1
|
Lindner K, Gavin AC. Isoform- and cell-state-specific APOE homeostasis and function. Neural Regen Res 2024; 19:2456-2466. [PMID: 38526282 PMCID: PMC11090418 DOI: 10.4103/nrr.nrr-d-23-01470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/17/2023] [Accepted: 12/26/2023] [Indexed: 03/26/2024] Open
Abstract
Apolipoprotein E is the major lipid transporter in the brain and an important player in neuron-astrocyte metabolic coupling. It ensures the survival of neurons under stressful conditions and hyperactivity by nourishing and detoxifying them. Apolipoprotein E polymorphism, combined with environmental stresses and/or age-related alterations, influences the risk of developing late-onset Alzheimer's disease. In this review, we discuss our current knowledge of how apolipoprotein E homeostasis, i.e. its synthesis, secretion, degradation, and lipidation, is affected in Alzheimer's disease.
Collapse
Affiliation(s)
- Karina Lindner
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Anne-Claude Gavin
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
2
|
Meyer-Acosta KK, Diaz-Guerra E, Varma P, Aruk A, Mirsadeghi S, Perez AM, Rafati Y, Hosseini A, Nieto-Estevez V, Giugliano M, Navara C, Hsieh J. APOE4 impacts cortical neurodevelopment and alters network formation in human brain organoids. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.07.617044. [PMID: 39416105 PMCID: PMC11482793 DOI: 10.1101/2024.10.07.617044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Apolipoprotein E4 ( APOE4 ) is the leading genetic risk factor for Alzheimer's disease. While most studies examine the role of APOE4 in aging, imaging, and cognitive assessments reveal that APOE4 influences brain structure and function as early as infancy. Here, we examined human-relevant cellular phenotypes across neurodevelopment using induced pluripotent stem cell (iPSC) derived cortical and ganglionic eminence organoids (COs and GEOs). In COs, we showed that APOE4 decreased BRN2+ and SATB2+ cortical neurons, increased astrocytes and outer radial glia, and was associated with increased cell death and dysregulated GABA-related gene expression. In GEOs, APOE4 accelerated maturation of neural progenitors and neurons. Multi-electrode array recordings in assembloids revealed that APOE4 disrupted network formation and altered response to GABA, resulting in heightened excitability and synchronicity. Together, our data provides new insights into how APOE4 may influence cortical neurodevelopmental processes and network formation in the human brain.
Collapse
|
3
|
Zuniga NR, Earls NE, Denos AEA, Elison JM, Jones BS, Smith EG, Moran NG, Brown KL, Romero GM, Hyer CD, Wagstaff KB, Almughamsi HM, Transtrum MK, Price JC. Quantitative and Kinetic Proteomics Reveal ApoE Isoform-dependent Proteostasis Adaptations in Mouse Brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.13.607719. [PMID: 39185235 PMCID: PMC11343127 DOI: 10.1101/2024.08.13.607719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Apolipoprotein E (ApoE) polymorphisms modify the risk of neurodegenerative disease with the ApoE4 isoform increasing and ApoE2 isoform decreasing risk relative to the 'wild-type control' ApoE3 isoform. To elucidate how ApoE isoforms alter the proteome, we measured relative protein abundance and turnover in transgenic mice expressing a human ApoE gene (isoform 2, 3, or 4). This data provides insight into how ApoE isoforms affect the in vivo synthesis and degradation of a wide variety of proteins. We identified 4849 proteins and tested for ApoE isoform-dependent changes in the homeostatic regulation of ~2700 ontologies. In the brain, we found that ApoE4 and ApoE2 both lead to modified regulation of mitochondrial membrane proteins relative to the wild-type control ApoE3. In ApoE4 mice, this regulation is not cohesive suggesting that aerobic respiration is impacted by proteasomal and autophagic dysregulation. ApoE2 mice exhibited a matching change in mitochondrial matrix proteins and the membrane which suggests coordinated maintenance of the entire organelle. In the liver, we did not observe these changes suggesting that the ApoE-effect on proteostasis is amplified in the brain relative to other tissues. Our findings underscore the utility of combining protein abundance and turnover rates to decipher proteome regulatory mechanisms and their potential role in biology.
Collapse
Affiliation(s)
- Nathan R. Zuniga
- Department of Chemistry and Biochemistry, College of Computational, Physical, and Mathematical Sciences, Brigham Young University, Provo, UT, USA
| | - Noah E. Earls
- Department of Chemistry and Biochemistry, College of Computational, Physical, and Mathematical Sciences, Brigham Young University, Provo, UT, USA
| | - Ariel E. A. Denos
- Department of Chemistry and Biochemistry, College of Computational, Physical, and Mathematical Sciences, Brigham Young University, Provo, UT, USA
| | - Jared M. Elison
- Department of Chemistry and Biochemistry, College of Computational, Physical, and Mathematical Sciences, Brigham Young University, Provo, UT, USA
| | - Benjamin S. Jones
- Department of Chemistry and Biochemistry, College of Computational, Physical, and Mathematical Sciences, Brigham Young University, Provo, UT, USA
| | - Ethan G. Smith
- Department of Chemistry and Biochemistry, College of Computational, Physical, and Mathematical Sciences, Brigham Young University, Provo, UT, USA
| | - Noah G. Moran
- Department of Chemistry and Biochemistry, College of Computational, Physical, and Mathematical Sciences, Brigham Young University, Provo, UT, USA
| | - Katie L. Brown
- Department of Chemistry and Biochemistry, College of Computational, Physical, and Mathematical Sciences, Brigham Young University, Provo, UT, USA
| | - Gerome M. Romero
- Department of Chemistry and Biochemistry, College of Computational, Physical, and Mathematical Sciences, Brigham Young University, Provo, UT, USA
| | - Chad D. Hyer
- Department of Chemistry and Biochemistry, College of Computational, Physical, and Mathematical Sciences, Brigham Young University, Provo, UT, USA
| | - Kimberly B. Wagstaff
- Department of Chemistry and Biochemistry, College of Computational, Physical, and Mathematical Sciences, Brigham Young University, Provo, UT, USA
| | - Haifa M. Almughamsi
- Department of Chemistry and Biochemistry, College of Computational, Physical, and Mathematical Sciences, Brigham Young University, Provo, UT, USA
- Department of Chemistry, College of Science, Taif University, Taif, Saudi Arabia
| | - Mark K. Transtrum
- Department of Physics and Astronomy, College of Computational, Physical, and Mathematical Sciences, Brigham Young University, Provo, UT, USA
| | - John C. Price
- Department of Chemistry and Biochemistry, College of Computational, Physical, and Mathematical Sciences, Brigham Young University, Provo, UT, USA
| |
Collapse
|
4
|
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
|
5
|
Hees JT, Wanderoy S, Lindner J, Helms M, Murali Mahadevan H, Harbauer AB. Insulin signalling regulates Pink1 mRNA localization via modulation of AMPK activity to support PINK1 function in neurons. Nat Metab 2024; 6:514-530. [PMID: 38504131 PMCID: PMC10963278 DOI: 10.1038/s42255-024-01007-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 02/06/2024] [Indexed: 03/21/2024]
Abstract
Mitochondrial quality control failure is frequently observed in neurodegenerative diseases. The detection of damaged mitochondria by stabilization of PTEN-induced kinase 1 (PINK1) requires transport of Pink1 messenger RNA (mRNA) by tethering it to the mitochondrial surface. Here, we report that inhibition of AMP-activated protein kinase (AMPK) by activation of the insulin signalling cascade prevents Pink1 mRNA binding to mitochondria. Mechanistically, AMPK phosphorylates the RNA anchor complex subunit SYNJ2BP within its PDZ domain, a phosphorylation site that is necessary for its interaction with the RNA-binding protein SYNJ2. Notably, loss of mitochondrial Pink1 mRNA association upon insulin addition is required for PINK1 protein activation and its function as a ubiquitin kinase in the mitophagy pathway, thus placing PINK1 function under metabolic control. Induction of insulin resistance in vitro by the key genetic Alzheimer risk factor apolipoprotein E4 retains Pink1 mRNA at the mitochondria and prevents proper PINK1 activity, especially in neurites. Our results thus identify a metabolic switch controlling Pink1 mRNA localization and PINK1 activity via insulin and AMPK signalling in neurons and propose a mechanistic connection between insulin resistance and mitochondrial dysfunction.
