1
|
Karan KR, Andrzejewski S, Stiles KM, Hackett NR, Crystal RG. Suppression of CNS APOE4 Expression by miRNAs Delivered by the S2 AAVrh.10 Capsid-Modified AAV Vector. Hum Gene Ther 2024. [PMID: 39318239 DOI: 10.1089/hum.2024.112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2024] Open
Abstract
The homozygous Apolipoprotein E (APOE4) genotype is the major risk factor for the development of early Alzheimer's disease. Genome engineering studies in mouse models of human APOE4-dependent pathology have established that reduction of APOE4 expression can rescue the phenotype. We hypothesized that APOE4 could be suppressed in the CNS of APOE4 homozygotes using adeno-associated virus (AAV) expression of microRNAs (miRNA) designed to hybridize to APOE mRNA. We screened nine different miRNAs targeting APOE following transfection in HEK293T and Huh7 cells. Optimal APOE suppression was obtained with mir2A (targeting coding region nt330-351) and mirN4 (3' untranslated region nt1142-1162). miRNA expression cassettes were designed with two copies of each of these two miRNAs co-expressed with a mCherry transgene. To optimize delivery of these miRNAs, an engineered AAVrh.10 variant was identified from a screen of multiple peptide insertions into capsid loop IV and substitutions in loop VIII. This led to identifying the AAV.S2 capsid with enhanced transduction of both neurons and glia and enhanced distribution in the brain. The engineered capsid was used to deliver the APOE miRNA suppression cassette to the hippocampus of TRE4 mice (human APOE4 knock-in replacement of the murine apoE locus). Two weeks after intra-hippocampus administration, regional expression of miRNA at the injection site was quantified at the mRNA level relative to an endogenous reference. The AAV.S2 capsid provided 2.31 ± 0.37-fold higher expression of miRNA over that provided by AAVrh.10 (p < 0.05). In the targeted region, a single intra-hippocampus AAV.S2 administration suppressed hippocampal APOE4 mRNA levels by 76.5 ± 3.9% compared with 41.3 ± 3.3% with the same cassette delivered by the wildtype AAVrh.10 capsid (p < 0.0001). We conclude that an expression cassette with two different miRNAs targeting APOE4 delivered by the AAV.S2 capsid will generate highly significant suppression of APOE4 in the CNS.
Collapse
Affiliation(s)
- Kalpita R Karan
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Slawomir Andrzejewski
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Katie M Stiles
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Neil R Hackett
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Ronald G Crystal
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York, USA
| |
Collapse
|
2
|
Laslo A, Laslo L, Arbănași EM, Ujlaki-Nagi AA, Chinezu L, Ivănescu AD, Arbănași EM, Cărare RO, Cordoș BA, Popa IA, Brînzaniuc K. Pathways to Alzheimer's Disease: The Intersecting Roles of Clusterin and Apolipoprotein E in Amyloid-β Regulation and Neuronal Health. PATHOPHYSIOLOGY 2024; 31:545-558. [PMID: 39449522 PMCID: PMC11503414 DOI: 10.3390/pathophysiology31040040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 09/23/2024] [Accepted: 09/27/2024] [Indexed: 10/26/2024] Open
Abstract
One of the hallmarks of Alzheimer's disease (AD) is the deposition of amyloid-β (Aβ) within the extracellular spaces of the brain as plaques and along the blood vessels in the brain, a condition also known as cerebral amyloid angiopathy (CAA). Clusterin (CLU), or apolipoprotein J (APOJ), is a multifunctional glycoprotein that has a role in many physiological and neurological conditions, including AD. The apolipoprotein E (APOE) is a significant genetic factor in AD, and while the primary physiological role of APOE in the brain and peripheral tissues is to regulate lipid transport, it also participates in various other biological processes, having three basic human forms: APOE2, APOE3, and APOE4. Notably, the APOE4 allele substantially increases the risk of developing late-onset AD. The main purpose of this review is to examine the roles of CLU and APOE in AD pathogenesis in order to acquire a better understanding of AD pathogenesis from which to develop targeted therapeutic approaches.
Collapse
Affiliation(s)
- Alexandru Laslo
- Department of Urology, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540139 Targu Mures, Romania;
- Department of Anatomy, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540139 Targu Mures, Romania; (A.D.I.); (K.B.)
- Doctoral School of Medicine and Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540139 Targu Mures, Romania;
| | - Laura Laslo
- Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540139 Targu Mures, Romania; (L.L.); (B.A.C.)
| | - Eliza-Mihaela Arbănași
- Doctoral School of Medicine and Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540139 Targu Mures, Romania;
- Regenerative Medicine Laboratory, Centre for Advanced Medical and Pharmaceutical Research (CCAMF), George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540139 Targu Mures, Romania
| | | | - Laura Chinezu
- Department of Histology, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540139 Targu Mures, Romania;
| | - Adrian Dumitru Ivănescu
- Department of Anatomy, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540139 Targu Mures, Romania; (A.D.I.); (K.B.)
| | - Emil-Marian Arbănași
- Doctoral School of Medicine and Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540139 Targu Mures, Romania;
- Regenerative Medicine Laboratory, Centre for Advanced Medical and Pharmaceutical Research (CCAMF), George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540139 Targu Mures, Romania
- Department of Vascular Surgery, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540139 Targu Mures, Romania
- Clinic of Vascular Surgery, Mures County Emergency Hospital, 540136 Targu Mures, Romania
| | | | - Bogdan Andrei Cordoș
- Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540139 Targu Mures, Romania; (L.L.); (B.A.C.)
- Centre for Experimental Medical and Imaging Studies, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540139 Targu Mures, Romania
| | - Ioana Adriana Popa
- Clinic of Radiology, Mures County Emergency Hospital, 540136 Targu Mures, Romania;
| | - Klara Brînzaniuc
- Department of Anatomy, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540139 Targu Mures, Romania; (A.D.I.); (K.B.)
| |
Collapse
|
3
|
Anfray A, Schaeffer S, Hattori Y, Santisteban MM, Casey N, Wang G, Strickland M, Zhou P, Holtzman DM, Anrather J, Park L, Iadecola C. A cell-autonomous role for border-associated macrophages in ApoE4 neurovascular dysfunction and susceptibility to white matter injury. Nat Neurosci 2024:10.1038/s41593-024-01757-6. [PMID: 39294490 DOI: 10.1038/s41593-024-01757-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 08/07/2024] [Indexed: 09/20/2024]
Abstract
Apolipoprotein E4 (ApoE4), the strongest genetic risk factor for sporadic Alzheimer's disease, is also a risk factor for microvascular pathologies leading to cognitive impairment, particularly subcortical white matter injury. These effects have been attributed to alterations in the regulation of the brain blood supply, but the cellular source of ApoE4 and the underlying mechanisms remain unclear. In mice expressing human ApoE3 or ApoE4, we report that border-associated macrophages (BAMs), myeloid cells closely apposed to neocortical microvessels, are both sources and effectors of ApoE4 mediating the neurovascular dysfunction through reactive oxygen species. ApoE4 in BAMs is solely responsible for the increased susceptibility to oligemic white matter damage in ApoE4 mice and is sufficient to enhance damage in ApoE3 mice. The data unveil a new aspect of BAM pathobiology and highlight a previously unrecognized cell-autonomous role of BAM in the neurovascular dysfunction of ApoE4 with potential therapeutic implications.
Collapse
Affiliation(s)
- Antoine Anfray
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Samantha Schaeffer
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Yorito Hattori
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Monica M Santisteban
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Nicole Casey
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Gang Wang
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Michael Strickland
- Department 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
| | - Ping Zhou
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - David M Holtzman
- Department 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
| | - Josef Anrather
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Laibaik Park
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
4
|
Chen F, Zhao J, Meng F, He F, Ni J, Fu Y. The vascular contribution of apolipoprotein E to Alzheimer's disease. Brain 2024; 147:2946-2965. [PMID: 38748848 DOI: 10.1093/brain/awae156] [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: 12/24/2023] [Revised: 03/23/2024] [Accepted: 04/21/2024] [Indexed: 09/04/2024] Open
Abstract
Alzheimer's disease, the most prevalent form of dementia, imposes a substantial societal burden. The persistent inadequacy of disease-modifying drugs targeting amyloid plaques and neurofibrillary tangles suggests the contribution of alternative pathogenic mechanisms. A frequently overlooked aspect is cerebrovascular dysfunction, which may manifest early in the progression of Alzheimer's disease pathology. Mounting evidence underscores the pivotal role of the apolipoprotein E gene, particularly the apolipoprotein ε4 allele as the strongest genetic risk factor for late-onset Alzheimer's disease, in the cerebrovascular pathology associated with Alzheimer's disease. In this review, we examine the evidence elucidating the cerebrovascular impact of both central and peripheral apolipoprotein E on the pathogenesis of Alzheimer's disease. We present a novel three-hit hypothesis, outlining potential mechanisms that shed light on the intricate relationship among different pathogenic events. Finally, we discuss prospective therapeutics targeting the cerebrovascular pathology associated with apolipoprotein E and explore their implications for future research endeavours.
Collapse
Affiliation(s)
- Feng Chen
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jing Zhao
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, China
| | - Fanxia Meng
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Fangping He
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jie Ni
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Yuan Fu
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| |
Collapse
|
5
|
Chandrashekar DV, Roules GC, Jagadeesan N, Panchal UR, Oyegbesan A, Imiruaye OE, Zhang H, Garcia J, Kaur K, Win S, Than TA, Kaplowitz N, Roosan MR, Han D, Sumbria RK. Hepatic LRP-1 plays an important role in amyloidosis in Alzheimer's disease mice: Potential role in chronic heavy alcohol feeding. Neurobiol Dis 2024; 199:106570. [PMID: 38885850 DOI: 10.1016/j.nbd.2024.106570] [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: 04/08/2024] [Revised: 06/13/2024] [Accepted: 06/14/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Hepatic lipoprotein receptor-related protein 1 (LRP-1) plays a central role in peripheral amyloid beta (Aβ) clearance, but its importance in Alzheimer's disease (AD) pathology is understudied. Our previous work showed that intragastric alcohol feeding to C57BL/6 J mice reduced hepatic LRP-1 expression which correlated with significant AD-relevant brain changes. Herein, we examined the role of hepatic LRP-1 in AD pathogenesis in APP/PS1 AD mice using two approaches to modulate hepatic LRP-1, intragastric alcohol feeding to model chronic heavy drinking shown by us to reduce hepatic LRP-1, and hepato-specific LRP-1 silencing. METHODS Eight-month-old male APP/PS1 mice were fed ethanol or control diet intragastrically for 5 weeks (n = 7-11/group). Brain and liver Aβ were assessed using immunoassays. Three important mechanisms of brain amyloidosis were investigated: hepatic LRP-1 (major peripheral Aβ regulator), blood-brain barrier (BBB) function (vascular Aβ regulator), and microglia (major brain Aβ regulator) using immunoassays. Spatial LRP-1 gene expression in the periportal versus pericentral hepatic regions was confirmed using NanoString GeoMx Digital Spatial Profiler. Further, hepatic LRP-1 was silenced by injecting LRP-1 microRNA delivered by the adeno-associated virus 8 (AAV8) and the hepato-specific thyroxine-binding globulin (TBG) promoter to 4-month-old male APP/PS1 mice (n = 6). Control male APP/PS1 mice received control AAV8 (n = 6). Spatial memory and locomotion were assessed 12 weeks after LRP-1 silencing using Y-maze and open-field test, respectively, and brain and liver Aβ were measured. RESULTS Alcohol feeding reduced plaque-associated microglia in APP/PS1 mice brains and increased aggregated Aβ (p < 0.05) by ELISA and 6E10-positive Aβ load by immunostaining (p < 0.05). Increased brain Aβ corresponded with a significant downregulation of hepatic LRP-1 (p < 0.01) at the protein and transcript level, primarily in pericentral hepatocytes (zone 3) where alcohol-induced injury occurs. Hepato-specific LRP-1 silencing significantly increased brain Aβ and locomotion hyperactivity (p < 0.05) in APP/PS1 mice. CONCLUSION Chronic heavy alcohol intake reduced hepatic LRP-1 expression and increased brain Aβ. The hepato-specific LRP-1 silencing similarly increased brain Aβ which was associated with behavioral deficits in APP/PS1 mice. Collectively, our results suggest that hepatic LRP-1 is a key regulator of brain amyloidosis in alcohol-dependent AD.
Collapse
Affiliation(s)
- Devaraj V Chandrashekar
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, United States
| | - G Chuli Roules
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, United States
| | - Nataraj Jagadeesan
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, United States
| | - Urvashi R Panchal
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, United States
| | - Adenike Oyegbesan
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, United States
| | - Oghenetega E Imiruaye
- School of Pharmacy and Health Sciences, Keck Graduate Institute, Claremont, CA, United States
| | - Hai Zhang
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, United States
| | - Jerome Garcia
- Department of Biology, University of La Verne, La Verne, CA, United States
| | - Kamaljit Kaur
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, United States
| | - Sanda Win
- Department of Medicine, Division of Gastroenterology and Liver Diseases, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Tin A Than
- Department of Medicine, Division of Gastroenterology and Liver Diseases, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Neil Kaplowitz
- Department of Medicine, Division of Gastroenterology and Liver Diseases, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Moom R Roosan
- Pharmacy Practice, School of Pharmacy, Chapman University, Irvine, CA, United States
| | - Derick Han
- School of Pharmacy and Health Sciences, Keck Graduate Institute, Claremont, CA, United States.
| | - Rachita K Sumbria
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, United States; Department of Neurology, University of California, Irvine, CA, United States.
| |
Collapse
|
6
|
Wieg L, Ciola JC, Wasén CC, Gaba F, Colletti BR, Schroeder MK, Hinshaw RG, Ekwudo MN, Holtzman DM, Saito T, Sasaguri H, Saido TC, Cox LM, Lemere CA. Cognitive Effects of Simulated Galactic Cosmic Radiation Are Mediated by ApoE Status, Sex, and Environment in APP Knock-In Mice. Int J Mol Sci 2024; 25:9379. [PMID: 39273325 PMCID: PMC11394682 DOI: 10.3390/ijms25179379] [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: 08/15/2024] [Revised: 08/25/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024] Open
Abstract
Cosmic radiation experienced during space travel may increase the risk of cognitive impairment. While simulated galactic cosmic radiation (GCRsim) has led to memory deficits in wildtype (WT) mice, it has not been investigated whether GCRsim in combination with genetic risk factors for Alzheimer's disease (AD) worsens memory further in aging mice. Here, we investigated the central nervous system (CNS) effects of 0 Gy (sham) or 0.75 Gy five-ion GCRsim or 2 Gy gamma radiation (IRR) in 14-month-old female and male APPNL-F/NL-F knock-in (KI) mice bearing humanized ApoE3 or ApoE4 (APP;E3F and APP;E4F). As travel to a specialized facility was required for irradiation, both traveled sham-irradiated C57BL/6J WT and KI mice and non-traveled (NT) KI mice acted as controls for potential effects of travel. Mice underwent four behavioral tests at 20 months of age and were euthanized for pathological and biochemical analyses 1 month later. Fecal samples were collected pre- and post-irradiation at four different time points. GCRsim seemed to impair memory in male APP;E3F mice compared to their sham counterparts. Travel tended to improve cognition in male APP;E3F mice and lowered total Aβ in female and male APP;E3F mice compared to their non-traveled counterparts. Sham-irradiated male APP;E4F mice accumulated more fibrillar amyloid than their APP;E3F counterparts. Radiation exposure had only modest effects on behavior and brain changes, but travel-, sex-, and genotype-specific effects were seen. Irradiated mice had immediate and long-term differences in their gut bacterial composition that correlated to Alzheimer's disease phenotypes.
