finland Neulaniementie 2 Kuopio 70211 Finland
| | - Sanna-Kaisa Herukka
- Department of Neurology Institute of Clinical Medicine University of Eastern Finland P.O. Box 1627 Kuopio 70211 Finland.,Neurocenter Neurology Kuopio University Hospital Kuopio Finland
| | - Johanna Kuusisto
- Institute of Clinical Medicine Internal Medicine University of Eastern Finland P.O. Box 1627 Kuopio 70211 Finland.,Kuopio University Hospital Kuopio Finland
| | - Hilkka Soininen
- Department of Neurology Institute of Clinical Medicine University of Eastern Finland P.O. Box 1627 Kuopio 70211 Finland.,Neurocenter Neurology Kuopio University Hospital Kuopio Finland
| | - Miia Kivipelto
- Department of Neurology Institute of Clinical Medicine University of Eastern Finland P.O. Box 1627 Kuopio 70211 Finland.,Division of Clinical Geriatrics Center for Alzheimer Research NVS, Karolinska Institutet Novum 5th floor Huddinge 14157 Sweden.,Department of Public Health Solutions Public Health Promotion Unit National Institute for Health and Welfare PO Box 30 Helsinki 00271 Finland
| | - Markku Laakso
- Institute of Clinical Medicine Internal Medicine University of Eastern Finland P.O. Box 1627 Kuopio 70211 Finland.,Kuopio University Hospital Kuopio Finland
| | - Mikko Hiltunen
- Institute of Biomedicine University of Eastern Finland Yliopistonranta 1 E, P.O. Box 1627 Kuopio 70211 Finland
| |
Collapse
|
50
|
Deng H, Ordaz A, Upadhyayula PS, Gillis-Buck EM, Suen CG, Melhado CG, Mohammed N, Lam T, Yue JK. Apolipoprotein E Epsilon 4 Genotype, Mild Traumatic Brain Injury, and the Development of Chronic Traumatic Encephalopathy. Med Sci (Basel) 2018; 6:E78. [PMID: 30223506 PMCID: PMC6163513 DOI: 10.3390/medsci6030078] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 09/04/2018] [Accepted: 09/12/2018] [Indexed: 12/14/2022] Open
Abstract
The annual incidence of mild traumatic brain injury (MTBI) is 3.8 million in the USA with 10⁻15% experiencing persistent morbidity beyond one year. Chronic traumatic encephalopathy (CTE), a neurodegenerative disease characterized by accumulation of hyperphosphorylated tau, can occur with repetitive MTBI. Risk factors for CTE are challenging to identify because injury mechanisms of MTBI are heterogeneous, clinical manifestations and management vary, and CTE is a postmortem diagnosis, making prospective studies difficult. There is growing interest in the genetic influence on head trauma and development of CTE. Apolipoprotein epsilon 4 (APOE-ε4) associates with many neurologic diseases, and consensus on the ε4 allele as a risk factor is lacking. This review investigates the influence of APOE-ε4 on MTBI and CTE. A comprehensive PubMed literature search (1966 to 12 June 2018) identified 24 unique reports on the topic (19 MTBI studies: 8 athletic, 5 military, 6 population-based; 5 CTE studies: 4 athletic and military, 1 leucotomy group). APOE-ε4 genotype is found to associate with outcomes in 4/8 athletic reports, 3/5 military reports, and 5/6 population-based reports following MTBI. Evidence on the association between APOE-ε4 and CTE from case series is equivocal. Refining modalities to aid CTE diagnosis in larger samples is needed in MTBI.
Collapse
Affiliation(s)
- Hansen Deng
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94122, USA.
| | - Angel Ordaz
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94122, USA.
| | - Pavan S Upadhyayula
- Department of Neurological Surgery, University of California San Diego, San Diego, CA 92093, USA.
| | - Eva M Gillis-Buck
- Department of Surgery, University of California San Francisco, San Francisco, CA 94122, USA.
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| | - Catherine G Suen
- Department of Neurology, University of Utah, Salt Lake City, UT 84112, USA.
| | - Caroline G Melhado
- Department of Surgery, University of California San Francisco, San Francisco, CA 94122, USA.
| | - Nebil Mohammed
- Department of Pathology, University of California San Francisco, San Francisco, CA 94122, USA.
| | - Troy Lam
- Department of Oral and Maxillofacial Surgery, University of California San Francisco, San Francisco, CA 94122, USA.
| | - John K Yue
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94122, USA.
| |
Collapse
|