Collapse
Affiliation(s)
- J Tabitha Hees
- TUM Medical Graduate Center, Technical University of Munich, Munich, Germany
- Max Planck Institute for Biological Intelligence, Martinsried, Germany
| | - Simone Wanderoy
- TUM Medical Graduate Center, Technical University of Munich, Munich, Germany
- Max Planck Institute for Biological Intelligence, Martinsried, Germany
| | - Jana Lindner
- Max Planck Institute for Biological Intelligence, Martinsried, Germany
| | - Marlena Helms
- Max Planck Institute for Biological Intelligence, Martinsried, Germany
| | - Hariharan Murali Mahadevan
- TUM Medical Graduate Center, Technical University of Munich, Munich, Germany
- Max Planck Institute for Biological Intelligence, Martinsried, Germany
| | - Angelika B Harbauer
- Max Planck Institute for Biological Intelligence, Martinsried, Germany.
- Technical University of Munich, Institute of Neuronal Cell Biology, Munich, Germany.
- Munich Cluster for Systems Neurology, Munich, Germany.
| |
Collapse
|
6
|
Shafqat A, Khan S, Omer MH, Niaz M, Albalkhi I, AlKattan K, Yaqinuddin A, Tchkonia T, Kirkland JL, Hashmi SK. Cellular senescence in brain aging and cognitive decline. Front Aging Neurosci 2023; 15:1281581. [PMID: 38076538 PMCID: PMC10702235 DOI: 10.3389/fnagi.2023.1281581] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 11/01/2023] [Indexed: 10/16/2024] Open
Abstract
Cellular senescence is a biological aging hallmark that plays a key role in the development of neurodegenerative diseases. Clinical trials are currently underway to evaluate the effectiveness of senotherapies for these diseases. However, the impact of senescence on brain aging and cognitive decline in the absence of neurodegeneration remains uncertain. Moreover, patient populations like cancer survivors, traumatic brain injury survivors, obese individuals, obstructive sleep apnea patients, and chronic kidney disease patients can suffer age-related brain changes like cognitive decline prematurely, suggesting that they may suffer accelerated senescence in the brain. Understanding the role of senescence in neurocognitive deficits linked to these conditions is crucial, especially considering the rapidly evolving field of senotherapeutics. Such treatments could help alleviate early brain aging in these patients, significantly reducing patient morbidity and healthcare costs. This review provides a translational perspective on how cellular senescence plays a role in brain aging and age-related cognitive decline. We also discuss important caveats surrounding mainstream senotherapies like senolytics and senomorphics, and present emerging evidence of hyperbaric oxygen therapy and immune-directed therapies as viable modalities for reducing senescent cell burden.
Collapse
Affiliation(s)
- Areez Shafqat
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | | | - Mohamed H. Omer
- School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Mahnoor Niaz
- Medical College, Aga Khan University, Karachi, Pakistan
| | | | - Khaled AlKattan
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | | | - Tamara Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, United States
| | - James L. Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, United States
| | - Shahrukh K. Hashmi
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
- Clinical Affairs, Khalifa University, Abu Dhabi, United Arab Emirates
- Department of Medicine, SSMC, Abu Dhabi, United Arab Emirates
| |
Collapse
|
7
|
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
|
8
|
Wynne ME, Ogunbona O, Lane AR, Gokhale A, Zlatic SA, Xu C, Wen Z, Duong DM, Rayaprolu S, Ivanova A, Ortlund EA, Dammer EB, Seyfried NT, Roberts BR, Crocker A, Shanbhag V, Petris M, Senoo N, Kandasamy S, Claypool SM, Barrientos A, Wingo A, Wingo TS, Rangaraju S, Levey AI, Werner E, Faundez V. APOE expression and secretion are modulated by mitochondrial dysfunction. eLife 2023; 12:e85779. [PMID: 37171075 PMCID: PMC10231934 DOI: 10.7554/elife.85779] [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: 12/23/2022] [Accepted: 05/11/2023] [Indexed: 05/13/2023] Open
Abstract
Mitochondria influence cellular function through both cell-autonomous and non-cell autonomous mechanisms, such as production of paracrine and endocrine factors. Here, we demonstrate that mitochondrial regulation of the secretome is more extensive than previously appreciated, as both genetic and pharmacological disruption of the electron transport chain caused upregulation of the Alzheimer's disease risk factor apolipoprotein E (APOE) and other secretome components. Indirect disruption of the electron transport chain by gene editing of SLC25A mitochondrial membrane transporters as well as direct genetic and pharmacological disruption of either complexes I, III, or the copper-containing complex IV of the electron transport chain elicited upregulation of APOE transcript, protein, and secretion, up to 49-fold. These APOE phenotypes were robustly expressed in diverse cell types and iPSC-derived human astrocytes as part of an inflammatory gene expression program. Moreover, age- and genotype-dependent decline in brain levels of respiratory complex I preceded an increase in APOE in the 5xFAD mouse model. We propose that mitochondria act as novel upstream regulators of APOE-dependent cellular processes in health and disease.