Collapse
Affiliation(s)
- Laura Wieg
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Jason C Ciola
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Caroline C Wasén
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Fidelia Gaba
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Brianna R Colletti
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Maren K Schroeder
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Robert G Hinshaw
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Millicent N Ekwudo
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Science, Nagoya 467-8601, Aichi, Japan
| | - Hiroki Sasaguri
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako City 351-0198, Saitama, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako City 351-0198, Saitama, Japan
| | - Laura M Cox
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA
- Department of Neurology, Harvard Medical School, Boston, MA 02115, USA
| | - Cynthia A Lemere
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA
- Department of Neurology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
7
|
Jackson RJ, Hyman BT, Serrano-Pozo A. Multifaceted roles of APOE in Alzheimer disease. Nat Rev Neurol 2024; 20:457-474. [PMID: 38906999 DOI: 10.1038/s41582-024-00988-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/24/2024] [Indexed: 06/23/2024]
Abstract
For the past three decades, apolipoprotein E (APOE) has been known as the single greatest genetic modulator of sporadic Alzheimer disease (AD) risk, influencing both the average age of onset and the lifetime risk of developing AD. The APOEε4 allele significantly increases AD risk, whereas the ε2 allele is protective relative to the most common ε3 allele. However, large differences in effect size exist across ethnoracial groups that are likely to depend on both global genetic ancestry and local genetic ancestry, as well as gene-environment interactions. Although early studies linked APOE to amyloid-β - one of the two culprit aggregation-prone proteins that define AD - in the past decade, mounting work has associated APOE with other neurodegenerative proteinopathies and broader ageing-related brain changes, such as neuroinflammation, energy metabolism failure, loss of myelin integrity and increased blood-brain barrier permeability, with potential implications for longevity and resilience to pathological protein aggregates. Novel mouse models and other technological advances have also enabled a number of therapeutic approaches aimed at either attenuating the APOEε4-linked increased AD risk or enhancing the APOEε2-linked AD protection. This Review summarizes this progress and highlights areas for future research towards the development of APOE-directed therapeutics.
Collapse
Affiliation(s)
- Rosemary J Jackson
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Bradley T Hyman
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, USA.
| | - Alberto Serrano-Pozo
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, USA.
| |
Collapse
|
8
|
Han SW, Lee SH, Kim JH, Lee JJ, Park YH, Kim S, Nho K, Sohn JH. Association of liver function markers and apolipoprotein E ε4 with pathogenesis and cognitive decline in Alzheimer's disease. Front Aging Neurosci 2024; 16:1411466. [PMID: 39114318 PMCID: PMC11303325 DOI: 10.3389/fnagi.2024.1411466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 07/09/2024] [Indexed: 08/10/2024] Open
Abstract
Background Alzheimer's disease (AD) is a complex neurodegenerative disorder influenced by various factors, including liver function, which may impact the clearance of amyloid-β (Aβ) in the brain. This study aimed to explore how the apolipoprotein E (APOE) ε4 allele affects the relationship of liver function markers with AD pathology and cognition. Methods We analyzed data from two independent cohorts, including 732 participants from the Hallym University Medical Center and 483 from the Alzheimer's Disease Neuroimaging Initiative, each group consisting of individuals with and without the APOE ε4 allele. Cross-sectional analyses evaluated the associations of liver enzymes (aspartate aminotransferase [AST], alanine aminotransferase [ALT], alkaline phosphatase, total bilirubin, and albumin) with AD diagnosis, amyloid positron emission tomography (PET) burden, and cerebrospinal fluid biomarkers for AD (Aβ42, total tau, and phosphorylated tau181) at baseline. Longitudinally, we investigated the associations between these liver enzymes and changes in cognitive performance over the course of a year. Logistic and linear regression models were used to analyze these associations and mediation analyses were conducted to assess whether age and amyloid PET burden mediated these associations. Results Only in the APOE ε4 carrier group, a high AST to ALT ratio and low ALT levels were significantly associated with AD diagnosis, increased amyloid PET burden, and faster longitudinal decline in cognitive function in both cohorts. In particular, the AST to ALT ratio was associated with cerebrospinal fluid Aβ42 levels exclusively in the APOE ε4 carrier group in the Alzheimer's Disease Neuroimaging Initiative cohort but not with phosphorylated tau181 or total tau levels. Moreover, mediation analyses from both cohorts revealed that in the APOE ε4 carriers group, age did not mediate the associations between liver enzymes and AD diagnosis or amyloid PET burden. However, amyloid PET burden partially mediated the association between liver enzymes and AD diagnosis exclusively in the APOE ε4 carriers group. Conclusion This study provides valuable insights into the significant association of the APOE ε4 allele with liver enzymes and their potential role in Aβ-related pathogenesis and cognition in AD. Further research is required to elucidate the underlying mechanisms and potential therapeutic implications of these findings.
Collapse
Affiliation(s)
- Sang-Won Han
- Department of Neurology, Chuncheon Sacred Heart Hospital, Hallym University College of Medicine, Chuncheon-si, Gangwon-do, Republic of Korea
| | - Sang-Hwa Lee
- Department of Neurology, Chuncheon Sacred Heart Hospital, Hallym University College of Medicine, Chuncheon-si, Gangwon-do, Republic of Korea
| | - Jong Ho Kim
- Department of Anesthesiology and Pain Medicine, Chuncheon Sacred Heart Hospital, Hallym University College of Medicine, Chuncheon-si, Gangwon-do, Republic of Korea
| | - Jae-Jun Lee
- Department of Anesthesiology and Pain Medicine, Chuncheon Sacred Heart Hospital, Hallym University College of Medicine, Chuncheon-si, Gangwon-do, Republic of Korea
- Institute of New Frontier Research Team, Hallym University, Chuncheon-si, Gangwon-do, Republic of Korea
| | - Young Ho Park
- Department of Neurology, Seoul National University College of Medicine and Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - SangYun Kim
- Department of Neurology, Seoul National University College of Medicine and Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Kwangsik Nho
- Department of Radiology and Imaging Sciences, Center for Computational Biology and Bioinformatics, Indiana Alzheimer’s Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Jong-Hee Sohn
- Department of Neurology, Chuncheon Sacred Heart Hospital, Hallym University College of Medicine, Chuncheon-si, Gangwon-do, Republic of Korea
- Institute of New Frontier Research Team, Hallym University, Chuncheon-si, Gangwon-do, Republic of Korea
| |
Collapse
|
9
|
Collins HM, Greenfield S. Rodent Models of Alzheimer's Disease: Past Misconceptions and Future Prospects. Int J Mol Sci 2024; 25:6222. [PMID: 38892408 PMCID: PMC11172947 DOI: 10.3390/ijms25116222] [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: 04/03/2024] [Revised: 05/28/2024] [Accepted: 06/03/2024] [Indexed: 06/21/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease with no effective treatments, not least due to the lack of authentic animal models. Typically, rodent models recapitulate the effects but not causes of AD, such as cholinergic neuron loss: lesioning of cholinergic neurons mimics the cognitive decline reminiscent of AD but not its neuropathology. Alternative models rely on the overexpression of genes associated with familial AD, such as amyloid precursor protein, or have genetically amplified expression of mutant tau. Yet transgenic rodent models poorly replicate the neuropathogenesis and protein overexpression patterns of sporadic AD. Seeding rodents with amyloid or tau facilitates the formation of these pathologies but cannot account for their initial accumulation. Intracerebral infusion of proinflammatory agents offer an alternative model, but these fail to replicate the cause of AD. A novel model is therefore needed, perhaps similar to those used for Parkinson's disease, namely adult wildtype rodents with neuron-specific (dopaminergic) lesions within the same vulnerable brainstem nuclei, 'the isodendritic core', which are the first to degenerate in AD. Site-selective targeting of these nuclei in adult rodents may recapitulate the initial neurodegenerative processes in AD to faithfully mimic its pathogenesis and progression, ultimately leading to presymptomatic biomarkers and preventative therapies.
Collapse
Affiliation(s)
- Helen M. Collins
- Neuro-Bio Ltd., Building F5 The Culham Campus, Abingdon OX14 3DB, UK;
| | | |
Collapse
|
10
|
Grenon MB, Papavergi MT, Bathini P, Sadowski M, Lemere CA. Temporal Characterization of the Amyloidogenic APPswe/PS1dE9;hAPOE4 Mouse Model of Alzheimer's Disease. Int J Mol Sci 2024; 25:5754. [PMID: 38891941 PMCID: PMC11172317 DOI: 10.3390/ijms25115754] [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: 05/04/2024] [Revised: 05/16/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
Alzheimer's disease (AD) is a devastating disorder with a global prevalence estimated at 55 million people. In clinical studies administering certain anti-beta-amyloid (Aβ) antibodies, amyloid-related imaging abnormalities (ARIAs) have emerged as major adverse events. The frequency of these events is higher among apolipoprotein ε4 allele carriers (APOE4) compared to non-carriers. To reflect patients most at risk for vascular complications of anti-Aβ immunotherapy, we selected an APPswe/PS1dE9 transgenic mouse model bearing the human APOE4 gene (APPPS1:E4) and compared it with the same APP/PS1 mouse model bearing the human APOE3 gene (APOE ε3 allele; APPPS1:E3). Using histological and biochemical analyses, we characterized mice at three ages: 8, 12, and 16 months. Female and male mice were assayed for general cerebral fibrillar and pyroglutamate (pGlu-3) Aβ deposition, cerebral amyloid angiopathy (CAA), microhemorrhages, apoE and cholesterol composition, astrocytes, microglia, inflammation, lysosomal dysfunction, and neuritic dystrophy. Amyloidosis, lipid deposition, and astrogliosis increased with age in APPPS1:E4 mice, while inflammation did not reveal significant changes with age. In general, APOE4 carriers showed elevated Aβ, apoE, reactive astrocytes, pro-inflammatory cytokines, microglial response, and neuritic dystrophy compared to APOE3 carriers at different ages. These results highlight the potential of the APPPS1:E4 mouse model as a valuable tool in investigating the vascular side effects associated with anti-amyloid immunotherapy.
Collapse
Affiliation(s)
- Martine B. Grenon
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (M.B.G.); (M.-T.P.); (P.B.)
- Section Neuropsychology & Psychopharmacology, Faculty of Psychology and Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Maria-Tzousi Papavergi
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (M.B.G.); (M.-T.P.); (P.B.)
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, 6200 MD Maastricht, The Netherlands
| | - Praveen Bathini
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (M.B.G.); (M.-T.P.); (P.B.)
| | - Martin Sadowski
- Departments of Neurology, Psychiatry, and Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY 10016, USA;
| | - Cynthia A. Lemere
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (M.B.G.); (M.-T.P.); (P.B.)
| |
Collapse
|
11
|
Jackson RJ, Keiser MS, Meltzer JC, Fykstra DP, Dierksmeier SE, Hajizadeh S, Kreuzer J, Morris R, Melloni A, Nakajima T, Tecedor L, Ranum PT, Carrell E, Chen Y, Nishtar MA, Holtzman DM, Haas W, Davidson BL, Hyman BT. APOE2 gene therapy reduces amyloid deposition and improves markers of neuroinflammation and neurodegeneration in a mouse model of Alzheimer disease. Mol Ther 2024; 32:1373-1386. [PMID: 38504517 PMCID: PMC11081918 DOI: 10.1016/j.ymthe.2024.03.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 01/05/2024] [Accepted: 03/15/2024] [Indexed: 03/21/2024] Open
Abstract
Epidemiological studies show that individuals who carry the relatively uncommon APOE ε2 allele rarely develop Alzheimer disease, and if they do, they have a later age of onset, milder clinical course, and less severe neuropathological findings than people without this allele. The contrast is especially stark when compared with the major genetic risk factor for Alzheimer disease, APOE ε4, which has an age of onset several decades earlier, a more aggressive clinical course and more severe neuropathological findings, especially in terms of the amount of amyloid deposition. Here, we demonstrate that brain exposure to APOE ε2 via a gene therapy approach, which bathes the entire cortical mantle in the gene product after transduction of the ependyma, reduces Aβ plaque deposition, neurodegenerative synaptic loss, and, remarkably, reduces microglial activation in an APP/PS1 mouse model despite continued expression of human APOE ε4. This result suggests a promising protective effect of exogenous APOE ε2 and reveals a cell nonautonomous effect of the protein on microglial activation, which we show is similar to plaque-associated microglia in the brain of Alzheimer disease patients who inherit APOE ε2. These data increase the potential that an APOE ε2 therapeutic could be effective in Alzheimer disease, even in individuals born with the risky ε4 allele.
Collapse
Affiliation(s)
- Rosemary J Jackson
- Alzheimer Research Unit, Massachusetts General Hospital Institute for Neurodegenerative Disease, Charlestown, MA 02129, USA; Department of Neurology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Boston, MA 02114, USA.
| | - Megan S Keiser
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Jonah C Meltzer
- Alzheimer Research Unit, Massachusetts General Hospital Institute for Neurodegenerative Disease, Charlestown, MA 02129, USA; Department of Neurology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Boston, MA 02114, USA
| | - Dustin P Fykstra
- Alzheimer Research Unit, Massachusetts General Hospital Institute for Neurodegenerative Disease, Charlestown, MA 02129, USA; Department of Neurology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Boston, MA 02114, USA
| | - Steven E Dierksmeier
- Alzheimer Research Unit, Massachusetts General Hospital Institute for Neurodegenerative Disease, Charlestown, MA 02129, USA; Department of Neurology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Boston, MA 02114, USA; Medical Sciences Division, University of Oxford, Oxford OX3 9DU, UK
| | - Soroush Hajizadeh
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, UK; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - Johannes Kreuzer
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, UK; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Robert Morris
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, UK
| | - Alexandra Melloni
- Alzheimer Research Unit, Massachusetts General Hospital Institute for Neurodegenerative Disease, Charlestown, MA 02129, USA
| | - Tsuneo Nakajima
- Alzheimer Research Unit, Massachusetts General Hospital Institute for Neurodegenerative Disease, Charlestown, MA 02129, USA; Department of Neurology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Boston, MA 02114, USA
| | - Luis Tecedor
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Paul T Ranum
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Ellie Carrell
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - YongHong Chen
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Maryam A Nishtar
- Alzheimer Research Unit, Massachusetts General Hospital Institute for Neurodegenerative Disease, Charlestown, MA 02129, USA; Department of Neurology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Boston, MA 02114, USA
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - Wilhelm Haas
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, UK; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Beverly L Davidson
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bradley T Hyman
- Alzheimer Research Unit, Massachusetts General Hospital Institute for Neurodegenerative Disease, Charlestown, MA 02129, USA; Department of Neurology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
12
|
Strickland MR, Rau MJ, Summers B, Basore K, Wulf J, Jiang H, Chen Y, Ulrich JD, Randolph GJ, Zhang R, Fitzpatrick JAJ, Cashikar AG, Holtzman DM. Apolipoprotein E secreted by astrocytes forms antiparallel dimers in discoidal lipoproteins. Neuron 2024; 112:1100-1109.e5. [PMID: 38266643 PMCID: PMC10994765 DOI: 10.1016/j.neuron.2023.12.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 11/24/2023] [Accepted: 12/22/2023] [Indexed: 01/26/2024]
Abstract
The Apolipoprotein E gene (APOE) is of great interest due to its role as a risk factor for late-onset Alzheimer's disease. ApoE is secreted by astrocytes in the central nervous system in high-density lipoprotein (HDL)-like lipoproteins. Structural models of lipidated ApoE of high resolution could aid in a mechanistic understanding of how ApoE functions in health and disease. Using monoclonal Fab and F(ab')2 fragments, we characterize the structure of lipidated ApoE on astrocyte-secreted lipoproteins. Our results provide support for the "double-belt" model of ApoE in nascent discoidal HDL-like lipoproteins, where two ApoE proteins wrap around the nanodisc in an antiparallel conformation. We further show that lipidated, recombinant ApoE accurately models astrocyte-secreted ApoE lipoproteins. Cryogenic electron microscopy of recombinant lipidated ApoE further supports ApoE adopting antiparallel dimers in nascent discoidal lipoproteins.
Collapse
Affiliation(s)
| | - Michael J Rau
- Washington University Center for Cellular Imaging, 660 S. Euclid Ave., St. Louis, MO 63110, USA
| | - Brock Summers
- Washington University Center for Cellular Imaging, 660 S. Euclid Ave., St. Louis, MO 63110, USA
| | - Katherine Basore
- Washington University Center for Cellular Imaging, 660 S. Euclid Ave., St. Louis, MO 63110, USA
| | - John Wulf
- Washington University Center for Cellular Imaging, 660 S. Euclid Ave., St. Louis, MO 63110, USA
| | - Hong Jiang
- Department of Neurology, 660 S. Euclid Ave., St. Louis, MO 63110, USA
| | - Yun Chen
- Department of Neurology, 660 S. Euclid Ave., St. Louis, MO 63110, USA; Department of Pathology and Immunology, 660 S. Euclid Ave., St. Louis, MO 63110, USA
| | - Jason D Ulrich
- Department of Neurology, 660 S. Euclid Ave., St. Louis, MO 63110, USA; Hope Center for Neurological Disorders, 660 S. Euclid Ave., St. Louis, MO 63110, USA; Knight Alzheimer's Disease Research Center, 4488 Forest Park Ave., St. Louis, MO 63108, USA
| | - Gwendalyn J Randolph
- Department of Pathology and Immunology, 660 S. Euclid Ave., St. Louis, MO 63110, USA
| | - Rui Zhang
- Department of Biochemistry and Molecular Biophysics, 660 S. Euclid Ave., St. Louis, MO 63110, USA
| | - James A J Fitzpatrick
- Washington University Center for Cellular Imaging, 660 S. Euclid Ave., St. Louis, MO 63110, USA
| | - Anil G Cashikar
- Hope Center for Neurological Disorders, 660 S. Euclid Ave., St. Louis, MO 63110, USA; Department of Psychiatry, 660 S. Euclid Ave., St. Louis, MO 63110, USA; Taylor Family institute for Innovative Psychiatric Research, 660 S. Euclid Ave., St. Louis, MO 63110, USA
| | - David M Holtzman
- Department of Neurology, 660 S. Euclid Ave., St. Louis, MO 63110, USA; Hope Center for Neurological Disorders, 660 S. Euclid Ave., St. Louis, MO 63110, USA; Knight Alzheimer's Disease Research Center, 4488 Forest Park Ave., St. Louis, MO 63108, USA.
| |
Collapse
|
13
|
Ohgita T, Sakai K, Fukui N, Namba N, Nakano M, Kiguchi Y, Morita I, Oyama H, Yamaki K, Nagao K, Kobayashi N, Saito H. Generation of novel anti-apoE monoclonal antibodies that selectively recognize apoE isoforms. FEBS Lett 2024; 598:902-914. [PMID: 38529702 DOI: 10.1002/1873-3468.14858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 12/21/2023] [Accepted: 12/22/2023] [Indexed: 03/27/2024]
Abstract
Apolipoprotein E (apoE) is a regulator of lipid metabolism, cholesterol transport, and the clearance and aggregation of amyloid β in the brain. The three human apoE isoforms apoE2, apoE3, and apoE4 only differ in one or two residues. Nevertheless, the functions highly depend on the isoform types and lipidated states. Here, we generated novel anti-apoE monoclonal antibodies (mAbs) and obtained an apoE4-selective mAb whose epitope is within residues 110-117. ELISA and bio-layer interferometry measurements demonstrated that the dissociation constants of mAbs are within the nanomolar range. Using the generated antibodies, we successfully constructed sandwich ELISA systems, which can detect all apoE isoforms or selectively detect apoE4. These results suggest the usability of the generated anti-apoE mAbs for selective detection of apoE isoforms.
Collapse
Affiliation(s)
- Takashi Ohgita
- Laboratory of Biophysical Chemistry, Kyoto Pharmaceutical University, Japan
- Center for Instrumental Analysis, Kyoto Pharmaceutical University, Japan
| | - Koto Sakai
- Laboratory of Biophysical Chemistry, Kyoto Pharmaceutical University, Japan
| | - Nodoka Fukui
- Laboratory of Biophysical Chemistry, Kyoto Pharmaceutical University, Japan
| | - Norihiro Namba
- Laboratory of Biophysical Chemistry, Kyoto Pharmaceutical University, Japan
| | - Miyu Nakano
- Laboratory of Biophysical Chemistry, Kyoto Pharmaceutical University, Japan
| | - Yuki Kiguchi
- Laboratory of Bioanalytical Chemistry, Kobe Pharmaceutical University, Japan
| | - Izumi Morita
- Laboratory of Bioanalytical Chemistry, Kobe Pharmaceutical University, Japan
| | - Hiroyuki Oyama
- Laboratory of Bioanalytical Chemistry, Kobe Pharmaceutical University, Japan
| | - Kouya Yamaki
- Laboratory of Pharmacology, Kobe Pharmaceutical University, Japan
| | - Kohjiro Nagao
- Laboratory of Biophysical Chemistry, Kyoto Pharmaceutical University, Japan
| | - Norihiro Kobayashi
- Laboratory of Bioanalytical Chemistry, Kobe Pharmaceutical University, Japan
| | - Hiroyuki Saito
- Laboratory of Biophysical Chemistry, Kyoto Pharmaceutical University, Japan
| |
Collapse
|
14
|
Ferguson CM, Hildebrand S, Godinho BMDC, Buchwald J, Echeverria D, Coles A, Grigorenko A, Vangjeli L, Sousa J, McHugh N, Hassler M, Santarelli F, Heneka MT, Rogaev E, Khvorova A. Silencing Apoe with divalent-siRNAs improves amyloid burden and activates immune response pathways in Alzheimer's disease. Alzheimers Dement 2024; 20:2632-2652. [PMID: 38375983 PMCID: PMC11032532 DOI: 10.1002/alz.13703] [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: 05/12/2023] [Revised: 10/30/2023] [Accepted: 11/14/2023] [Indexed: 02/21/2024]
Abstract
INTRODUCTION The most significant genetic risk factor for late-onset Alzheimer's disease (AD) is APOE4, with evidence for gain- and loss-of-function mechanisms. A clinical need remains for therapeutically relevant tools that potently modulate APOE expression. METHODS We optimized small interfering RNAs (di-siRNA, GalNAc) to potently silence brain or liver Apoe and evaluated the impact of each pool of Apoe on pathology. RESULTS In adult 5xFAD mice, siRNAs targeting CNS Apoe efficiently silenced Apoe expression and reduced amyloid burden without affecting systemic cholesterol, confirming that potent silencing of brain Apoe is sufficient to slow disease progression. Mechanistically, silencing Apoe reduced APOE-rich amyloid cores and activated immune system responses. DISCUSSION These results establish siRNA-based modulation of Apoe as a viable therapeutic approach, highlight immune activation as a key pathway affected by Apoe modulation, and provide the technology to further evaluate the impact of APOE silencing on neurodegeneration.
Collapse
Affiliation(s)
- Chantal M. Ferguson
- RNA Therapeutics Institute, University of Massachusetts Medical SchoolWorcesterMassachusettsUSA
| | - Samuel Hildebrand
- RNA Therapeutics Institute, University of Massachusetts Medical SchoolWorcesterMassachusettsUSA
| | - Bruno M. D. C. Godinho
- RNA Therapeutics Institute, University of Massachusetts Medical SchoolWorcesterMassachusettsUSA
| | - Julianna Buchwald
- RNA Therapeutics Institute, University of Massachusetts Medical SchoolWorcesterMassachusettsUSA
| | - Dimas Echeverria
- RNA Therapeutics Institute, University of Massachusetts Medical SchoolWorcesterMassachusettsUSA
| | - Andrew Coles
- RNA Therapeutics Institute, University of Massachusetts Medical SchoolWorcesterMassachusettsUSA
| | - Anastasia Grigorenko
- Department of PsychiatryUniversity of Massachusetts Medical SchoolWorcesterMassachusettsUSA
| | - Lorenc Vangjeli
- RNA Therapeutics Institute, University of Massachusetts Medical SchoolWorcesterMassachusettsUSA
| | - Jacquelyn Sousa
- RNA Therapeutics Institute, University of Massachusetts Medical SchoolWorcesterMassachusettsUSA
| | - Nicholas McHugh
- RNA Therapeutics Institute, University of Massachusetts Medical SchoolWorcesterMassachusettsUSA
| | - Matthew Hassler
- RNA Therapeutics Institute, University of Massachusetts Medical SchoolWorcesterMassachusettsUSA
| | | | - Michael T. Heneka
- Luxembourg Centre for Systems Biomedicine (LCSB)Esch‐sur‐AlzetteLuxembourg
| | - Evgeny Rogaev
- Department of PsychiatryUniversity of Massachusetts Medical SchoolWorcesterMassachusettsUSA
| | - Anastasia Khvorova
- RNA Therapeutics Institute, University of Massachusetts Medical SchoolWorcesterMassachusettsUSA
| |
Collapse
|
15
|
de Leeuw SM, Nuriel T. Intracellular cholesterol visualization in brain tissue using D4H ∗. STAR Protoc 2024; 5:102779. [PMID: 38100357 PMCID: PMC10762518 DOI: 10.1016/j.xpro.2023.102779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 10/31/2023] [Accepted: 11/30/2023] [Indexed: 12/17/2023] Open
Abstract
Studying cholesterol biology in the brain has been greatly hindered by the lack of adequate cholesterol visualization techniques. Here, we present a protocol for using a high-affinity cholesterol probe D4H∗-mCherry as a histology reagent in mouse or human brain tissue. We describe steps for D4H∗ tissue treatment and crosslinking leading to stable labeling of intracellular membrane cholesterol. Furthermore, co-labeling with Rab5 endosomal marker and optimized buffers to reduce background enable punctate cholesterol visualization within the organelle membranes.
Collapse
Affiliation(s)
- Sherida M de Leeuw
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, 630 West 168th Street, New York, NY 10032, USA; Department of Pathology and Cell Biology, Columbia University, 630 West 168th Street, New York, NY 10032, USA.
| | - Tal Nuriel
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, 630 West 168th Street, New York, NY 10032, USA; Department of Pathology and Cell Biology, Columbia University, 630 West 168th Street, New York, NY 10032, USA.
| |
Collapse
|
16
|
Sepulveda J, Kim JY, Binder J, Vicini S, Rebeck GW. APOE4 genotype and aging impair injury-induced microglial behavior in brain slices, including toward Aβ, through P2RY12. Mol Neurodegener 2024; 19:24. [PMID: 38468308 DOI: 10.1186/s13024-024-00714-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 02/22/2024] [Indexed: 03/13/2024] Open
Abstract
Microglia are highly dynamic cells that play a critical role in tissue homeostasis through the surveillance of brain parenchyma and response to cues associated with damage. Aging and APOE4 genotype are the strongest risk factors for Alzheimer's disease (AD), but how they affect microglial dynamics remains unclear. Using ex vivo confocal microscopy, we analyzed microglial dynamic behaviors in the entorhinal cortex (EC) and hippocampus CA1 of 6-, 12-, and 21-month-old mice APOE3 or APOE4 knock-in mice expressing GFP under the CX3CR1 promoter. To study microglia surveillance, we imaged microglia baseline motility for 20 min and measured the extension and retraction of processes. We found that APOE4 microglia exhibited significantly less brain surveillance (27%) compared to APOE3 microglia in 6-month-old mice; aging exacerbated this deficit. To measure microglia response to damage, we imaged process motility in response to ATP, an injury-associated signal, for 30 min. We found APOE4 microglia extended their processes significantly slower (0.9 µm/min, p < 0.005) than APOE3 microglia (1.1 μm/min) in 6-month-old animals. APOE-associated alterations in microglia motility were observed in 12- and 21-month-old animals, and this effect was exacerbated with aging in APOE4 microglia. We measured protein and mRNA levels of P2RY12, a core microglial receptor required for process movement in response to damage. We found that APOE4 microglia express significantly less P2RY12 receptors compared to APOE3 microglia despite no changes in P2RY12 transcripts. To examine if the effect of APOE4 on the microglial response to ATP also applied to amyloid β (Aβ), we infused locally Hi-Lyte Fluor 555-labeled Aβ in acute brain slices of 6-month-old mice and imaged microglia movement for 2 h. APOE4 microglia showed a significantly slower (p < 0.0001) process movement toward the Aβ, and less Aβ coverage at early time points after Aβ injection. To test whether P2RY12 is involved in process movement in response to Aβ, we treated acute brain slices with a P2RY12 antagonist before Aβ injection; microglial processes no longer migrated towards Aβ. These results provide mechanistic insights into the impact of APOE4 genotype and aging in dynamic microglial behaviors prior to gross Aβ pathology and could help explain how APOE4 brains are more susceptible to AD pathogenesis.
Collapse
Affiliation(s)
- Jordy Sepulveda
- Department of Pharmacology & Physiology, Georgetown University, Washington, DC, 20007, USA
| | - Jennifer Yejean Kim
- Department of Neuroscience, Georgetown University, Washington, DC, 20007, USA
| | - Joseph Binder
- Department of Neuroscience, Georgetown University, Washington, DC, 20007, USA
| | - Stefano Vicini
- Department of Pharmacology & Physiology, Georgetown University, Washington, DC, 20007, USA
| | - G William Rebeck
- Department of Neuroscience, Georgetown University, Washington, DC, 20007, USA.
| |
Collapse
|
17
|
Zhong MZ, Peng T, Duarte ML, Wang M, Cai D. Updates on mouse models of Alzheimer's disease. Mol Neurodegener 2024; 19:23. [PMID: 38462606 PMCID: PMC10926682 DOI: 10.1186/s13024-024-00712-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 02/14/2024] [Indexed: 03/12/2024] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease in the United States (US). Animal models, specifically mouse models have been developed to better elucidate disease mechanisms and test therapeutic strategies for AD. A large portion of effort in the field was focused on developing transgenic (Tg) mouse models through over-expression of genetic mutations associated with familial AD (FAD) patients. Newer generations of mouse models through knock-in (KI)/knock-out (KO) or CRISPR gene editing technologies, have been developed for both familial and sporadic AD risk genes with the hope to more accurately model proteinopathies without over-expression of human AD genes in mouse brains. In this review, we summarized the phenotypes of a few commonly used as well as newly developed mouse models in translational research laboratories including the presence or absence of key pathological features of AD such as amyloid and tau pathology, synaptic and neuronal degeneration as well as cognitive and behavior deficits. In addition, advantages and limitations of these AD mouse models have been elaborated along with discussions of any sex-specific features. More importantly, the omics data from available AD mouse models have been analyzed to categorize molecular signatures of each model reminiscent of human AD brain changes, with the hope to guide future selection of most suitable models for specific research questions to be addressed in the AD field.
Collapse
Affiliation(s)
- Michael Z Zhong
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Biology, College of Arts and Science, Boston University, Boston, MA, 02215, USA
| | - Thomas Peng
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Science Research Program, Scarsdale High School, New York, NY, 10583, USA
| | - Mariana Lemos Duarte
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Research & Development, James J Peters VA Medical Center, Bronx, NY, 10468, USA.
| | - Minghui Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA.
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA.
| | - Dongming Cai
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Research & Development, James J Peters VA Medical Center, Bronx, NY, 10468, USA.
- Alzheimer's Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Neurology, N. Bud Grossman Center for Memory Research and Care, The University of Minnesota, Minneapolis, MN, 55455, USA.