Collapse
Affiliation(s)
- Meghan E Wynne
- Department of Cell Biology, Emory UniversityAtlantaUnited States
| | - Oluwaseun Ogunbona
- Department of Cell Biology, Emory UniversityAtlantaUnited States
- Department of Pathology and Laboratory Medicine, Emory UniversityAtlantaUnited States
| | - Alicia R Lane
- Department of Cell Biology, Emory UniversityAtlantaUnited States
| | - Avanti Gokhale
- Department of Cell Biology, Emory UniversityAtlantaUnited States
| | | | - Chongchong Xu
- Department of Psychiatry and Behavioral Sciences, Emory UniversityAtlantaUnited States
| | - Zhexing Wen
- Department of Cell Biology, Emory UniversityAtlantaUnited States
- Department of Psychiatry and Behavioral Sciences, Emory UniversityAtlantaUnited States
- Department of Neurology and Human Genetics, Emory UniversityAtlantaUnited States
| | - Duc M Duong
- Department of Biochemistry, Emory UniversityAtlantaUnited States
| | - Sruti Rayaprolu
- Department of Neurology and Human Genetics, Emory UniversityAtlantaUnited States
| | - Anna Ivanova
- Department of Biochemistry, Emory UniversityAtlantaUnited States
| | - Eric A Ortlund
- Department of Biochemistry, Emory UniversityAtlantaUnited States
| | - Eric B Dammer
- Department of Biochemistry, Emory UniversityAtlantaUnited States
| | | | - Blaine R Roberts
- Department of Biochemistry, Emory UniversityAtlantaUnited States
| | - Amanda Crocker
- Program in Neuroscience, Middlebury CollegeMiddleburyUnited States
| | - Vinit Shanbhag
- Department of Biochemistry, University of MissouriColumbiaUnited States
| | - Michael Petris
- Department of Biochemistry, University of MissouriColumbiaUnited States
| | - Nanami Senoo
- Department of Physiology, Johns Hopkins UniversityBaltimoreUnited States
| | | | | | - Antoni Barrientos
- Department of Neurology and Biochemistry & Molecular Biology, University of MiamiMiamiUnited States
| | - Aliza Wingo
- Department of Neurology and Human Genetics, Emory UniversityAtlantaUnited States
| | - Thomas S Wingo
- Department of Neurology and Human Genetics, Emory UniversityAtlantaUnited States
| | - Srikant Rangaraju
- Department of Neurology and Human Genetics, Emory UniversityAtlantaUnited States
| | - Allan I Levey
- Department of Neurology and Human Genetics, Emory UniversityAtlantaUnited States
| | - Erica Werner
- Department of Cell Biology, Emory UniversityAtlantaUnited States
| | - Victor Faundez
- Department of Cell Biology, Emory UniversityAtlantaUnited States
| |
Collapse
|
9
|
Lee S, Devanney NA, Golden LR, Smith CT, Schwartz JL, Walsh AE, Clarke HA, Goulding DS, Allenger EJ, Morillo-Segovia G, Friday CM, Gorman AA, Hawkinson TR, MacLean SM, Williams HC, Sun RC, Morganti JM, Johnson LA. APOE modulates microglial immunometabolism in response to age, amyloid pathology, and inflammatory challenge. Cell Rep 2023; 42:112196. [PMID: 36871219 PMCID: PMC10117631 DOI: 10.1016/j.celrep.2023.112196] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 11/29/2022] [Accepted: 02/14/2023] [Indexed: 03/06/2023] Open
Abstract
The E4 allele of Apolipoprotein E (APOE) is associated with both metabolic dysfunction and a heightened pro-inflammatory response: two findings that may be intrinsically linked through the concept of immunometabolism. Here, we combined bulk, single-cell, and spatial transcriptomics with cell-specific and spatially resolved metabolic analyses in mice expressing human APOE to systematically address the role of APOE across age, neuroinflammation, and AD pathology. RNA sequencing (RNA-seq) highlighted immunometabolic changes across the APOE4 glial transcriptome, specifically in subsets of metabolically distinct microglia enriched in the E4 brain during aging or following an inflammatory challenge. E4 microglia display increased Hif1α expression and a disrupted tricarboxylic acid (TCA) cycle and are inherently pro-glycolytic, while spatial transcriptomics and mass spectrometry imaging highlight an E4-specific response to amyloid that is characterized by widespread alterations in lipid metabolism. Taken together, our findings emphasize a central role for APOE in regulating microglial immunometabolism and provide valuable, interactive resources for discovery and validation research.
Collapse
Affiliation(s)
- Sangderk Lee
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA
| | - Nicholas A Devanney
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA; Sanders Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA
| | - Lesley R Golden
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
| | - Cathryn T Smith
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
| | - James L Schwartz
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA
| | - Adeline E Walsh
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
| | - Harrison A Clarke
- Department of Neuroscience, University of Kentucky, Lexington, KY 40536, USA; Department of Biochemistry & Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA; Center for Advanced Spatial Biomolecule Research, University of Florida, Gainesville, FL, USA
| | - Danielle S Goulding
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA
| | | | | | - Cassi M Friday
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
| | - Amy A Gorman
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA
| | - Tara R Hawkinson
- Department of Neuroscience, University of Kentucky, Lexington, KY 40536, USA; Department of Biochemistry & Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA; Center for Advanced Spatial Biomolecule Research, University of Florida, Gainesville, FL, USA
| | - Steven M MacLean
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
| | - Holden C Williams
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
| | - Ramon C Sun
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA; Department of Neuroscience, University of Kentucky, Lexington, KY 40536, USA; Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA; Department of Biochemistry & Molecular Biology, College of Medicine, University of Florida, Gainesville, FL, USA; Center for Advanced Spatial Biomolecule Research, University of Florida, Gainesville, FL, USA
| | - Josh M Morganti
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA; Department of Neuroscience, University of Kentucky, Lexington, KY 40536, USA.
| | - Lance A Johnson
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA; Sanders Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA.
| |
Collapse
|
10
|
Ramos J, Caywood LJ, Prough MB, Clouse JE, Herington SD, Slifer SH, Fuzzell MD, Fuzzell SL, Hochstetler SD, Miskimen KL, Main LR, Osterman MD, Zaman AF, Whitehead PL, Adams LD, Laux RA, Song YE, Foroud TM, Mayeux RP, George-Hyslop PS, Ogrocki PK, Lerner AJ, Vance JM, Cuccaro ML, Haines JL, Pericak-Vance MA, Scott WK. Genetic variants in the SHISA6 gene are associated with delayed cognitive impairment in two family datasets. Alzheimers Dement 2023; 19:611-620. [PMID: 35490390 PMCID: PMC9622429 DOI: 10.1002/alz.12686] [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: 09/23/2021] [Revised: 03/08/2022] [Accepted: 03/28/2022] [Indexed: 11/12/2022]
Abstract
INTRODUCTION Studies of cognitive impairment (CI) in Amish communities have identified sibships containing CI and cognitively unimpaired (CU) individuals. We hypothesize that CU individuals may carry protective alleles delaying age at onset (AAO) of CI. METHODS A total of 1522 individuals screened for CI were genotyped. The outcome studied was AAO for CI individuals or age at last normal exam for CU individuals. Cox mixed-effects models examined association between age and single nucleotide variants (SNVs). RESULTS Three SNVs were significantly associated (P < 5 × 10-8 ) with AAO on chromosomes 6 (rs14538074; hazard ratio [HR] = 3.35), 9 (rs534551495; HR = 2.82), and 17 (rs146729640; HR = 6.38). The chromosome 17 association was replicated in the independent National Institute on Aging Genetics Initiative for Late-Onset Alzheimer's Disease dataset. DISCUSSION The replicated genome-wide significant association with AAO on chromosome 17 is located in the SHISA6 gene, which is involved in post-synaptic transmission in the hippocampus and is a biologically plausible candidate gene for Alzheimer's disease.