- Geriatric Research Education & Clinical Center (GRECC), The Minneapolis VA Health Care System, Minneapolis, MN, 55417, USA.
| |
Collapse
|
18
|
Litvinchuk A, Suh JH, Guo JL, Lin K, Davis SS, Bien-Ly N, Tycksen E, Tabor GT, Remolina Serrano J, Manis M, Bao X, Lee C, Bosch M, Perez EJ, Yuede CM, Cashikar AG, Ulrich JD, Di Paolo G, Holtzman DM. Amelioration of Tau and ApoE4-linked glial lipid accumulation and neurodegeneration with an LXR agonist. Neuron 2024; 112:384-403.e8. [PMID: 37995685 PMCID: PMC10922706 DOI: 10.1016/j.neuron.2023.10.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 09/13/2023] [Accepted: 10/18/2023] [Indexed: 11/25/2023]
Abstract
Apolipoprotein E (APOE) is a strong genetic risk factor for late-onset Alzheimer's disease (LOAD). APOE4 increases and APOE2 decreases risk relative to APOE3. In the P301S mouse model of tauopathy, ApoE4 increases tau pathology and neurodegeneration when compared with ApoE3 or the absence of ApoE. However, the role of ApoE isoforms and lipid metabolism in contributing to tau-mediated degeneration is unknown. We demonstrate that in P301S tau mice, ApoE4 strongly promotes glial lipid accumulation and perturbations in cholesterol metabolism and lysosomal function. Increasing lipid efflux in glia via an LXR agonist or Abca1 overexpression strongly attenuates tau pathology and neurodegeneration in P301S/ApoE4 mice. We also demonstrate reductions in reactive astrocytes and microglia, as well as changes in cholesterol biosynthesis and metabolism in glia of tauopathy mice in response to LXR activation. These data suggest that promoting efflux of glial lipids may serve as a therapeutic approach to ameliorate tau and ApoE4-linked neurodegeneration.
Collapse
Affiliation(s)
- Alexandra Litvinchuk
- Department 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 63108, USA
| | - Jung H Suh
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA
| | - Jing L Guo
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA
| | - Karin Lin
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA
| | - Sonnet S Davis
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA
| | - Nga Bien-Ly
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA
| | - Eric Tycksen
- Genome Technology Access Center, McDonnell Genome Institute, St. Louis, MO 63110, USA
| | - G Travis Tabor
- Department 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 63108, USA
| | - Javier Remolina Serrano
- Department 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 63108, USA
| | - Melissa Manis
- Department 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 63108, USA
| | - Xin Bao
- Department 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 63108, USA
| | - Choonghee Lee
- Department 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 63108, USA
| | - Megan Bosch
- Department 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 63108, USA
| | - Enmanuel J Perez
- Department 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 63108, USA
| | - Carla M Yuede
- Department 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 63108, USA
| | - Anil G Cashikar
- Department of Psychiatry, Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Jason D Ulrich
- Department 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 63108, USA
| | | | - David M Holtzman
- Department 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 63108, USA.
| |
Collapse
|
19
|
Blumenfeld J, Yip O, Kim MJ, Huang Y. Cell type-specific roles of APOE4 in Alzheimer disease. Nat Rev Neurosci 2024; 25:91-110. [PMID: 38191720 PMCID: PMC11073858 DOI: 10.1038/s41583-023-00776-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/28/2023] [Indexed: 01/10/2024]
Abstract
The ɛ4 allele of the apolipoprotein E gene (APOE), which translates to the APOE4 isoform, is the strongest genetic risk factor for late-onset Alzheimer disease (AD). Within the CNS, APOE is produced by a variety of cell types under different conditions, posing a challenge for studying its roles in AD pathogenesis. However, through powerful advances in research tools and the use of novel cell culture and animal models, researchers have recently begun to study the roles of APOE4 in AD in a cell type-specific manner and at a deeper and more mechanistic level than ever before. In particular, cutting-edge omics studies have enabled APOE4 to be studied at the single-cell level and have allowed the identification of critical APOE4 effects in AD-vulnerable cellular subtypes. Through these studies, it has become evident that APOE4 produced in various types of CNS cell - including astrocytes, neurons, microglia, oligodendrocytes and vascular cells - has diverse roles in AD pathogenesis. Here, we review these scientific advances and propose a cell type-specific APOE4 cascade model of AD. In this model, neuronal APOE4 emerges as a crucial pathological initiator and driver of AD pathogenesis, instigating glial responses and, ultimately, neurodegeneration. In addition, we provide perspectives on future directions for APOE4 research and related therapeutic developments in the context of AD.
Collapse
Affiliation(s)
- Jessica Blumenfeld
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Oscar Yip
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Min Joo Kim
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Yadong Huang
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA.
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA.
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA.
- Gladstone Center for Translational Advancement, Gladstone Institutes, San Francisco, CA, USA.
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA.
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
20
|
Chen Y, Song S, Parhizkar S, Lord J, Zhu Y, Strickland MR, Wang C, Park J, Travis Tabor G, Jiang H, Li K, Davis AA, Yuede CM, Colonna M, Ulrich JD, Holtzman DM. APOE3ch alters microglial response and suppresses Aβ-induced tau seeding and spread. Cell 2024; 187:428-445.e20. [PMID: 38086389 PMCID: PMC10842861 DOI: 10.1016/j.cell.2023.11.029] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 10/24/2023] [Accepted: 11/27/2023] [Indexed: 12/20/2023]
Abstract
A recent case report described an individual who was a homozygous carrier of the APOE3 Christchurch (APOE3ch) mutation and resistant to autosomal dominant Alzheimer's Disease (AD) caused by a PSEN1-E280A mutation. Whether APOE3ch contributed to the protective effect remains unclear. We generated a humanized APOE3ch knock-in mouse and crossed it to an amyloid-β (Aβ) plaque-depositing model. We injected AD-tau brain extract to investigate tau seeding and spreading in the presence or absence of amyloid. Similar to the case report, APOE3ch expression resulted in peripheral dyslipidemia and a marked reduction in plaque-associated tau pathology. Additionally, we observed decreased amyloid response and enhanced microglial response around plaques. We also demonstrate increased myeloid cell phagocytosis and degradation of tau aggregates linked to weaker APOE3ch binding to heparin sulfate proteoglycans. APOE3ch influences the microglial response to Aβ plaques, which suppresses Aβ-induced tau seeding and spreading. The results reveal new possibilities to target Aβ-induced tauopathy.
Collapse
Affiliation(s)
- Yun Chen
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sihui Song
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Samira Parhizkar
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA
- Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jennifer Lord
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yiyang Zhu
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Michael R. Strickland
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Chanung Wang
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jiyu Park
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - G. Travis Tabor
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Hong Jiang
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA
- Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Kevin Li
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Albert A. Davis
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Carla M. Yuede
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA
- Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jason D. Ulrich
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA
- Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - David M. Holtzman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA
- Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
21
|
Valencia-Olvera AC, Balu D, Moore A, Shah M, Ainis R, Xiang B, Saleh Y, Cai D, LaDu MJ, Tai LM. APOE2 Heterozygosity Reduces Hippocampal Soluble Amyloid-β42 Levels in Non-Hyperlipidemic Mice. J Alzheimers Dis 2024; 97:1629-1639. [PMID: 38306049 DOI: 10.3233/jad-231210] [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: 02/03/2024]
Abstract
APOE2 lowers Alzheimer's disease (AD) risk; unfortunately, the mechanism remains poorly understood and the use of mice models is problematic as APOE2 homozygosity is associated with hyperlipidemia. In this study, we developed mice that are heterozygous for APOE2 and APOE3 or APOE4 and overexpress amyloid-β peptide (Aβ) (EFAD) to evaluate the effect of APOE2 dosage on Aβ pathology. We found that heterozygous mice do not exhibit hyperlipidemia. Hippocampal but not cortical levels of soluble Aβ42 followed the order E2/2FAD > E2/3FAD≤E3/3FAD and E2/2FAD > E2/4FAD < E4/4FAD without an effect on insoluble Aβ42. These findings offer initial insights on the impact of APOE2 on Aβ pathology.
Collapse
Affiliation(s)
- Ana C Valencia-Olvera
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA
| | - Deebika Balu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA
| | | | - Maitri Shah
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA
| | - Rebecca Ainis
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA
| | | | - Yaseen Saleh
- University of Miami/Jackson Healthcare System, Miami, FL, USA
| | - Dongming Cai
- Alzheimer Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Research and Development Service, James J. Peters VA Medical Center, Bronx, NY, USA
- Department of Neurology, N. Bud Grossman Center for Memory Research and Care, University of Minnesota, Minneapolis, MN, USA
- Geriatric Research Education and Clinical Center (GRECC), Minneapolis VA Health Care System, Minneapolis, MN, USA
| | - Mary Jo LaDu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA
| | - Leon M Tai
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
22
|
Liemisa B, Newbury SF, Novy MJ, Pasato JA, Morales-Corraliza J, Peng KY, Mathews PM. Brain apolipoprotein E levels in mice challenged by a Western diet increase in an allele-dependent manner. AGING BRAIN 2023; 4:100102. [PMID: 38058491 PMCID: PMC10696459 DOI: 10.1016/j.nbas.2023.100102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 11/05/2023] [Accepted: 11/17/2023] [Indexed: 12/08/2023] Open
Abstract
Human apolipoprotein E (APOE) is the greatest determinant of genetic risk for memory deficits and Alzheimer's disease (AD). While APOE4 drives memory loss and high AD risk, APOE2 leads to healthy brain aging and reduced AD risk compared to the common APOE3 variant. We examined brain APOE protein levels in humanized mice homozygous for these alleles and found baseline levels to be age- and isoform-dependent: APOE2 levels were greater than APOE3, which were greater than APOE4. Despite the understanding that APOE lipoparticles do not traverse the blood-brain barrier, we show that brain APOE levels are responsive to dietary fat intake. Challenging mice for 6 months on a Western diet high in fat and cholesterol increased APOE protein levels in an allele-dependent fashion with a much greater increase within blood plasma than within the brain. In the brain, APOE2 levels responded most to the Western diet challenge, increasing by 20 % to 30 %. While increased lipoparticles are generally deleterious in the periphery, we propose that higher brain APOE2 levels may represent a readily available pool of beneficial lipid particles for neurons.
Collapse
Affiliation(s)
- Braison Liemisa
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY 10962, USA
| | - Samantha F. Newbury
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY 10962, USA
| | - Mariah J. Novy
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY 10962, USA
| | - Jonathan A. Pasato
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY 10962, USA
| | - Jose Morales-Corraliza
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY 10962, USA
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Katherine Y. Peng
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY 10962, USA
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Paul M. Mathews
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY 10962, USA
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY 10016, USA
- NYU Neuroscience Institute, New York University Grossman School of Medicine, New York, NY 10016, USA
| |
Collapse
|
23
|
Yin Z, Rosenzweig N, Kleemann KL, Zhang X, Brandão W, Margeta MA, Schroeder C, Sivanathan KN, Silveira S, Gauthier C, Mallah D, Pitts KM, Durao A, Herron S, Shorey H, Cheng Y, Barry JL, Krishnan RK, Wakelin S, Rhee J, Yung A, Aronchik M, Wang C, Jain N, Bao X, Gerrits E, Brouwer N, Deik A, Tenen DG, Ikezu T, Santander NG, McKinsey GL, Baufeld C, Sheppard D, Krasemann S, Nowarski R, Eggen BJL, Clish C, Tanzi RE, Madore C, Arnold TD, Holtzman DM, Butovsky O. APOE4 impairs the microglial response in Alzheimer's disease by inducing TGFβ-mediated checkpoints. Nat Immunol 2023; 24:1839-1853. [PMID: 37749326 PMCID: PMC10863749 DOI: 10.1038/s41590-023-01627-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 08/15/2023] [Indexed: 09/27/2023]
Abstract
The APOE4 allele is the strongest genetic risk factor for late-onset Alzheimer's disease (AD). The contribution of microglial APOE4 to AD pathogenesis is unknown, although APOE has the most enriched gene expression in neurodegenerative microglia (MGnD). Here, we show in mice and humans a negative role of microglial APOE4 in the induction of the MGnD response to neurodegeneration. Deletion of microglial APOE4 restores the MGnD phenotype associated with neuroprotection in P301S tau transgenic mice and decreases pathology in APP/PS1 mice. MGnD-astrocyte cross-talk associated with β-amyloid (Aβ) plaque encapsulation and clearance are mediated via LGALS3 signaling following microglial APOE4 deletion. In the brains of AD donors carrying the APOE4 allele, we found a sex-dependent reciprocal induction of AD risk factors associated with suppression of MGnD genes in females, including LGALS3, compared to individuals homozygous for the APOE3 allele. Mechanistically, APOE4-mediated induction of ITGB8-transforming growth factor-β (TGFβ) signaling impairs the MGnD response via upregulation of microglial homeostatic checkpoints, including Inpp5d, in mice. Deletion of Inpp5d in microglia restores MGnD-astrocyte cross-talk and facilitates plaque clearance in APP/PS1 mice. We identify the microglial APOE4-ITGB8-TGFβ pathway as a negative regulator of microglial response to AD pathology, and restoring the MGnD phenotype via blocking ITGB8-TGFβ signaling provides a promising therapeutic intervention for AD.
Collapse
Affiliation(s)
- Zhuoran Yin
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Neta Rosenzweig
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kilian L Kleemann
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- School of Computing, University of Portsmouth, Portsmouth, UK
| | - Xiaoming Zhang
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Wesley Brandão
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Milica A Margeta
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Caitlin Schroeder
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kisha N Sivanathan
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Evergrande Center for Immunologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sebastian Silveira
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Christian Gauthier
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Dania Mallah
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kristen M Pitts
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Ana Durao
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Shawn Herron
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Hannah Shorey
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yiran Cheng
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jen-Li Barry
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Rajesh K Krishnan
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Evergrande Center for Immunologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sam Wakelin
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jared Rhee
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Anthony Yung
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael Aronchik
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Chao Wang
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
- Institute for Brain Science and Disease, Chongqing Medical University, Chongqing, China
| | - Nimansha Jain
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Xin Bao
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Emma Gerrits
- Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Nieske Brouwer
- Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Amy Deik
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Daniel G Tenen
- Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA
- Cancer Science Institute, National University of Singapore, Singapore, Singapore
| | - Tsuneya Ikezu
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL, USA
| | - Nicolas G Santander
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
- Instituto de Ciencias de la Salud, Universidad de O´Higgins, Rancagua, Chile
| | - Gabriel L McKinsey
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Caroline Baufeld
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Dean Sheppard
- Department of Medicine, Cardiovascular Research Center, University of California, San Francisco, San Francisco, CA, USA
| | - Susanne Krasemann
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf UKE, Hamburg, Germany
| | - Roni Nowarski
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Evergrande Center for Immunologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Bart J L Eggen
- Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Clary Clish
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Rudolph E Tanzi
- Genetics and Aging Research Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Charlotte Madore
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Laboratoire NutriNeuro, UMR1286, INRAE, Bordeaux INP, University of Bordeaux, Bordeaux, France
| | - Thomas D Arnold
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Oleg Butovsky
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Evergrande Center for Immunologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
24
|
Liu CC, Wang N, Chen Y, Inoue Y, Shue F, Ren Y, Wang M, Qiao W, Ikezu TC, Li Z, Zhao J, Martens Y, Doss SV, Rosenberg CL, Jeevaratnam S, Jia L, Raulin AC, Qi F, Zhu Y, Alnobani A, Knight J, Chen Y, Linares C, Kurti A, Fryer JD, Zhang B, Wu LJ, Kim BYS, Bu G. Cell-autonomous effects of APOE4 in restricting microglial response in brain homeostasis and Alzheimer's disease. Nat Immunol 2023; 24:1854-1866. [PMID: 37857825 DOI: 10.1038/s41590-023-01640-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 09/06/2023] [Indexed: 10/21/2023]
Abstract
Microglial involvement in Alzheimer's disease (AD) pathology has emerged as a risk-determining pathogenic event. While apolipoprotein E (APOE) is known to modify AD risk, it remains unclear how microglial apoE impacts brain cognition and AD pathology. Here, using conditional mouse models expressing apoE isoforms in microglia and central nervous system-associated macrophages (CAMs), we demonstrate a cell-autonomous effect of apoE3-mediated microglial activation and function, which are negated by apoE4. Expression of apoE3 in microglia/CAMs improves cognitive function, increases microglia surrounding amyloid plaque and reduces amyloid pathology and associated toxicity, whereas apoE4 expression either compromises or has no effects on these outcomes by impairing lipid metabolism. Single-cell transcriptomic profiling reveals increased antigen presentation and interferon pathways upon apoE3 expression. In contrast, apoE4 expression downregulates complement and lysosomal pathways, and promotes stress-related responses. Moreover, in the presence of mouse endogenous apoE, microglial apoE4 exacerbates amyloid pathology. Finally, we observed a reduction in Lgals3-positive responsive microglia surrounding amyloid plaque and an increased accumulation of lipid droplets in APOE4 human brains and induced pluripotent stem cell-derived microglia. Our findings establish critical isoform-dependent effects of microglia/CAM-expressed apoE in brain function and the development of amyloid pathology, providing new insight into how apoE4 vastly increases AD risk.
Collapse
Affiliation(s)
- Chia-Chen Liu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA.
- Neurodegenerative Diseases Research Unit, Biogen, Cambridge, MA, USA.
| | - Na Wang
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Yuanxin Chen
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Yasuteru Inoue
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Francis Shue
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Yingxue Ren
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL, USA
| | - Minghui Wang
- Department of Genetics and Genomic Sciences, Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Wenhui Qiao
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | - Zonghua Li
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Jing Zhao
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Yuka Martens
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Sydney V Doss
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | | | - Lin Jia
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | - Fangfang Qi
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Yiyang Zhu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Alla Alnobani
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Joshua Knight
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Yixing Chen
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Cynthia Linares
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Aishe Kurti
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - John D Fryer
- Department of Neuroscience, Mayo Clinic, Scottsdale, AZ, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Betty Y S Kim
- Department of Neurosurgery, The Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA.
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong, China.
| |
Collapse
|
25
|
Balu D, Valencia-Olvera AC, Islam Z, Mielczarek C, Hansen A, Perez Ramos TM, York J, LaDu MJ, Tai LM. APOE genotype and sex modulate Alzheimer's disease pathology in aged EFAD transgenic mice. Front Aging Neurosci 2023; 15:1279343. [PMID: 38020764 PMCID: PMC10644540 DOI: 10.3389/fnagi.2023.1279343] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 10/10/2023] [Indexed: 12/01/2023] Open
Abstract
Increasing evidence supports that age, APOE and sex interact to modulate Alzheimer's disease (AD) risk, however the underlying pathways are unclear. One way that AD risk factors may modulate cognition is by impacting amyloid beta (Aβ) accumulation as plaques, and/or neuroinflammation Therefore, the goal of the present study was to evaluate the extent to which age, APOE and sex modulate Aβ pathology, neuroinflammation and behavior in vivo. To achieve this goal, we utilized the EFAD mice, which express human APOE3 or APOE4 and have five familial AD mutations (FAD) that result in Aβ42 overproduction. We assessed Aβ levels, reactive glia and Morris water maze performance in 6-, 10-, 14-, and 18-month-old EFAD mice. Female APOE4 mice had the highest Aβ deposition, fibrillar amyloid deposits and neuroinflammation as well as earlier behavior deficits. Interestingly, we found that female APOE3 mice and male APOE4 mice had similar levels of pathology. Collectively our data support that the combination of APOE4 and female sex is the most detrimental combination for AD, and that at older ages, female sex may be equivalent to APOE4 genotype.
Collapse
Affiliation(s)
- Deebika Balu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Ana C. Valencia-Olvera
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Zarak Islam
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
- University of Illinois College of Medicine, Chicago, IL, United States
| | - Clare Mielczarek
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Allison Hansen
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
- University of Illinois College of Medicine, Peoria, IL, United States
| | - Tamara M. Perez Ramos
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
- School of Medicine, St. George’s University, St. George’s, Grenada
| | - Jason York
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Mary Jo LaDu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Leon M. Tai
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
26
|
Marottoli FM, Zhang H, Flores-Barrera E, Artur de la Villarmois E, Damen FC, Miguelez Fernández AM, Blesson HV, Chaudhary R, Nguyen AL, Nwokeji AE, Talati R, John AS, Madadakere K, Lutz SE, Cai K, Tseng KY, Tai LM. Endothelial Cell APOE3 Regulates Neurovascular, Neuronal, and Behavioral Function. Arterioscler Thromb Vasc Biol 2023; 43:1952-1966. [PMID: 37650329 PMCID: PMC10521805 DOI: 10.1161/atvbaha.123.319816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 08/17/2023] [Indexed: 09/01/2023]
Abstract
BACKGROUND Specialized brain endothelial cells and human APOE3 are independently important for neurovascular function, yet whether APOE3 expression by endothelial cells contributes to brain function is currently unknown. In the present study, we determined whether the loss of endothelial cell APOE3 impacts brain vascular and neural function. METHODS We developed APOE3fl/fl/Cdh5(PAC)-CreERT2+/- (APOE3Cre+/-) and APOE3fl/fl/Cdh5(PAC)-CreERT2-/- (APOE3Cre-/-, control) mice and induced endothelial cell APOE3 knockdown with tamoxifen at ≈4 to 5 weeks of age. Neurovascular and neuronal function were evaluated by biochemistry, immunohistochemistry, behavioral testing, and electrophysiology at 9 months of age. RESULTS We found that the loss of endothelial APOE3 expression was sufficient to cause neurovascular dysfunction including higher permeability and lower vessel coverage in tandem with deficits in spatial memory and fear memory extinction and a disruption of cortical excitatory/inhibitory balance. CONCLUSIONS Our data collectively support the novel concept that endothelial APOE3 plays a critical role in the regulation of the neurovasculature, neural circuit function, and behavior.
Collapse
Affiliation(s)
- Felecia M. Marottoli
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Hui Zhang
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Eden Flores-Barrera
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Emilce Artur de la Villarmois
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | | | - Anabel M.M. Miguelez Fernández
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Hannah V. Blesson
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Rohan Chaudhary
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Anthony L. Nguyen
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Amanda E. Nwokeji
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Ruju Talati
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Ashwin S. John
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Kushi Madadakere
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Sarah E. Lutz
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Kejia Cai
- Radiology (F.C.D., K.C.), University of Illinois at Chicago
- Bioengineering (K.C.), University of Illinois at Chicago
| | - Kuei Y. Tseng
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| | - Leon M. Tai
- Departments of Anatomy and Cell Biology (F.M.M., H.Z., E.F.-B., E.A.d.l.V., A.M.M.M.F., H.V.B., R.C., A.L.N., A.E.N., R.T., A.S.J., K.M., S.E.L., K.Y.T., L.M.T.), University of Illinois at Chicago
| |
Collapse
|
27
|
Chen F, Ke Q, Wei W, Cui L, Wang Y. Apolipoprotein E and viral infection: Risks and Mechanisms. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 33:529-542. [PMID: 37588688 PMCID: PMC10425688 DOI: 10.1016/j.omtn.2023.07.031] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/18/2023]
Abstract
Apolipoprotein E (ApoE) is a multifunctional protein critical for lipid metabolism and cholesterol homeostasis. In addition to being a well known genetic determinant of both neurodegenerative and cardiovascular diseases, ApoE is frequently involved in various viral infection-related diseases. Human ApoE protein is functionally polymorphic with three isoforms, namely, ApoE2, ApoE3, and ApoE4, with markedly altered protein structures and functions. ApoE4 is associated with increased susceptibility to infection with herpes simplex virus type-1 and HIV. Conversely, ApoE4 protects against hepatitis C virus and hepatitis B virus infection. With the outbreak of coronavirus disease 2019, ApoE4 has been shown to determine the incidence and progression of severe acute respiratory syndrome coronavirus 2 infection. These findings clearly indicate the critical role of ApoE in viral infection. Furthermore, ApoE polymorphism has various or even opposite effects in these infection processes, which are partly related to the structural features that distinguish the different ApoE statuses. In the current review, we summarize the emerging relationship between ApoE and viral infection, discuss the potential mechanisms, and identify future directions that may help to advance our understanding of the link between ApoE and viral infection.
Collapse
Affiliation(s)
- Feng Chen
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Qiongwei Ke
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Wenyan Wei
- Department of Gerontology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Lili Cui
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| | - Yan Wang
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China
| |
Collapse
|
28
|
Cozachenco D, Zimmer ER, Lourenco MV. Emerging concepts towards a translational framework in Alzheimer's disease. Neurosci Biobehav Rev 2023; 152:105246. [PMID: 37236385 DOI: 10.1016/j.neubiorev.2023.105246] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 05/20/2023] [Accepted: 05/22/2023] [Indexed: 05/28/2023]
Abstract
Over the past decades, significant efforts have been made to understand the precise mechanisms underlying the pathogenesis of Alzheimer's disease (AD), the most common cause of dementia. However, clinical trials targeting AD pathological hallmarks have consistently failed. Refinement of AD conceptualization, modeling, and assessment is key to developing successful therapies. Here, we review critical findings and discuss emerging ideas to integrate molecular mechanisms and clinical approaches in AD. We further propose a refined workflow for animal studies incorporating multimodal biomarkers used in clinical studies - delineating critical paths for drug discovery and translation. Addressing unresolved questions with the proposed conceptual and experimental framework may accelerate the development of effective disease-modifying strategies for AD.
Collapse
Affiliation(s)
- Danielle Cozachenco
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Eduardo R Zimmer
- Department of Pharmacology, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Graduate Program in Biological Sciences: Biochemistry (PPGBioq), UFRGS, Porto Alegre, RS, Brazil; Pharmacology and Therapeutics (PPGFT), UFRGS, Porto Alegre, RS, Brazil; McGill Centre for Studies in Aging, McGill University, Montreal, Canada; Brain Institute of Rio Grande do Sul, PUCRS, Porto Alegre, Brazil.
| | - Mychael V Lourenco
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
29
|
Jackson RJ, Keiser MS, Meltzer JC, Fykstra DP, Dierksmeier SE, Melloni A, Nakajima T, Tecedor L, Ranum PT, Carrell E, Chen Y, Holtzman DM, Davidson BL, Hyman BT. APOE2 gene therapy reduces amyloid deposition, and improves markers of neuroinflammation and neurodegeneration in a mouse model of Alzheimer disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.14.552850. [PMID: 37645718 PMCID: PMC10461997 DOI: 10.1101/2023.08.14.552850] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Epidemiological studies show that individuals who carry the relatively uncommon APOE ε2 allele rarely develop Alzheimer disease, and if they do they have a later age of onset, milder clinical course, and less severe neuropathological findings than others with Alzheimer disease. The contrast is especially stark in comparison to the phenotype associated with the major genetic risk factor for Alzheimer disease, APOE ε4, which has an age of onset several decades earlier, as well as a more aggressive clinical course and notably more severe neuropathological findings, especially in terms of the amount of amyloid deposition. Even one APOE ε2 allele improves phenotype, but it is uncertain if that is due to the replacement of a more toxic allele by APOE ε2, or if APOE ε2 has a protective, neuro-modulatory effect. Here, we demonstrate that brain exposure to APOE2 via a gene therapy approach which bathes the entire cortical mantle in the gene product after transduction of the ependyma, rapidly ameliorates established Aβ plaque deposition, neurodegenerative synaptic loss, and, remarkably, reduces microglial activation in an APP/PS1 mouse model despite continued expression of human APOE4. This result suggests a promising protective effect of exogenous APOE2, revealing a cell non-autonomous effect of the protein on microglial activation. We also show that plaque associated microglia in the brain of patients who inherit APOE2 similarly have less microglial reactivity to plaques. These data raise the potential that an APOE2 therapeutic could be effective in Alzheimer disease even in individuals born with the risk ε4 allele. One Sentence Summary Introduction of ApoE2 using an AAV that transduces the ependymal cells of the ventricle causes a reduction in amyloid load and plaque associated synapse loss, and reduces neuroinflammation by modulating microglial responsiveness to plaques.
Collapse
|
30
|
Iadecola C, Anfray A, Schaeffer S, Hattori Y, Santisteban M, Casey N, Wang G, Strickland M, Zhou P, Holtzman D, Anrather J, Park L. Cell autonomous role of border associated macrophages in ApoE4 neurovascular dysfunction and susceptibility to white matter injury. RESEARCH SQUARE 2023:rs.3.rs-3222611. [PMID: 37577565 PMCID: PMC10418550 DOI: 10.21203/rs.3.rs-3222611/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Apolipoprotein-E4 (ApoE4), the strongest genetic risk factor for sporadic Alzheimer's disease, is also a risk factor for microvascular pathologies leading to cognitive impairment, particularly subcortical white matter injury. These effects have been attributed to alterations in the regulation of the brain blood supply, but the cellular source of ApoE4 and the underlying mechanisms remain unclear. In mice expressing human ApoE3 or ApoE4 we report that border associated macrophages (BAM), myeloid cells closely apposed to neocortical microvessels, are both the source and the target of the ApoE4 mediating the neurovascular dysfunction through reactive oxygen species. ApoE4 in BAM is solely responsible for the increased susceptibility to oligemic white matter damage in ApoE4 mice and is sufficient to enhance damage in ApoE3 mice. The data unveil a new aspect of BAM pathobiology and highlight a previously unrecognized cell autonomous role of BAM in the neurovascular dysfunction of ApoE4 with potential therapeutic implications.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Gang Wang
- Feil Family Brain and Mind Research Institute - Weill Cornell Medicine
| | | | | | | | | | - Laibaik Park
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY
| |
Collapse
|
31
|
Yang LG, March ZM, Stephenson RA, Narayan PS. Apolipoprotein E in lipid metabolism and neurodegenerative disease. Trends Endocrinol Metab 2023; 34:430-445. [PMID: 37357100 PMCID: PMC10365028 DOI: 10.1016/j.tem.2023.05.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/05/2023] [Accepted: 05/09/2023] [Indexed: 06/27/2023]
Abstract
Dysregulation of lipid metabolism has emerged as a central component of many neurodegenerative diseases. Variants of the lipid transport protein, apolipoprotein E (APOE), modulate risk and resilience in several neurodegenerative diseases including late-onset Alzheimer's disease (LOAD). Allelic variants of the gene, APOE, alter the lipid metabolism of cells and tissues and have been broadly associated with several other cellular and systemic phenotypes. Targeting APOE-associated metabolic pathways may offer opportunities to alter disease-related phenotypes and consequently, attenuate disease risk and impart resilience to multiple neurodegenerative diseases. We review the molecular, cellular, and tissue-level alterations to lipid metabolism that arise from different APOE isoforms. These changes in lipid metabolism could help to elucidate disease mechanisms and tune neurodegenerative disease risk and resilience.
Collapse
Affiliation(s)
- Linda G Yang
- Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD, USA
| | - Zachary M March
- Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD, USA
| | - Roxan A Stephenson
- Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD, USA
| | - Priyanka S Narayan
- Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD, USA.; National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health, Bethesda, MD, USA; Center for Alzheimer's and Related Dementias (CARD), National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
32
|
Sadleir KR, Vassar R. Connections between ApoE, sleep, and Aβ and tau pathologies in Alzheimer's disease. J Clin Invest 2023; 133:e171838. [PMID: 37463448 PMCID: PMC10348763 DOI: 10.1172/jci171838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2023] Open
Abstract
In this issue of the JCI, Wang and colleagues investigate the relationship between sleep disturbances, an environmental risk factor for Alzheimer's disease (AD), and the apolipoprotein 4 (APOEε4) allele, a strong genetic risk factor for AD. The authors subjected an amyloid mouse model expressing human APOE3 or APOE4, with and without human AD-tau injection, to sleep deprivation and observed that amyloid and tau pathologies were worsened in the presence of APOE4. Moreover, decreased microglial clustering and increased dystrophic neurites around plaques were observed in sleep-deprived APOE4 mice. In addition, aquaporin 4, important for clearing amyloid-β through the glymphatic system, was reduced and less polarized to astrocytic endfeet. These APOE4-induced changes caused alterations in sleep behavior during recovery from sleep deprivation, suggesting a feed-forward cycle of sleep disturbance and increased AD pathology that can further disrupt sleep in the presence of APOE4.