Collapse
Affiliation(s)
- Jairo Ramos
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Laura J. Caywood
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Michael B. Prough
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jason E. Clouse
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Sharlene D. Herington
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Susan H. Slifer
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - M. Denise Fuzzell
- Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Sarada L. Fuzzell
- Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | | | | | - Leighanne R. Main
- Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Michael D. Osterman
- Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, USA
| | - Andrew F. Zaman
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Patrice L. Whitehead
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Larry D. Adams
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Renee A. Laux
- Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Yeunjoo E. Song
- Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Tatiana M. Foroud
- Indiana Alzheimer’s Disease Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Richard P. Mayeux
- Taub Institute on Alzheimer’s Disease and the Aging Brain, Department of Neurology, Columbia University, New York, NY, USA
- Gertrude H. Sergievsky Center, Columbia University, New York, NY, USA
- Department of Neurology, Columbia University, New York, NY, USA
| | | | - Paula K. Ogrocki
- University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Alan J. Lerner
- University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Jeffery M. Vance
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
- The Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Michael L. Cuccaro
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
- The Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jonathan L. Haines
- Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, USA
| | - Margaret A. Pericak-Vance
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
- The Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - William K. Scott
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
- The Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
11
|
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
|
12
|
Steele OG, Stuart AC, Minkley L, Shaw K, Bonnar O, Anderle S, Penn AC, Rusted J, Serpell L, Hall C, King S. A multi-hit hypothesis for an APOE4-dependent pathophysiological state. Eur J Neurosci 2022; 56:5476-5515. [PMID: 35510513 PMCID: PMC9796338 DOI: 10.1111/ejn.15685] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 03/31/2022] [Accepted: 04/25/2022] [Indexed: 01/01/2023]
Abstract
The APOE gene encoding the Apolipoprotein E protein is the single most significant genetic risk factor for late-onset Alzheimer's disease. The APOE4 genotype confers a significantly increased risk relative to the other two common genotypes APOE3 and APOE2. Intriguingly, APOE4 has been associated with neuropathological and cognitive deficits in the absence of Alzheimer's disease-related amyloid or tau pathology. Here, we review the extensive literature surrounding the impact of APOE genotype on central nervous system dysfunction, focussing on preclinical model systems and comparison of APOE3 and APOE4, given the low global prevalence of APOE2. A multi-hit hypothesis is proposed to explain how APOE4 shifts cerebral physiology towards pathophysiology through interconnected hits. These hits include the following: neurodegeneration, neurovascular dysfunction, neuroinflammation, oxidative stress, endosomal trafficking impairments, lipid and cellular metabolism disruption, impaired calcium homeostasis and altered transcriptional regulation. The hits, individually and in combination, leave the APOE4 brain in a vulnerable state where further cumulative insults will exacerbate degeneration and lead to cognitive deficits in the absence of Alzheimer's disease pathology and also a state in which such pathology may more easily take hold. We conclude that current evidence supports an APOE4 multi-hit hypothesis, which contributes to an APOE4 pathophysiological state. We highlight key areas where further study is required to elucidate the complex interplay between these individual mechanisms and downstream consequences, helping to frame the current landscape of existing APOE-centric literature.
Collapse
Affiliation(s)
| | | | - Lucy Minkley
- School of Life SciencesUniversity of SussexBrightonUK
| | - Kira Shaw
- School of Life SciencesUniversity of SussexBrightonUK
| | - Orla Bonnar
- School of Life SciencesUniversity of SussexBrightonUK
| | | | | | | | | | | | - Sarah King
- School of PsychologyUniversity of SussexBrightonUK
| |
Collapse
|
13
|
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
|
14
|
Sex and APOE Genotype Alter the Basal and Induced Inflammatory States of Primary Microglia from APOE Targeted Replacement Mice. Int J Mol Sci 2022; 23:ijms23179829. [PMID: 36077227 PMCID: PMC9456163 DOI: 10.3390/ijms23179829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/17/2022] [Accepted: 08/19/2022] [Indexed: 11/16/2022] Open
Abstract
The sex and APOE4 genotype are significant risk factors for Alzheimer’s disease (AD); however, the mechanism(s) responsible for this interaction are still a matter of debate. Here, we assess the responses of mixed-sex and sex-specific APOE3 and APOE4 primary microglia (PMG) to lipopolysaccharide and interferon-gamma. In our investigation, inflammatory cytokine profiles were assessed by qPCR and multiplex ELISA assays. Mixed-sex APOE4 PMG exhibited higher basal mRNA expression and secreted levels of TNFa and IL1b. In sex-specific cultures, basal expression and secreted levels of IL1b, TNFa, IL6, and NOS2 were 2−3 fold higher in APOE4 female PMG compared to APOE4 males, with both higher than APOE3 cells. Following an inflammatory stimulus, the expression of pro-inflammatory cytokines and the secreted cytokine level were upregulated in the order E4 female > E4 male > E3 female > E3 male in sex-specific cultures. These data indicate that the APOE4 genotype and female sex together contribute to a greater inflammatory response in PMG isolated from targeted replacement humanized APOE mice. These data are consistent with clinical data and indicate that sex-specific PMG may provide a platform for exploring mechanisms of genotype and sex differences in AD related to neuroinflammation and neurodegeneration.
Collapse
|
15
|
Rueter J, Rimbach G, Huebbe P. Functional diversity of apolipoprotein E: from subcellular localization to mitochondrial function. Cell Mol Life Sci 2022; 79:499. [PMID: 36018414 PMCID: PMC9418098 DOI: 10.1007/s00018-022-04516-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/27/2022] [Accepted: 08/07/2022] [Indexed: 11/26/2022]
Abstract
Human apolipoprotein E (APOE), originally known for its role in lipid metabolism, is polymorphic with three major allele forms, namely, APOEε2, APOEε3, and APOEε4, leading to three different human APOE isoforms. The ε4 allele is a genetic risk factor for Alzheimer's disease (AD); therefore, the vast majority of APOE research focuses on its role in AD pathology. However, there is increasing evidence for other functions of APOE through the involvement in other biological processes such as transcriptional regulation, mitochondrial metabolism, immune response, and responsiveness to dietary factors. Therefore, the aim of this review is to provide an overview of the potential novel functions of APOE and their characterization. The detection of APOE in various cell organelles points to previously unrecognized roles in mitochondria and others, although it is actually considered a secretory protein. Furthermore, numerous interactions of APOE with other proteins have been detected, providing indications for new metabolic pathways involving APOE. The present review summarizes the current evidence on APOE beyond its original role in lipid metabolism, to change the perspective and encourage novel approaches to future research on APOE and its isoform-dependent role in the cellular metabolism.