Collapse
Affiliation(s)
| | - Robert Vassar
- Davee Department of Neurology and
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
33
|
Wang C, Nambiar A, Strickland MR, Lee C, Parhizkar S, Moore AC, Musiek ES, Ulrich JD, Holtzman DM. APOE-ε4 synergizes with sleep disruption to accelerate Aβ deposition and Aβ-associated tau seeding and spreading. J Clin Invest 2023; 133:e169131. [PMID: 37279069 PMCID: PMC10351966 DOI: 10.1172/jci169131] [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: 01/25/2023] [Accepted: 04/27/2023] [Indexed: 06/07/2023] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia. The APOE-ε4 allele of the apolipoprotein E (APOE) gene is the strongest genetic risk factor for late-onset AD. The APOE genotype modulates the effect of sleep disruption on AD risk, suggesting a possible link between apoE and sleep in AD pathogenesis, which is relatively unexplored. We hypothesized that apoE modifies Aβ deposition and Aβ plaque-associated tau seeding and spreading in the form of neuritic plaque-tau (NP-tau) pathology in response to chronic sleep deprivation (SD) in an apoE isoform-dependent fashion. To test this hypothesis, we used APPPS1 mice expressing human APOE-ε3 or -ε4 with or without AD-tau injection. We found that SD in APPPS1 mice significantly increased Aβ deposition and peri-plaque NP-tau pathology in the presence of APOE4 but not APOE3. SD in APPPS1 mice significantly decreased microglial clustering around plaques and aquaporin-4 (AQP4) polarization around blood vessels in the presence of APOE4 but not APOE3. We also found that sleep-deprived APPPS1:E4 mice injected with AD-tau had significantly altered sleep behaviors compared with APPPS1:E3 mice. These findings suggest that the APOE-ε4 genotype is a critical modifier in the development of AD pathology in response to SD.
Collapse
|
34
|
Vuu YM, Kadar Shahib A, Rastegar M. The Potential Therapeutic Application of Simvastatin for Brain Complications and Mechanisms of Action. Pharmaceuticals (Basel) 2023; 16:914. [PMID: 37513826 PMCID: PMC10385015 DOI: 10.3390/ph16070914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/14/2023] [Accepted: 06/15/2023] [Indexed: 07/30/2023] Open
Abstract
Statins are common drugs that are clinically used to reduce elevated plasma cholesterol levels. Based on their solubility, statins are considered to be either hydrophilic or lipophilic. Amongst them, simvastatin has the highest lipophilicity to facilitate its ability to cross the blood-brain barrier. Recent studies have suggested that simvastatin could be a promising therapeutic option for different brain complications and diseases ranging from brain tumors (i.e., medulloblastoma and glioblastoma) to neurological disorders (i.e., Alzheimer's disease, Parkinson's disease, and Huntington's disease). Specific mechanisms of disease amelioration, however, are still unclear. Independent studies suggest that simvastatin may reduce the risk of developing certain neurodegenerative disorders. Meanwhile, other studies point towards inducing cell death in brain tumor cell lines. In this review, we outline the potential therapeutic effects of simvastatin on brain complications and review the clinically relevant molecular mechanisms in different cases.
Collapse
Affiliation(s)
| | | | - Mojgan Rastegar
- Department of Biochemistry and Medical Genetics, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| |
Collapse
|
35
|
Huntoon K, Anderson SK, Ballman KV, Twohy E, Dooley K, Jiang W, An Y, Li J, von Roemeling C, Qie Y, Ross OA, Cerhan JH, Whitton AC, Greenspoon JN, Parney IF, Ashman JB, Bahary JP, Hadjipanayis C, Urbanic JJ, Farace E, Khuntia D, Laack NN, Brown PD, Roberge D, Kim BYS. Association of circulating markers with cognitive decline after radiation therapy for brain metastasis. Neuro Oncol 2023; 25:1123-1131. [PMID: 36472389 PMCID: PMC10237411 DOI: 10.1093/neuonc/noac262] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND A recent phase III trial (NCT01372774) comparing use of stereotactic radiosurgery [SRS] versus whole-brain radiation therapy [WBRT] after surgical resection of a single brain metastasis revealed that declines in cognitive function were more common with WBRT than with SRS. A secondary endpoint in that trial, and the primary objective in this secondary analysis, was to identify baseline biomarkers associated with cognitive impairment after either form of radiotherapy for brain metastasis. Here we report our findings on APOE genotype and serum levels of associated proteins and their association with radiation-induced neurocognitive decline. METHODS In this retrospective analysis of prospectively collected samples from a completed randomized clinical trial, patients provided blood samples every 3 months that were tested by genotyping and enzyme-linked immunosorbent assay, and results were analyzed in association with cognitive impairment. RESULTS The APOE genotype was not associated with neurocognitive impairment at 3 months. However, low serum levels of ApoJ, ApoE, or ApoA protein (all P < .01) and higher amyloid beta (Aβ 1-42) levels (P = .048) at baseline indicated a greater likelihood of neurocognitive decline at 3 months after SRS, whereas lower ApoJ levels were associated with decline after WBRT (P = .014). CONCLUSIONS Patients with these pretreatment serum markers should be counseled about radiation-related neurocognitive decline.
Collapse
Affiliation(s)
- Kristin Huntoon
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- The Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - S Keith Anderson
- Alliance Statistics and Data Management Center, Mayo Clinic, Rochester, Minnesota, USA
| | - Karla V Ballman
- Alliance Statistics and Data Management Center, Mayo Clinic, Rochester, Minnesota, USA
- Department of Biostatistics and Epidemiology, Weill Medical College of Cornell University, New York, New York, USA
| | - Erin Twohy
- Alliance Statistics and Data Management Center, Mayo Clinic, Rochester, Minnesota, USA
| | - Katharine Dooley
- Alliance Statistics and Data Management Center, Mayo Clinic, Rochester, Minnesota, USA
| | - Wen Jiang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas,USA
| | - Yi An
- Department of Therapeutic Radiology, Yale-New Haven Hospital, North Haven, Connecticut, USA
| | - Jing Li
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas,USA
| | | | - Yaqing Qie
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- The Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Owen A Ross
- Department of Neuroscience, Mayo Clinic, Jacksonville, Florida, USA
| | - Jane H Cerhan
- Department of Psychiatry and Psychology Mayo Clinic, Rochester, Minnesota, USA
| | - Anthony C Whitton
- Department of Radiation Oncology, McMaster University, Hamilton, Ontario, Canada
| | - Jeffrey N Greenspoon
- Department of Radiation Oncology, McMaster University, Hamilton, Ontario, Canada
| | - Ian F Parney
- Department of Neurosurgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Jonathan B Ashman
- Department of Radiation Oncology, Mayo Clinic, Phoenix/Scottsdale, Arizona, USA
| | - Jean-Paul Bahary
- Department of Radiation Oncology, CHUM, Montreal, Quebec, Canada
| | | | - James J Urbanic
- Department of Radiation Oncology, University of California San Diego, Moores Cancer Center, La Jolla, California, USA
| | - Elana Farace
- Department of Public Health Sciences, Penn State University College of Medicine, Hershey, Pennsylvania, USA
| | - Deepak Khuntia
- Department of Radiation Oncology, Precision Cancer Specialists and Varian Medical Systems, Palo Alto, California, USA
| | - Nadia N Laack
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Paul D Brown
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - David Roberge
- Department of Radiation Oncology, CHUM, Montreal, Quebec, Canada
| | - Betty Y S Kim
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- The Brain Tumor Center, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
36
|
Padmanabhan P, Götz J. Clinical relevance of animal models in aging-related dementia research. NATURE AGING 2023; 3:481-493. [PMID: 37202516 DOI: 10.1038/s43587-023-00402-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 03/21/2023] [Indexed: 05/20/2023]
Abstract
Alzheimer's disease (AD) and other, less prevalent dementias are complex age-related disorders that exhibit multiple etiologies. Over the past decades, animal models have provided pathomechanistic insight and evaluated countless therapeutics; however, their value is increasingly being questioned due to the long history of drug failures. In this Perspective, we dispute this criticism. First, the utility of the models is limited by their design, as neither the etiology of AD nor whether interventions should occur at a cellular or network level is fully understood. Second, we highlight unmet challenges shared between animals and humans, including impeded drug transport across the blood-brain barrier, limiting effective treatment development. Third, alternative human-derived models also suffer from the limitations mentioned above and can only act as complementary resources. Finally, age being the strongest AD risk factor should be better incorporated into the experimental design, with computational modeling expected to enhance the value of animal models.
Collapse
Affiliation(s)
- Pranesh Padmanabhan
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, the University of Queensland, Brisbane, Queensland, Australia
| | - Jürgen Götz
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, the University of Queensland, Brisbane, Queensland, Australia.
| |
Collapse
|
37
|
Cashikar AG, Toral-Rios D, Timm D, Romero J, Strickland M, Long JM, Han X, Holtzman DM, Paul SM. Regulation of astrocyte lipid metabolism and ApoE secretionby the microglial oxysterol, 25-hydroxycholesterol. J Lipid Res 2023; 64:100350. [PMID: 36849076 PMCID: PMC10060115 DOI: 10.1016/j.jlr.2023.100350] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 01/30/2023] [Accepted: 02/13/2023] [Indexed: 02/27/2023] Open
Abstract
Neuroinflammation, a major hallmark of Alzheimer's disease and several other neurological and psychiatric disorders, is often associated with dysregulated cholesterol metabolism. Relative to homeostatic microglia, activated microglia express higher levels of Ch25h, an enzyme that hydroxylates cholesterol to produce 25-hydroxycholesterol (25HC). 25HC is an oxysterol with interesting immune roles stemming from its ability to regulate cholesterol metabolism. Since astrocytes synthesize cholesterol in the brain and transport it to other cells via ApoE-containing lipoproteins, we hypothesized that secreted 25HC from microglia may influence lipid metabolism as well as extracellular ApoE derived from astrocytes. Here, we show that astrocytes take up externally added 25HC and respond with altered lipid metabolism. Extracellular levels of ApoE lipoprotein particles increased after treatment of astrocytes with 25HC without an increase in Apoe mRNA expression. In mouse astrocytes-expressing human ApoE3 or ApoE4, 25HC promoted extracellular ApoE3 better than ApoE4. Increased extracellular ApoE was due to elevated efflux from increased Abca1 expression via LXRs as well as decreased lipoprotein reuptake from suppressed Ldlr expression via inhibition of SREBP. 25HC also suppressed expression of Srebf2, but not Srebf1, leading to reduced cholesterol synthesis in astrocytes without affecting fatty acid levels. We further show that 25HC promoted the activity of sterol-o-acyl transferase that led to a doubling of the amount of cholesteryl esters and their concomitant storage in lipid droplets. Our results demonstrate an important role for 25HC in regulating astrocyte lipid metabolism.
Collapse
Affiliation(s)
- Anil G Cashikar
- Department of Psychiatry, Washington University School of Medicine, St Louis, Missouri, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St Louis, Missouri, USA; Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St Louis, Missouri, USA; Department of Neurology, Washington University School of Medicine, St Louis, Missouri, USA.
| | - Danira Toral-Rios
- Department of Psychiatry, Washington University School of Medicine, St Louis, Missouri, USA
| | - David Timm
- Department of Psychiatry, Washington University School of Medicine, St Louis, Missouri, USA
| | - Johnathan Romero
- Department of Psychiatry, Washington University School of Medicine, St Louis, Missouri, USA
| | - Michael Strickland
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Justin M Long
- Hope Center for Neurological Disorders, Washington University School of Medicine, St Louis, Missouri, USA; Department of Neurology, Washington University School of Medicine, St Louis, Missouri, USA; Knight Alzheimer Disease Research Center, Washington University School of Medicine, St Louis, Missouri, USA
| | - Xianlin Han
- Barshop Institute for Longevity and Aging Studies, Department of Medicine, University of Texas Health Science Center, San Antonio, Texas, USA
| | - David M Holtzman
- Hope Center for Neurological Disorders, Washington University School of Medicine, St Louis, Missouri, USA; Department of Neurology, Washington University School of Medicine, St Louis, Missouri, USA; Knight Alzheimer Disease Research Center, Washington University School of Medicine, St Louis, Missouri, USA
| | - Steven M Paul
- Department of Psychiatry, Washington University School of Medicine, St Louis, Missouri, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St Louis, Missouri, USA; Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St Louis, Missouri, USA; Department of Neurology, Washington University School of Medicine, St Louis, Missouri, USA
| |
Collapse
|
38
|
Xiong M, Wang C, Gratuze M, Saadi F, Bao X, Bosch ME, Lee C, Jiang H, Serrano JR, Gonzales ER, Kipnis M, Holtzman DM. Astrocytic APOE4 removal confers cerebrovascular protection despite increased cerebral amyloid angiopathy. Mol Neurodegener 2023; 18:17. [PMID: 36922879 PMCID: PMC10018855 DOI: 10.1186/s13024-023-00610-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 03/02/2023] [Indexed: 03/18/2023] Open
Abstract
BACKGROUND Alzheimer Disease (AD) and cerebral amyloid angiopathy (CAA) are both characterized by amyloid-β (Aβ) accumulation in the brain, although Aβ deposits mostly in the brain parenchyma in AD and in the cerebrovasculature in CAA. The presence of CAA can exacerbate clinical outcomes of AD patients by promoting spontaneous intracerebral hemorrhage and ischemia leading to CAA-associated cognitive decline. Genetically, AD and CAA share the ε4 allele of the apolipoprotein E (APOE) gene as the strongest genetic risk factor. Although tremendous efforts have focused on uncovering the role of APOE4 on parenchymal plaque pathogenesis in AD, mechanistic studies investigating the role of APOE4 on CAA are still lacking. Here, we addressed whether abolishing APOE4 generated by astrocytes, the major producers of APOE, is sufficient to ameliorate CAA and CAA-associated vessel damage. METHODS We generated transgenic mice that deposited both CAA and plaques in which APOE4 expression can be selectively suppressed in astrocytes. At 2-months-of-age, a timepoint preceding CAA and plaque formation, APOE4 was removed from astrocytes of 5XFAD APOE4 knock-in mice. Mice were assessed at 10-months-of-age for Aβ plaque and CAA pathology, gliosis, and vascular integrity. RESULTS Reducing the levels of APOE4 in astrocytes shifted the deposition of fibrillar Aβ from the brain parenchyma to the cerebrovasculature. However, despite increased CAA, astrocytic APOE4 removal reduced overall Aβ-mediated gliosis and also led to increased cerebrovascular integrity and function in vessels containing CAA. CONCLUSION In a mouse model of CAA, the reduction of APOE4 derived specifically from astrocytes, despite increased fibrillar Aβ deposition in the vasculature, is sufficient to reduce Aβ-mediated gliosis and cerebrovascular dysfunction.
Collapse
Affiliation(s)
- Monica Xiong
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110 USA
- Division of Biology and Biomedical Sciences (DBBS), Washington University School of Medicine, St. Louis, MO 63110 USA
- Present Address: Genentech, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Chao Wang
- Institute for Brain Science and Disease, Chongqing Medical University, Chongqing, 400016 China
| | - Maud Gratuze
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110 USA
- Present address: Institute of Neurophysiopathology (INP UMR7051), CNRS, Aix-Marseille Université, Marseille, 13005 France
| | - Fareeha Saadi
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Xin Bao
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Megan E. Bosch
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Choonghee Lee
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Hong Jiang
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Javier Remolina Serrano
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Ernesto R. Gonzales
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Michal Kipnis
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - David M. Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110 USA
| |
Collapse
|
39
|
Chen X, Firulyova M, Manis M, Herz J, Smirnov I, Aladyeva E, Wang C, Bao X, Finn MB, Hu H, Shchukina I, Kim MW, Yuede CM, Kipnis J, Artyomov MN, Ulrich JD, Holtzman DM. Microglia-mediated T cell infiltration drives neurodegeneration in tauopathy. Nature 2023; 615:668-677. [PMID: 36890231 PMCID: PMC10258627 DOI: 10.1038/s41586-023-05788-0] [Citation(s) in RCA: 206] [Impact Index Per Article: 206.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 02/03/2023] [Indexed: 03/10/2023]
Abstract
Extracellular deposition of amyloid-β as neuritic plaques and intracellular accumulation of hyperphosphorylated, aggregated tau as neurofibrillary tangles are two of the characteristic hallmarks of Alzheimer's disease1,2. The regional progression of brain atrophy in Alzheimer's disease highly correlates with tau accumulation but not amyloid deposition3-5, and the mechanisms of tau-mediated neurodegeneration remain elusive. Innate immune responses represent a common pathway for the initiation and progression of some neurodegenerative diseases. So far, little is known about the extent or role of the adaptive immune response and its interaction with the innate immune response in the presence of amyloid-β or tau pathology6. Here we systematically compared the immunological milieux in the brain of mice with amyloid deposition or tau aggregation and neurodegeneration. We found that mice with tauopathy but not those with amyloid deposition developed a unique innate and adaptive immune response and that depletion of microglia or T cells blocked tau-mediated neurodegeneration. Numbers of T cells, especially those of cytotoxic T cells, were markedly increased in areas with tau pathology in mice with tauopathy and in the Alzheimer's disease brain. T cell numbers correlated with the extent of neuronal loss, and the cells dynamically transformed their cellular characteristics from activated to exhausted states along with unique TCR clonal expansion. Inhibition of interferon-γ and PDCD1 signalling both significantly ameliorated brain atrophy. Our results thus reveal a tauopathy- and neurodegeneration-related immune hub involving activated microglia and T cell responses, which could serve as therapeutic targets for preventing neurodegeneration in Alzheimer's disease and primary tauopathies.
Collapse
MESH Headings
- Animals
- Mice
- Alzheimer Disease/immunology
- Alzheimer Disease/metabolism
- Alzheimer Disease/pathology
- Amyloid beta-Peptides/immunology
- Amyloid beta-Peptides/metabolism
- Brain/immunology
- Brain/metabolism
- Brain/pathology
- Microglia/immunology
- Microglia/metabolism
- Neurofibrillary Tangles/immunology
- Neurofibrillary Tangles/metabolism
- Neurofibrillary Tangles/pathology
- tau Proteins/immunology
- tau Proteins/metabolism
- Tauopathies/immunology
- Tauopathies/metabolism
- Tauopathies/pathology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- T-Lymphocytes/pathology
- Plaque, Amyloid/immunology
- Plaque, Amyloid/metabolism
- Plaque, Amyloid/pathology
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- T-Lymphocytes, Cytotoxic/pathology
- Clone Cells/immunology
- Clone Cells/metabolism
- Clone Cells/pathology
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Immunity, Innate
Collapse
Affiliation(s)
- Xiaoying Chen
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St Louis, MO, USA
| | - Maria Firulyova
- Almazov National Medical Research Centre, St Petersburg, Russia
| | - Melissa Manis
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St Louis, MO, USA
| | - Jasmin Herz
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
- Center for Brain Immunology and Glia (BIG), Washington University School of Medicine, St Louis, MO, USA
| | - Igor Smirnov
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
- Center for Brain Immunology and Glia (BIG), Washington University School of Medicine, St Louis, MO, USA
| | - Ekaterina Aladyeva
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Chanung Wang
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St Louis, MO, USA
| | - Xin Bao
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St Louis, MO, USA
| | - Mary Beth Finn
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St Louis, MO, USA
| | - Hao Hu
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St Louis, MO, USA
| | - Irina Shchukina
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Min Woo Kim
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
- Center for Brain Immunology and Glia (BIG), Washington University School of Medicine, St Louis, MO, USA
| | - Carla M Yuede
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St Louis, MO, USA
| | - Jonathan Kipnis
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St Louis, MO, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
- Center for Brain Immunology and Glia (BIG), Washington University School of Medicine, St Louis, MO, USA
| | - Maxim N Artyomov
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Jason D Ulrich
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St Louis, MO, USA
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St Louis, MO, USA.
- Center for Brain Immunology and Glia (BIG), Washington University School of Medicine, St Louis, MO, USA.
| |
Collapse
|
40
|
Kessler K, Giannisis A, Bial G, Foquet L, Nielsen HM, Raber J. Behavioral and cognitive performance of humanized APOEε3/ε3 liver mice in relation to plasma apolipoprotein E levels. Sci Rep 2023; 13:1728. [PMID: 36720957 PMCID: PMC9889814 DOI: 10.1038/s41598-023-28165-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 01/13/2023] [Indexed: 02/01/2023] Open
Abstract
Plasma apolipoprotein E levels were previously associated with the risk of developing Alzheimer's disease (AD), levels of cerebrospinal fluid AD biomarkers, cognition and imaging brain measures. Outside the brain, the liver is the primary source of apoE and liver transplantation studies have demonstrated that liver-derived apoE does not cross the blood-brain-barrier. How hepatic apoE may be implicated in behavioral and cognitive performance is not clear. In the current study, we behaviorally tested FRGN mice with humanized liver harboring the ε3/ε3 genotype (E3-human liver (HL)) and compared their behavioral and cognitive performance with that of age-matched ε3/ε3 targeted replacement (E3-TR) mice, the latter produces human apoE3 throughout the body whereas the E3-HL mice endogenously produce human apoE3 only in the liver. We also compared the liver weights and plasma apoE levels, and assessed whether plasma apoE levels were correlated with behavioral or cognitive measures in both models. E3-HL were more active but performed cognitively worse than E3-TR mice. E3-HL mice moved more in the open field containing objects, showed higher activity levels in the Y maze, showed higher activity levels during the baseline period in the fear conditioning test than E3-TR mice, and swam faster than E3-TR mice during training to locate the visible platform in the water maze. However, E3-HL mice showed reduced spatial memory retention in the water maze and reduced fear learning and contextual and cued fear memory than E3-TR mice. Liver weights were greater in E3-HL than E3-TR mice and sex-dependent only in the latter model. Plasma apoE3 levels were similar to those found in humans and comparable in female and male E3-TR mice but higher in female E3-HL mice. Finally, we found correlations between plasma apoE levels and behavioral and cognitive measures which were predominantly model-dependent. Our study demonstrates mouse-model dependent associations between plasma apoE levels, behavior and cognition in an 'AD-neutral' setting and suggests that a humanized liver might be sufficient to induce mouse behavioral and cognitive phenotypes.
Collapse
Affiliation(s)
- Kat Kessler
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Andreas Giannisis
- Department of Biochemistry and Biophysics, Stockholm University, 10691, Stockholm, Sweden
| | - Greg Bial
- Yecuris Corporation, Tualatin, OR, 97062, USA
| | | | - Henrietta M Nielsen
- Department of Biochemistry and Biophysics, Stockholm University, 10691, Stockholm, Sweden.
| | - Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239, USA. .,Departments of Neurology and Radiation Medicine, Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, 97239, USA.
| |
Collapse
|
41
|
The role of ApoE-mediated microglial lipid metabolism in brain aging and disease. IMMUNOMETABOLISM (COBHAM (SURREY, ENGLAND)) 2023; 5:e00018. [PMID: 36710921 PMCID: PMC9869962 DOI: 10.1097/in9.0000000000000018] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 12/20/2022] [Indexed: 01/31/2023]
Abstract
Microglia are a unique population of immune cells resident in the brain that integrate complex signals and dynamically change phenotypes in response to the brain microenvironment. In recent years, single-cell sequencing analyses have revealed profound cellular heterogeneity and context-specific transcriptional plasticity of microglia during brain development, aging, and disease. Emerging evidence suggests that microglia adapt phenotypic plasticity by flexibly reprogramming cellular metabolism to fulfill distinct immune functions. The control of lipid metabolism is central to the appropriate function and homeostasis of the brain. Microglial lipid metabolism regulated by apolipoprotein E (ApoE), a crucial lipid transporter in the brain, has emerged as a critical player in regulating neuroinflammation. The ApoE gene allelic variant, ε4, is associated with a greater risk for neurodegenerative diseases. In this review, we explore novel discoveries in microglial lipid metabolism mediated by ApoE. We elaborate on the functional impact of perturbed microglial lipid metabolism on the underlying pathogenesis of brain aging and disease.
Collapse
|
42
|
Seo DO, O’Donnell D, Jain N, Ulrich JD, Herz J, Li Y, Lemieux M, Cheng J, Hu H, Serrano JR, Bao X, Franke E, Karlsson M, Meier M, Deng S, Desai C, Dodiya H, Lelwala-Guruge J, Handley SA, Kipnis J, Sisodia SS, Gordon JI, Holtzman DM. ApoE isoform- and microbiota-dependent progression of neurodegeneration in a mouse model of tauopathy. Science 2023; 379:eadd1236. [PMID: 36634180 PMCID: PMC9901565 DOI: 10.1126/science.add1236] [Citation(s) in RCA: 95] [Impact Index Per Article: 95.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 11/22/2022] [Indexed: 01/13/2023]
Abstract
Tau-mediated neurodegeneration is a hallmark of Alzheimer's disease. Primary tauopathies are characterized by pathological tau accumulation and neuronal and synaptic loss. Apolipoprotein E (ApoE)-mediated neuroinflammation is involved in the progression of tau-mediated neurodegeneration, and emerging evidence suggests that the gut microbiota regulates neuroinflammation in an APOE genotype-dependent manner. However, evidence of a causal link between the microbiota and tau-mediated neurodegeneration is lacking. In this study, we characterized a genetically engineered mouse model of tauopathy expressing human ApoE isoforms reared under germ-free conditions or after perturbation of their gut microbiota with antibiotics. Both of these manipulations reduced gliosis, tau pathology, and neurodegeneration in a sex- and ApoE isoform-dependent manner. The findings reveal mechanistic and translationally relevant interrelationships between the microbiota, neuroinflammation, and tau-mediated neurodegeneration.
Collapse
Affiliation(s)
- Dong-oh Seo
- Department of Neurology, Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO. USA
| | - David O’Donnell
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO. USA; Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO. USA
| | - Nimansha Jain
- Department of Neurology, Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO. USA
| | - Jason D. Ulrich
- Department of Neurology, Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO. USA
| | - Jasmin Herz
- Center for Brain Immunology and Glia (BIG), Washington University School of Medicine, St. Louis, MO. USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO. USA
| | - Yuhao Li
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, MO. USA
| | - Mackenzie Lemieux
- Center for Brain Immunology and Glia (BIG), Washington University School of Medicine, St. Louis, MO. USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO. USA
| | - Jiye Cheng
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO. USA; Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO. USA
| | - Hao Hu
- Department of Neurology, Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO. USA
| | - Javier R. Serrano
- Department of Neurology, Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO. USA
| | - Xin Bao
- Department of Neurology, Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO. USA
| | - Emily Franke
- Department of Neurology, Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO. USA
| | - Maria Karlsson
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO. USA; Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO. USA
| | - Martin Meier
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO. USA; Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO. USA
| | - Su Deng
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO. USA; Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO. USA
| | - Chandani Desai
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO. USA; Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO. USA
| | - Hemraj Dodiya
- Department of Neurobiology, The University of Chicago, Chicago, IL, 60637, USA
| | - Janaki Lelwala-Guruge
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO. USA; Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO. USA
| | - Scott A. Handley
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO. USA
| | - Jonathan Kipnis
- Center for Brain Immunology and Glia (BIG), Washington University School of Medicine, St. Louis, MO. USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO. USA
| | - Sangram S. Sisodia
- Department of Neurobiology, The University of Chicago, Chicago, IL, 60637, USA
| | - Jeffrey I. Gordon
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO. USA; Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO. USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO. USA
| | - David M. Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO. USA
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO. USA
| |
Collapse
|
43
|
Young CB, Johns E, Kennedy G, Belloy ME, Insel PS, Greicius MD, Sperling RA, Johnson KA, Poston KL, Mormino EC. APOE effects on regional tau in preclinical Alzheimer's disease. Mol Neurodegener 2023; 18:1. [PMID: 36597122 PMCID: PMC9811772 DOI: 10.1186/s13024-022-00590-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 12/14/2022] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND APOE variants are strongly associated with abnormal amyloid aggregation and additional direct effects of APOE on tau aggregation are reported in animal and human cell models. The degree to which these effects are present in humans when individuals are clinically unimpaired (CU) but have abnormal amyloid (Aβ+) remains unclear. METHODS We analyzed data from CU individuals in the Anti-Amyloid Treatment in Asymptomatic AD (A4) and Longitudinal Evaluation of Amyloid Risk and Neurodegeneration (LEARN) studies. Amyloid PET data were available for 4486 participants (3163 Aβ-, 1323 Aβ+) and tau PET data were available for a subset of 447 participants (55 Aβ-, 392 Aβ+). Linear models examined APOE (number of e2 and e4 alleles) associations with global amyloid and regional tau burden in medial temporal lobe (entorhinal, amygdala) and early neocortical regions (inferior temporal, inferior parietal, precuneus). Consistency of APOE4 effects on regional tau were examined in 220 Aβ + CU and mild cognitive impairment (MCI) participants from the Alzheimer's Disease Neuroimaging Initiative (ADNI). RESULTS APOE2 and APOE4 were associated with lower and higher amyloid positivity rates, respectively. Among Aβ+ CU, e2 and e4 were associated with reduced (-12 centiloids per allele) and greater (+15 centiloids per allele) continuous amyloid burden, respectively. APOE2 was associated with reduced regional tau in all regions (-0.05 to -0.09 SUVR per allele), whereas APOE4 was associated with greater regional tau (+0.02 to +0.07 SUVR per allele). APOE differences were confirmed by contrasting e3/e3 with e2/e3 and e3/e4. Mediation analyses among Aβ+ s showed that direct effects of e2 on regional tau were present in medial temporal lobe and early neocortical regions, beyond an indirect pathway mediated by continuous amyloid burden. For e4, direct effects on regional tau were only significant in medial temporal lobe. The magnitude of protective e2 effects on regional tau was consistent across brain regions, whereas detrimental e4 effects were greatest in medial temporal lobe. APOE4 patterns were confirmed in Aβ+ ADNI participants. CONCLUSIONS APOE influences early regional tau PET burden, above and beyond effects related to cross-sectional amyloid PET burden. Therapeutic strategies targeting underlying mechanisms related to APOE may modify tau accumulation among Aβ+ individuals.