Collapse
Affiliation(s)
- Johanna Rueter
- Devision of Food Science, Institute of Human Nutrition and Food Science, University of Kiel, Hermann-Rodewald-Strasse 6, 24118, Kiel, Germany
| | - Gerald Rimbach
- Devision of Food Science, Institute of Human Nutrition and Food Science, University of Kiel, Hermann-Rodewald-Strasse 6, 24118, Kiel, Germany.
| | - Patricia Huebbe
- Devision of Food Science, Institute of Human Nutrition and Food Science, University of Kiel, Hermann-Rodewald-Strasse 6, 24118, Kiel, Germany
| |
Collapse
|
16
|
Foley KE, Diemler CA, Hewes AA, Garceau DT, Sasner M, Howell GR. APOE ε4 and exercise interact in a sex-specific manner to modulate dementia risk factors. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2022; 8:e12308. [PMID: 35783454 PMCID: PMC9241167 DOI: 10.1002/trc2.12308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/23/2022] [Accepted: 05/02/2022] [Indexed: 12/01/2022]
Abstract
Introduction Apolipoprotein E (APOE) ε4 is the strongest genetic risk factor for Alzheimer's disease and related dementias (ADRDs), affecting many different pathways that lead to cognitive decline. Exercise is one of the most widely proposed prevention and intervention strategies to mitigate risk and symptomology of ADRDs. Importantly, exercise and APOE ε4 affect similar processes in the body and brain. While both APOE ε4 and exercise have been studied extensively, their interactive effects are not well understood. Methods To address this, male and female APOE ε3/ε3, APOE ε3/ε4, and APOE ε4/ε4 mice ran voluntarily from wean (1 month) to midlife (12 months). Longitudinal and cross-sectional phenotyping were performed on the periphery and the brain, assessing markers of risk for dementia such as weight, body composition, circulating cholesterol composition, murine daily activities, energy expenditure, and cortical and hippocampal transcriptional profiling. Results Data revealed chronic running decreased age-dependent weight gain, lean and fat mass, and serum low-density lipoprotein concentration dependent on APOE genotype. Additionally, murine daily activities and energy expenditure were significantly influenced by an interaction between APOE genotype and running in both sexes. Transcriptional profiling of the cortex and hippocampus predicted that APOE genotype and running interact to affect numerous biological processes including vascular integrity, synaptic/neuronal health, cell motility, and mitochondrial metabolism, in a sex-specific manner. Discussion These data in humanized mouse models provide compelling evidence that APOE genotype should be considered for population-based strategies that incorporate exercise to prevent ADRDs and other APOE-relevant diseases.
Collapse
Affiliation(s)
- Kate E. Foley
- The Jackson LaboratoryBar HarborMaineUSA
- School of Graduate Biomedical SciencesTufts University School of MedicineBostonMassachusettsUSA
| | | | - Amanda A. Hewes
- The Jackson LaboratoryBar HarborMaineUSA
- Department of PsychologyUniversity of MaineOronoMaineUSA
| | | | | | - Gareth R. Howell
- The Jackson LaboratoryBar HarborMaineUSA
- School of Graduate Biomedical SciencesTufts University School of MedicineBostonMassachusettsUSA
- Graduate School of Biomedical Sciences and EngineeringUniversity of MaineOronoMaineUSA
| |
Collapse
|
17
|
Mitochondrial function and Aβ in Alzheimer's disease postmortem brain. Neurobiol Dis 2022; 171:105781. [PMID: 35667615 DOI: 10.1016/j.nbd.2022.105781] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/15/2022] [Accepted: 05/31/2022] [Indexed: 10/18/2022] Open
Abstract
INTRODUCTION Mitochondrial dysfunction is observed in Alzheimer's disease (AD). However, the relationship between functional mitochondrial deficits and AD pathologies is not well established in human subjects. METHODS Post-mortem human brain tissue from 11 non-demented (ND) and 12 AD subjects was used to examine mitochondrial electron transport chain (ETC) function. Data were analyzed by neuropathology diagnosis and Apolipoprotein E (APOE) genotype. Relationships between AD pathology and mitochondrial function were determined. RESULTS AD subjects had reductions in brain cytochrome oxidase (COX) function and complex II Vmax. APOE ε4 carriers had COX, complex II and III deficits. AD subjects had reduced expression of Complex I-III ETC proteins, no changes were observed in APOE ε4 carriers. No correlation between p-Tau Thr 181 and mitochondrial outcomes was observed, although brains from non-demented subjects demonstrated positive correlations between Aβ concentration and COX Vmax. DISCUSSION These data support a dysregulated relationship between brain mitochondrial function and Aβ pathology in AD.
Collapse
|
18
|
Robb WH, Khan OA, Ahmed HA, Li J, Moore EE, Cambronero FE, Pechman KR, Liu D, Gifford KA, Landman BA, Donahue MJ, Hohman TJ, Jefferson AL. Lower cerebral oxygen utilization is associated with Alzheimer's disease-related neurodegeneration and poorer cognitive performance among apolipoprotein E ε4 carriers. J Cereb Blood Flow Metab 2022; 42:642-655. [PMID: 34743630 PMCID: PMC9051148 DOI: 10.1177/0271678x211056393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 08/05/2021] [Accepted: 09/28/2021] [Indexed: 11/15/2022]
Abstract
Oxygen extraction fraction (OEF) and cerebral metabolic rate of oxygen (CMRO2) are markers of cerebral oxygen homeostasis and metabolism that may offer insights into abnormal changes in brain aging. The present study cross-sectionally related OEF and CMRO2 to cognitive performance and structural neuroimaging variables among older adults (n = 246, 74 ± 7 years, 37% female) and tested whether apolipoprotein E (APOE)-ε4 status modified these associations. Main effects of OEF and CMRO2 were null (p-values >0.06), and OEF interactions with APOE-ε4 status on cognitive and structural imaging outcomes were null (p-values >0.06). However, CMRO2 interacted with APOE-ε4 status on language (p = 0.002), executive function (p = 0.03), visuospatial (p = 0.005), and episodic memory performances (p = 0.03), and on hippocampal (p = 0.006) and inferior lateral ventricle volumes (p = 0.02). In stratified analyses, lower oxygen metabolism related to worse language (p = 0.02) and episodic memory performance (p = 0.03) among APOE-ε4 carriers only. Associations between CMRO2 and cognitive performance were primarily driven by APOE-ε4 carriers with existing cognitive impairment. Congruence across language and episodic memory results as well as hippocampal and inferior lateral ventricle volume findings suggest that APOE-ε4 may interact with cerebral oxygen metabolism in the pathogenesis of Alzheimer's disease and related neurodegeneration.
Collapse
Affiliation(s)
- W Hudson Robb
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University
Medical Center, Nashville, TN, USA
| | - Omair A Khan
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University
Medical Center, Nashville, TN, USA
- Department of Biostatistics, Vanderbilt University Medical
Center, Vanderbilt University Medical Center, Nashville, TN,
USA
| | - Humza A Ahmed
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University
Medical Center, Nashville, TN, USA
| | - Judy Li
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University
Medical Center, Nashville, TN, USA
| | - Elizabeth E Moore
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University
Medical Center, Nashville, TN, USA
| | - Francis E Cambronero
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University
Medical Center, Nashville, TN, USA
| | - Kimberly R Pechman
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University
Medical Center, Nashville, TN, USA
| | - Dandan Liu
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University
Medical Center, Nashville, TN, USA
- Department of Biostatistics, Vanderbilt University Medical
Center, Vanderbilt University Medical Center, Nashville, TN,
USA
| | - Katherine A Gifford
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University
Medical Center, Nashville, TN, USA
- Department of Neurology, Vanderbilt University Medical
Center, Vanderbilt University Medical Center, Nashville, TN,
USA
| | - Bennett A Landman
- Department of Neurology, Vanderbilt University Medical
Center, Vanderbilt University Medical Center, Nashville, TN,
USA
- Department of Biomedical Engineering, Vanderbilt University, Vanderbilt University, Nashville, TN, USA
- Department of Electrical Engineering and Computer Science, Vanderbilt University, Vanderbilt University, Nashville, TN, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical
Center, Vanderbilt University Medical Center, Nashville, TN,
USA
| | - Manus J Donahue
- Department of Neurology, Vanderbilt University Medical
Center, Vanderbilt University Medical Center, Nashville, TN,
USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical
Center, Vanderbilt University Medical Center, Nashville, TN,
USA
- Department of Psychiatry and Behavioral Sciences, Vanderbilt
University Medical Center, Nashville, TN, USA
| | - Timothy J Hohman
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University
Medical Center, Nashville, TN, USA
- Department of Neurology, Vanderbilt University Medical
Center, Vanderbilt University Medical Center, Nashville, TN,
USA
| | - Angela L Jefferson
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University
Medical Center, Nashville, TN, USA
- Department of Neurology, Vanderbilt University Medical
Center, Vanderbilt University Medical Center, Nashville, TN,
USA
- Department of Medicine, Vanderbilt University Medical
Center, Vanderbilt University Medical Center, Nashville, TN,
USA
| |
Collapse
|
19
|
Miranda AM, Ashok A, Chan RB, Zhou B, Xu Y, McIntire LB, Area-Gomez E, Di Paolo G, Duff KE, Oliveira TG, Nuriel T. Effects of APOE4 allelic dosage on lipidomic signatures in the entorhinal cortex of aged mice. Transl Psychiatry 2022; 12:129. [PMID: 35351864 PMCID: PMC8964762 DOI: 10.1038/s41398-022-01881-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/10/2022] [Accepted: 02/25/2022] [Indexed: 12/22/2022] Open
Abstract
Apolipoprotein E ε4 (APOE4) is the primary genetic risk factor for the late-onset form of Alzheimer's disease (AD). Although the reason for this association is not completely understood, researchers have uncovered numerous effects of APOE4 expression on AD-relevant brain processes, including amyloid beta (Aβ) accumulation, lipid metabolism, endosomal-lysosomal trafficking, and bioenergetics. In this study, we aimed to determine the effect of APOE4 allelic dosage on regional brain lipid composition in aged mice, as well as in cultured neurons. We performed a targeted lipidomic analysis on an AD-vulnerable brain region (entorhinal cortex; EC) and an AD-resistant brain region (primary visual cortex; PVC) from 14-15 month-old APOE3/3, APOE3/4, and APOE4/4 targeted replacement mice, as well as on neurons cultured with conditioned media from APOE3/3 or APOE4/4 astrocytes. Our results reveal that the EC possesses increased susceptibility to APOE4-associated lipid alterations compared to the PVC. In the EC, APOE4 expression showed a dominant effect in decreasing diacylglycerol (DAG) levels, and a semi-dominant, additive effect in the upregulation of multiple ceramide, glycosylated sphingolipid, and bis(monoacylglycerol)phosphate (BMP) species, lipids known to accumulate as a result of endosomal-lysosomal dysfunction. Neurons treated with conditioned media from APOE4/4 vs. APOE3/3 astrocytes showed similar alterations of DAG and BMP species to those observed in the mouse EC. Our results suggest that APOE4 expression differentially modulates regional neuronal lipid signatures, which may underlie the increased susceptibility of EC-localized neurons to AD pathology.
Collapse
Affiliation(s)
- André Miguel Miranda
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Neuroradiology Unit, Department of Imagiology, Centro Hospitalar Vila Nova Gaia/Espinho, 4434-502, Vila Nova Gaia, Portugal
| | - Archana Ashok
- 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
| | - Robin Barry Chan
- 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
| | - Bowen Zhou
- 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
| | - Yimeng Xu
- 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
| | - Laura Beth McIntire
- 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
| | - Estela Area-Gomez
- 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 Neurology, Columbia University, 630 West 168th Street, New York, NY, 10032, USA
| | - Gilbert Di Paolo
- 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
- Denali Therapeutics Inc., South San Francisco, CA, 94080, USA
| | - Karen E Duff
- 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
- UK Dementia Research Institute, University College London, Cruciform Building, Gower Street, London, WC1E 6BT, UK
| | - Tiago Gil Oliveira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057, Braga, Portugal.
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
- Department of Neuroradiology, Hospital de Braga, 4710-243, Braga, Portugal.
| | - 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
|
20
|
Foley KE, Hewes AA, Garceau DT, Kotredes KP, Carter GW, Sasner M, Howell GR. The APOEε3/ε4 Genotype Drives Distinct Gene Signatures in the Cortex of Young Mice. Front Aging Neurosci 2022; 14:838436. [PMID: 35370604 PMCID: PMC8967347 DOI: 10.3389/fnagi.2022.838436] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 02/14/2022] [Indexed: 12/22/2022] Open
Abstract
IntroductionRestrictions on existing APOE mouse models have impacted research toward understanding the strongest genetic risk factor contributing to Alzheimer’s disease (AD) and dementia, APOEε4, by hindering observation of a key, common genotype in humans – APOEε3/ε4. Human studies are typically underpowered to address APOEε4 allele risk as the APOEε4/ε4 genotype is rare, which leaves human and mouse research unsupported to evaluate the APOEε3/ε4 genotype on molecular and pathological risk for AD and dementia.MethodsAs a part of MODEL-AD, we created and validated new versions of humanized APOEε3/ε3 and APOEε4/ε4 mouse strains that, due to unrestricted breeding, allow for the evaluation of the APOEε3/ε4 genotype. As biometric measures are often translatable between mouse and human, we profiled circulating lipid concentrations. We also performed transcriptional profiling of the cerebral cortex at 2 and 4 months (mos), comparing APOEε3/ε4 and APOEε4/ε4 to the reference APOEε3/ε3 using linear modeling and WGCNA. Further, APOE mice were exercised and compared to litter-matched sedentary controls, to evaluate the interaction between APOEε4 and exercise at a young age.ResultsExpression of human APOE isoforms were confirmed in APOEε3/ε3, APOEε3/ε4 and APOEε4/ε4 mouse brains. At two mos, cholesterol composition was influenced by sex, but not APOE genotype. Results show that the APOEε3/ε4 and APOEε4/ε4 genotype exert differential effects on cortical gene expression. APOEε3/ε4 uniquely impacts ‘hormone regulation’ and ‘insulin signaling,’ terms absent in APOEε4/ε4 data. At four mos, cholesterol and triglyceride levels were affected by sex and activity, with only triglyceride levels influenced by APOE genotype. Linear modeling revealed APOEε3/ε4, but not APOEε4/ε4, affected ‘extracellular matrix’ and ‘blood coagulation’ related terms. We confirmed these results using WGCNA, indicating robust, yet subtle, transcriptional patterns. While there was little evidence of APOE genotype by exercise interaction on the cortical transcriptome at this young age, running was predicted to affect myelination and gliogenesis, independent of APOE genotype with few APOE genotype-specific affects identified.DiscussionAPOEε4 allele dosage-specific effects were observed in circulating lipid levels and cortical transcriptional profiles. Future studies are needed to establish how these data may contribute to therapeutic development in APOEε3/ε4 and APOEε4/ε4 dementia patients.