Collapse
Affiliation(s)
- Christina B Young
- Stanford University School of Medicine, 453 Quarry Rd., Palo Alto, Stanford, CA, 94304, USA.
| | - Emily Johns
- Stanford University School of Medicine, 453 Quarry Rd., Palo Alto, Stanford, CA, 94304, USA
| | - Gabriel Kennedy
- Stanford University School of Medicine, 453 Quarry Rd., Palo Alto, Stanford, CA, 94304, USA
| | - Michael E Belloy
- Stanford University School of Medicine, 453 Quarry Rd., Palo Alto, Stanford, CA, 94304, USA
| | - Philip S Insel
- University of California San Francisco, San Francisco, CA, USA
| | - Michael D Greicius
- Stanford University School of Medicine, 453 Quarry Rd., Palo Alto, Stanford, CA, 94304, USA
| | - Reisa A Sperling
- Brigham and Women's Hospital, Boston, MA, USA
- Massachusetts General Hospital, Boston, MA, USA
| | - Keith A Johnson
- Brigham and Women's Hospital, Boston, MA, USA
- Massachusetts General Hospital, Boston, MA, USA
| | - Kathleen L Poston
- Stanford University School of Medicine, 453 Quarry Rd., Palo Alto, Stanford, CA, 94304, USA
| | - Elizabeth C Mormino
- Stanford University School of Medicine, 453 Quarry Rd., Palo Alto, Stanford, CA, 94304, USA
| |
Collapse
|
44
|
Nasab AS, Noorani F, Paeizi Z, Khani L, Banaei S, Sadeghi M, Shafeghat M, Shafie M, Mayeli M, Initiative (ADNI) TADN. A Comprehensive Investigation of the Potential Role of Lipoproteins and Metabolite Profile as Biomarkers of Alzheimer's Disease Compared to the Known CSF Biomarkers. Int J Alzheimers Dis 2023; 2023:3540020. [PMID: 36936136 PMCID: PMC10019964 DOI: 10.1155/2023/3540020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 12/22/2022] [Accepted: 01/30/2023] [Indexed: 03/14/2023] Open
Abstract
Introduction While cerebrospinal fluid (CSF) core biomarkers have been considered diagnostic biomarkers for a long time, special attention has been recently dedicated to lipoproteins and metabolites that could be potentially associated with Alzheimer's disease (AD) neurodegeneration. Herein, we aimed to investigate the relationship between the levels of CSF core biomarkers including Aβ-42, TAU, and P-TAU and plasma lipoproteins and metabolites of patients with AD from the baseline cohort of the Alzheimer's Disease Neuroimaging Initiative (ADNI) database. Method Using the ADNI database, fourteen subclasses of lipoproteins as well as a number of lipids and fatty acids and low-molecular metabolites including amino acids, ketone bodies, and glycolysis-related metabolites in blood samples were measured as potential noninvasive markers, and their association with the CSF core biomarkers was statistically investigated controlling for age and gender. Results A total number of 251 AD subjects were included, among whom 71 subjects were negative for the Apo-E ε4 allele and 150 were positive. There was no significant difference between the two groups regarding cognitive assessments, CSF core biomarkers, and lipoproteins and metabolites except the level of Aβ-42 (p < 0.001) and phenylalanine (p = 0.049), which were higher in the negative group. CSF TAU and P-TAU were significantly correlated with medium and small HDL in the negative group, and with extremely large VLDL in the positive group. Our results also indicated significant correlations of metabolites including unsaturated fatty acids, glycerol, and leucine with CSF core biomarkers. Conclusion Based on our findings, a number of lipoproteins and metabolites were associated with CSF core biomarkers of AD. These correlations showed some differences in Apo-E ε4 positive and negative groups, which reminds the role of Apo-E gene status in the pathophysiology of AD development. However, further research is warranted to explore the exact association of lipoproteins and other metabolites with AD core biomarkers and pathology.
Collapse
Affiliation(s)
- Azam Sajjadi Nasab
- 1NeuroTRACT Association, Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Noorani
- 1NeuroTRACT Association, Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Paeizi
- 1NeuroTRACT Association, Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Leila Khani
- 1NeuroTRACT Association, Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Saba Banaei
- 1NeuroTRACT Association, Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Sadeghi
- 1NeuroTRACT Association, Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Melika Shafeghat
- 1NeuroTRACT Association, Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
- 2School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahan Shafie
- 1NeuroTRACT Association, Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
- 2School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahsa Mayeli
- 1NeuroTRACT Association, Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
- 2School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
45
|
Johnson NR, Wang ACJ, Coughlan C, Sillau S, Lucero E, Viltz L, Markham N, Allen C, Dhanasekaran AR, Chial HJ, Potter H. Imipramine and olanzapine block apoE4-catalyzed polymerization of Aβ and show evidence of improving Alzheimer’s disease cognition. Alzheimers Res Ther 2022; 14:88. [PMID: 35768831 PMCID: PMC9241285 DOI: 10.1186/s13195-022-01020-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 05/11/2022] [Indexed: 01/18/2023]
Abstract
Background The apolipoprotein E (APOE) ε4 allele confers the strongest risk for late-onset Alzheimer’s disease (AD) besides age itself, but the mechanisms underlying this risk are debated. One hypothesis supported by evidence from multiple labs is that apoE4 binds to the amyloid-β (Aβ) peptide and catalyzes its polymerization into neurotoxic oligomers and fibrils. Inhibiting this early step in the amyloid cascade may thereby reduce or prevent neurodegeneration and AD. Methods Using a design of experiments (DOE) approach, we developed a high-throughput assay to identify inhibitors of apoE4-catalyzed polymerization of Aβ into oligomers and fibrils. We used it to screen the NIH Clinical Collection of small molecule drugs tested previously in human clinical trials. We then evaluated the efficacy and cytotoxicity of the hit compounds in primary neuron models of apoE4-induced Aβ and phosphorylated tau aggregation. Finally, we performed retrospective analyses of the National Alzheimer’s Coordinating Center (NACC) clinical dataset, using Cox regression and Cox proportional hazards models to determine if the use of two FDA-approved hit compounds was associated with better cognitive scores (Mini-Mental State Exam), or improved AD clinical diagnosis, when compared with other medications of the same clinical indication. Results Our high-throughput screen identified eight blood-brain barrier (BBB)-permeable hit compounds that reduced apoE4-catalyzed Aβ oligomer and fibril formation in a dose-dependent manner. Five hit compounds were non-toxic toward cultured neurons and also reduced apoE4-promoted Aβ and tau neuropathology in a dose-dependent manner. Three of the five compounds were determined to be specific inhibitors of apoE4, whereas the other two compounds were Aβ or tau aggregation inhibitors. When prescribed to AD patients for their normal clinical indications, two of the apoE4 inhibitors, imipramine and olanzapine, but not other (non-hit) antipsychotic or antidepressant medications, were associated with improvements in cognition and clinical diagnosis, especially among APOE4 carriers. Conclusions The critical test of any proposed AD mechanism is whether it leads to effective treatments. Our high-throughput screen identified two promising FDA-approved drugs, imipramine and olanzapine, which have no structural, functional, or clinical similarities other than their shared ability to inhibit apoE4-catalyzed Aβ polymerization, thus identifying this mechanism as an essential contribution of apoE4 to AD. Supplementary Information The online version contains supplementary material available at 10.1186/s13195-022-01020-9.
Collapse
|
46
|
Raulin AC, Doss SV, Trottier ZA, Ikezu TC, Bu G, Liu CC. ApoE in Alzheimer’s disease: pathophysiology and therapeutic strategies. Mol Neurodegener 2022; 17:72. [PMID: 36348357 PMCID: PMC9644639 DOI: 10.1186/s13024-022-00574-4] [Citation(s) in RCA: 160] [Impact Index Per Article: 80.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 10/08/2022] [Accepted: 10/13/2022] [Indexed: 11/10/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common cause of dementia worldwide, and its prevalence is rapidly increasing due to extended lifespans. Among the increasing number of genetic risk factors identified, the apolipoprotein E (APOE) gene remains the strongest and most prevalent, impacting more than half of all AD cases. While the ε4 allele of the APOE gene significantly increases AD risk, the ε2 allele is protective relative to the common ε3 allele. These gene alleles encode three apoE protein isoforms that differ at two amino acid positions. The primary physiological function of apoE is to mediate lipid transport in the brain and periphery; however, additional functions of apoE in diverse biological functions have been recognized. Pathogenically, apoE seeds amyloid-β (Aβ) plaques in the brain with apoE4 driving earlier and more abundant amyloids. ApoE isoforms also have differential effects on multiple Aβ-related or Aβ-independent pathways. The complexity of apoE biology and pathobiology presents challenges to designing effective apoE-targeted therapeutic strategies. This review examines the key pathobiological pathways of apoE and related targeting strategies with a specific focus on the latest technological advances and tools.
Collapse
|
47
|
Barisano G, Kisler K, Wilkinson B, Nikolakopoulou AM, Sagare AP, Wang Y, Gilliam W, Huuskonen MT, Hung ST, Ichida JK, Gao F, Coba MP, Zlokovic BV. A "multi-omics" analysis of blood-brain barrier and synaptic dysfunction in APOE4 mice. J Exp Med 2022; 219:e20221137. [PMID: 36040482 PMCID: PMC9435921 DOI: 10.1084/jem.20221137] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/02/2022] [Accepted: 08/04/2022] [Indexed: 02/02/2023] Open
Abstract
Apolipoprotein E4 (APOE4), the main susceptibility gene for Alzheimer's disease, leads to blood-brain barrier (BBB) breakdown in humans and mice. Remarkably, BBB dysfunction predicts cognitive decline and precedes synaptic deficits in APOE4 human carriers. How APOE4 affects BBB and synaptic function at a molecular level, however, remains elusive. Using single-nucleus RNA-sequencing and phosphoproteome and proteome analysis, we show that APOE4 compared with APOE3 leads to an early disruption of the BBB transcriptome in 2-3-mo-old APOE4 knock-in mice, followed by dysregulation in protein signaling networks controlling cell junctions, cytoskeleton, clathrin-mediated transport, and translation in brain endothelium, as well as transcription and RNA splicing suggestive of DNA damage in pericytes. Changes in BBB signaling mechanisms paralleled an early, progressive BBB breakdown and loss of pericytes, which preceded postsynaptic interactome disruption and behavioral deficits that developed 2-5 mo later. Thus, dysregulated signaling mechanisms in endothelium and pericytes in APOE4 mice reflect a molecular signature of a progressive BBB failure preceding changes in synaptic function and behavior.
Collapse
Affiliation(s)
- Giuseppe Barisano
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA
| | - Kassandra Kisler
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Brent Wilkinson
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Angeliki Maria Nikolakopoulou
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Abhay P. Sagare
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Yaoming Wang
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - William Gilliam
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Mikko T. Huuskonen
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Shu-Ting Hung
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at University of Southern California, Los Angeles, CA
| | - Justin K. Ichida
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at University of Southern California, Los Angeles, CA
| | - Fan Gao
- Caltech Bioinformatics Resource Center, Caltech, Pasadena, CA
| | - Marcelo P. Coba
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Berislav V. Zlokovic
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA
- Alzheimer’s Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, CA
| |
Collapse
|
48
|
Liver-ing in your head rent free: peripheral ApoE4 drives CNS pathology. Mol Neurodegener 2022; 17:65. [PMID: 36195891 PMCID: PMC9531445 DOI: 10.1186/s13024-022-00569-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 09/22/2022] [Indexed: 11/10/2022] Open
|
49
|
Chen Z, Yuan Z, Yang S, Zhu Y, Xue M, Zhang J, Leng L. Brain Energy Metabolism: Astrocytes in Neurodegenerative Diseases. CNS Neurosci Ther 2022; 29:24-36. [PMID: 36193573 PMCID: PMC9804080 DOI: 10.1111/cns.13982] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 08/23/2022] [Accepted: 09/11/2022] [Indexed: 02/06/2023] Open
Abstract
Astrocytes are the most abundant cells in the brain. They have many important functions in the central nervous system (CNS), including the maintenance of glutamate and ion homeostasis, the elimination of oxidative stress, energy storage in glycogen, tissue repair, regulating synaptic activity by releasing neurotransmitters, and participating in synaptic formation. Astrocytes have special highly ramified structure. Their branches contact with synapses of neurons inwardly, with fine structure and wrapping synapses; their feet contact with blood vessels of brain parenchyma outward, almost wrapping the whole brain. The adjacent astrocytes rarely overlap and communicate with each other through gap junction channels. The ideal location of astrocytes enables them to sense the weak changes of their surroundings and provide the structural basis for the energy supply of neurons. Neurons and astrocytes are closely coupled units of energy metabolism in the brain. Neurons consume a lot of ATPs in the process of neurotransmission. Astrocytes provide metabolic substrates for neurons, maintain high activity of neuron, and facilitate information transmission of neurons. This article reviews the characteristics of glucose metabolism, lipid metabolism, and amino acid metabolism of astrocytes. The metabolic interactions between astrocytes and neurons, astrocytes and microglia were also detailed discussed. Finally, we classified analyzed the role of metabolic disorder of astrocytes in the occurrence and development of neurodegenerative diseases.
Collapse
Affiliation(s)
- Zhenlei Chen
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging ResearchInstitute of Neuroscience, School of Medicine, Xiamen UniversityXiamenChina
| | - Ziqi Yuan
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging ResearchInstitute of Neuroscience, School of Medicine, Xiamen UniversityXiamenChina
| | - Shangchen Yang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging ResearchInstitute of Neuroscience, School of Medicine, Xiamen UniversityXiamenChina
| | - Yufei Zhu
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging ResearchInstitute of Neuroscience, School of Medicine, Xiamen UniversityXiamenChina
| | - Maoqiang Xue
- Department of Basic Medical Science, School of MedicineXiamen UniversityXiamenChina
| | - Jie Zhang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging ResearchInstitute of Neuroscience, School of Medicine, Xiamen UniversityXiamenChina
| | - Lige Leng
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging ResearchInstitute of Neuroscience, School of Medicine, Xiamen UniversityXiamenChina
| |
Collapse
|
50
|
Fernández-Calle R, Konings SC, Frontiñán-Rubio J, García-Revilla J, Camprubí-Ferrer L, Svensson M, Martinson I, Boza-Serrano A, Venero JL, Nielsen HM, Gouras GK, Deierborg T. APOE in the bullseye of neurodegenerative diseases: impact of the APOE genotype in Alzheimer's disease pathology and brain diseases. Mol Neurodegener 2022; 17:62. [PMID: 36153580 PMCID: PMC9509584 DOI: 10.1186/s13024-022-00566-4] [Citation(s) in RCA: 77] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 08/29/2022] [Indexed: 02/06/2023] Open
Abstract
ApoE is the major lipid and cholesterol carrier in the CNS. There are three major human polymorphisms, apoE2, apoE3, and apoE4, and the genetic expression of APOE4 is one of the most influential risk factors for the development of late-onset Alzheimer's disease (AD). Neuroinflammation has become the third hallmark of AD, together with Amyloid-β plaques and neurofibrillary tangles of hyperphosphorylated aggregated tau protein. This review aims to broadly and extensively describe the differential aspects concerning apoE. Starting from the evolution of apoE to how APOE's single-nucleotide polymorphisms affect its structure, function, and involvement during health and disease. This review reflects on how APOE's polymorphisms impact critical aspects of AD pathology, such as the neuroinflammatory response, particularly the effect of APOE on astrocytic and microglial function and microglial dynamics, synaptic function, amyloid-β load, tau pathology, autophagy, and cell-cell communication. We discuss influential factors affecting AD pathology combined with the APOE genotype, such as sex, age, diet, physical exercise, current therapies and clinical trials in the AD field. The impact of the APOE genotype in other neurodegenerative diseases characterized by overt inflammation, e.g., alpha- synucleinopathies and Parkinson's disease, traumatic brain injury, stroke, amyotrophic lateral sclerosis, and multiple sclerosis, is also addressed. Therefore, this review gathers the most relevant findings related to the APOE genotype up to date and its implications on AD and CNS pathologies to provide a deeper understanding of the knowledge in the APOE field.
Collapse
Affiliation(s)
- Rosalía Fernández-Calle
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden
| | - Sabine C. Konings
- Department of Experimental Medical Science, Experimental Dementia Research Unit, Lund University, Lund, Sweden
| | - Javier Frontiñán-Rubio
- Oxidative Stress and Neurodegeneration Group, Faculty of Medicine, Universidad de Castilla-La Mancha, Ciudad Real, Spain
| | - Juan García-Revilla
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden
- Departamento de Bioquímica Y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, and Instituto de Biomedicina de Sevilla-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Lluís Camprubí-Ferrer
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden
| | - Martina Svensson
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden
| | - Isak Martinson
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden
| | - Antonio Boza-Serrano
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden
- Departamento de Bioquímica Y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, and Instituto de Biomedicina de Sevilla-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - José Luís Venero
- Departamento de Bioquímica Y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, and Instituto de Biomedicina de Sevilla-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Henrietta M. Nielsen
- Department of Biochemistry and Biophysics at, Stockholm University, Stockholm, Sweden
| | - Gunnar K. Gouras
- Department of Experimental Medical Science, Experimental Dementia Research Unit, Lund University, Lund, Sweden
| | - Tomas Deierborg
- Department of Experimental Medical Science, Experimental Neuroinflammation Laboratory, Lund University, Lund, Sweden
| |
Collapse
|