Collapse
Affiliation(s)
- Kate E. Foley
- The Jackson Laboratory, Bar Harbor, ME, United States
- School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, United States
| | - Amanda A. Hewes
- The Jackson Laboratory, Bar Harbor, ME, United States
- Department of Psychology, University of Maine, Orono, ME, United States
| | | | | | - Gregory W. Carter
- The Jackson Laboratory, Bar Harbor, ME, United States
- School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, United States
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, United States
| | | | - Gareth R. Howell
- The Jackson Laboratory, Bar Harbor, ME, United States
- School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, United States
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, United States
- *Correspondence: Gareth R. Howell,
| |
Collapse
|
21
|
Khan N, Alimova Y, Clark SJ, Vekaria H, Walsh AE, Williams HC, Hawk GS, Sullivan P, Johnson LA, McClintock TS. Human APOE ɛ3 and APOE ɛ4 Alleles Have Differential Effects on Mouse Olfactory Epithelium. J Alzheimers Dis 2021; 85:1481-1494. [PMID: 34958025 DOI: 10.3233/jad-215152] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is a progressive age-dependent disorder whose risk is affected by genetic factors. Better models for investigating early effects of risk factors such as apolipoprotein E (APOE) genotype are needed. OBJECTIVE To determine whether APOE genotype produces neuropathologies in an AD-susceptible neural system, we compared effects of human APOE ɛ3 (E3) and APOE ɛ4 (E4) alleles on the mouse olfactory epithelium. METHODS RNA-Seq using the STAR aligner and DESeq2, immunohistochemistry for activated caspase-3 and phosphorylated histone H3, glucose uptake after oral gavage of 2-[1,2-3H (N)]-deoxy-D-glucose, and Seahorse Mito Stress tests on dissociated olfactory mucosal cells. RESULTS E3 and E4 olfactory mucosae show 121 differentially abundant mRNAs at age 6 months. These do not indicate differences in cell type proportions, but effects on 17 odorant receptor mRNAs suggest small differences in tissue development. Ten oxidoreductases mRNAs important for cellular metabolism and mitochondria are less abundant in E4 olfactory mucosae but this does not translate into differences in cellular respiration. E4 olfactory mucosae show lower glucose uptake, characteristic of AD susceptibility and consistent with greater expression of the glucose-sensitive gene, Asns. Olfactory sensory neuron apoptosis is unaffected at age 6 months but is greater in E4 mice at 10 months. CONCLUSION Effects of human APOE alleles on mouse olfactory epithelium phenotype are apparent in early adulthood, and neuronal loss begins to increase by middle age (10 months). The olfactory epithelium is an appropriate model for the ability of human APOE alleles to modulate age-dependent effects associated with the progression of AD.
Collapse
Affiliation(s)
- Naazneen Khan
- Department of Physiology, University of Kentucky, Lexington, KY, USA
| | - Yelena Alimova
- Department of Physiology, University of Kentucky, Lexington, KY, USA
| | - Sophie J Clark
- Department of Physiology, University of Kentucky, Lexington, KY, USA
| | - Hemendra Vekaria
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, USA
| | - Adeline E Walsh
- Department of Physiology, University of Kentucky, Lexington, KY, USA
| | - Holden C Williams
- Department of Physiology, University of Kentucky, Lexington, KY, USA.,Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Gregory S Hawk
- Department of Statistics, University of Kentucky, Lexington, KY, USA
| | - Patrick Sullivan
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, USA.,Department of Neuroscience, University of Kentucky, Lexington, KY, USA.,Lexington Veterans' Affairs Healthcare System, Lexington, KY, USA
| | - Lance A Johnson
- Department of Physiology, University of Kentucky, Lexington, KY, USA.,Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | | |
Collapse
|
22
|
Klein HU, Trumpff C, Yang HS, Lee AJ, Picard M, Bennett DA, De Jager PL. Characterization of mitochondrial DNA quantity and quality in the human aged and Alzheimer's disease brain. Mol Neurodegener 2021; 16:75. [PMID: 34742335 PMCID: PMC8572491 DOI: 10.1186/s13024-021-00495-8] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 10/11/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Mitochondrial dysfunction is a feature of neurodegenerative diseases, including Alzheimer's disease (AD). Changes in the mitochondrial DNA copy number (mtDNAcn) and increased mitochondrial DNA mutation burden have both been associated with neurodegenerative diseases and cognitive decline. This study aims to systematically identify which common brain pathologies in the aged human brain are associated with mitochondrial recalibrations and to disentangle the relationship between these pathologies, mtDNAcn, mtDNA heteroplasmy, aging, neuronal loss, and cognitive function. METHODS Whole-genome sequencing data from n = 1361 human brain samples from 5 different regions were used to quantify mtDNAcn as well as heteroplasmic mtDNA point mutations and small indels. Brain samples were assessed for 10 common pathologies. Annual cognitive test results were used to assess cognitive function proximal to death. For a subset of samples, neuronal proportions were estimated from RNA-seq profiles, and mass spectrometry was used to quantify the mitochondrial protein content of the tissue. RESULTS mtDNAcn was 7-14% lower in AD relative to control participants. When accounting for all 10 common neuropathologies, only tau was significantly associated with lower mtDNAcn in the dorsolateral prefrontal cortex. In the posterior cingulate cortex, TDP-43 pathology demonstrated a distinct association with mtDNAcn. No changes were observed in the cerebellum, which is affected late by pathologies. Neither age nor gender was associated with mtDNAcn in the studied brain regions when adjusting for pathologies. Mitochondrial content and mtDNAcn independently explained variance in cognitive function unaccounted by pathologies, implicating complex mitochondrial recalibrations in cognitive decline. In contrast, mtDNA heteroplasmy levels increased by 1.5% per year of life in the cortical regions, but displayed no association with any of the pathologies or cognitive function. CONCLUSIONS We studied mtDNA quantity and quality in relation to mixed pathologies of aging and showed that tau and not amyloid-β is primarily associated with reduced mtDNAcn. In the posterior cingulate cortex, the association of TDP-43 with low mtDNAcn points to a vulnerability of this region in limbic-predominant age-related TDP-43 encephalopathy. While we found low mtDNAcn in brain regions affected by pathologies, the absence of associations with mtDNA heteroplasmy burden indicates that mtDNA point mutations and small indels are unlikely to be involved in the pathogenesis of late-onset neurodegenerative diseases.
Collapse
Affiliation(s)
- Hans-Ulrich Klein
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032 USA
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032 USA
| | - Caroline Trumpff
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY 10032 USA
| | - Hyun-Sik Yang
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women’s Hospital, Boston, MA 02115 USA
| | - Annie J. Lee
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032 USA
| | - Martin Picard
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY 10032 USA
- Merritt Center and Columbia Translational Neuroscience Initiative, Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032 USA
| | - David A. Bennett
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612 USA
| | - Philip L. De Jager
- Center for Translational & Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032 USA
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032 USA
| |
Collapse
|
23
|
Amick KA, Mahapatra G, Bergstrom J, Gao Z, Craft S, Register TC, Shively CA, Molina AJA. Brain region-specific disruption of mitochondrial bioenergetics in cynomolgus macaques fed a Western versus a Mediterranean diet. Am J Physiol Endocrinol Metab 2021; 321:E652-E664. [PMID: 34569271 PMCID: PMC8791787 DOI: 10.1152/ajpendo.00165.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Mitochondrial dysfunction is evident in diseases affecting cognition and metabolism such as Alzheimer's disease and type 2 diabetes. Human studies of brain mitochondrial function are limited to postmortem tissue, preventing the assessment of bioenergetics by respirometry. Here, we investigated the effect of two diets on mitochondrial bioenergetics in three brain regions: the prefrontal cortex (PFC), the entorhinal cortex (ERC), and the cerebellum (CB), using middle-aged nonhuman primates. Eighteen female cynomolgus macaques aged 12.3 ± 0.7 yr were fed either a Mediterranean diet that is associated with healthy outcomes or a Western diet that is associated with poor cognitive and metabolic outcomes. Average bioenergetic capacity within each brain region did not differ between diets. Distinct brain regions have different metabolic requirements related to their function and disease susceptibility. Therefore, we also examined differences in bioenergetic capacity between brain regions. Mitochondria isolated from animals fed a Mediterranean diet maintained distinct differences in mitochondrial bioenergetics between brain regions, whereas animals fed the Western diet had diminished distinction in bioenergetics between brain regions. Notably, fatty acid β-oxidation was not affected between regions in animals fed a Western diet. In addition, bioenergetics in animals fed a Western diet had positive associations with fasting blood glucose and insulin levels in PFC and ERC mitochondria but not in CB mitochondria. Altogether, these data indicate that a Western diet disrupts bioenergetic patterns across brain regions and that circulating blood glucose and insulin levels in Western-diet fed animals influence bioenergetics in brain regions susceptible to Alzheimer's disease and type 2 diabetes.NEW & NOTEWORTHY We show that compared with cynomolgus macaques fed a Mediterranean diet, a Western diet resulted in diminished bioenergetic pattern between brain regions related to blood glucose and insulin levels, specifically in brain regions susceptible to neurodegeneration and diabetes. In addition, fatty acid metabolism not directly linked to the TCA cycle and glucose metabolism did not show differences in bioenergetics due to diet.
Collapse
Affiliation(s)
- K Allison Amick
- Section of Gerontology and Geriatrics, Sticht Center for Healthy Aging and Alzheimer's Prevention, Department of Internal Medicine, Wake Forest School of Medicine, Winston Salem, North Carolina
- Department of Neuroscience, Wake Forest School of Medicine, Winston Salem, North Carolina
| | - Gargi Mahapatra
- Section of Gerontology and Geriatrics, Sticht Center for Healthy Aging and Alzheimer's Prevention, Department of Internal Medicine, Wake Forest School of Medicine, Winston Salem, North Carolina
| | - Jaclyn Bergstrom
- Department of Family Medicine and Public Health, University of California San Diego School of Medicine, La Jolla, California
| | - Zhengrong Gao
- Section of Gerontology and Geriatrics, Sticht Center for Healthy Aging and Alzheimer's Prevention, Department of Internal Medicine, Wake Forest School of Medicine, Winston Salem, North Carolina
| | - Suzanne Craft
- Section of Gerontology and Geriatrics, Sticht Center for Healthy Aging and Alzheimer's Prevention, Department of Internal Medicine, Wake Forest School of Medicine, Winston Salem, North Carolina
| | - Thomas C Register
- Department of Pathology/Comparative Medicine, Wake Forest School of Medicine, Winston Salem, North Carolina
| | - Carol A Shively
- Department of Pathology/Comparative Medicine, Wake Forest School of Medicine, Winston Salem, North Carolina
| | - Anthony J A Molina
- Section of Gerontology and Geriatrics, Sticht Center for Healthy Aging and Alzheimer's Prevention, Department of Internal Medicine, Wake Forest School of Medicine, Winston Salem, North Carolina
- Division of Geriatrics and Gerontology, Department of Medicine, University of California San Diego School of Medicine, La Jolla, California
| |
Collapse
|
24
|
Ryu WI, Cohen BM, Sonntag KC. Hypothesis and Theory: Characterizing Abnormalities of Energy Metabolism Using a Cellular Platform as a Personalized Medicine Approach for Alzheimer's Disease. Front Cell Dev Biol 2021; 9:697578. [PMID: 34395428 PMCID: PMC8363296 DOI: 10.3389/fcell.2021.697578] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 06/27/2021] [Indexed: 01/07/2023] Open
Abstract
Sporadic or late-onset Alzheimer’s disease (LOAD) is characterized by slowly progressive deterioration and death of CNS neurons. There are currently no substantially disease-modifying therapies. LOAD pathology is closely related to changes with age and include, among others, accumulation of toxic molecules and altered metabolic, microvascular, biochemical and inflammatory processes. In addition, there is growing evidence that cellular energy deficits play a critical role in aging and LOAD pathophysiology. However, the exact mechanisms and causal relationships are largely unknown. In our studies we tested the hypothesis that altered bioenergetic and metabolic cell functions are key elements in LOAD, using a cellular platform consisting of skin fibroblasts derived from LOAD patients and AD-unaffected control individuals and therefrom generated induced pluripotent stem cells that are differentiated to brain-like cells to study LOAD pathogenic processes in context of age, disease, genetic background, cell development, and cell type. This model has revealed that LOAD cells exhibit a multitude of bioenergetic and metabolic alterations, providing evidence for an innate inefficient cellular energy management in LOAD as a prerequisite for the development of neurodegenerative disease with age. We propose that this cellular platform could ultimately be used as a conceptual basis for a personalized medicine tool to predict altered aging and risk for development of dementia, and to test or implement customized therapeutic or disease-preventive intervention strategies.
Collapse
Affiliation(s)
- Woo-In Ryu
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, United States.,Basic Neuroscience Division, McLean Hospital, Harvard Medical School, Belmont, MA, United States.,Program for Neuropsychiatric Research, McLean Hospital, Harvard Medical School, Belmont, MA, United States
| | - Bruce M Cohen
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, United States.,Program for Neuropsychiatric Research, McLean Hospital, Harvard Medical School, Belmont, MA, United States.,Psychotic Disorders Division, McLean Hospital, Harvard Medical School, Belmont, MA, United States
| | - Kai-C Sonntag
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, United States.,Basic Neuroscience Division, McLean Hospital, Harvard Medical School, Belmont, MA, United States.,Program for Neuropsychiatric Research, McLean Hospital, Harvard Medical School, Belmont, MA, United States
| |
Collapse
|
25
|
Amponsah AE, Guo R, Kong D, Feng B, He J, Zhang W, Liu X, Du X, Ma Z, Liu B, Ma J, Cui H. Patient-derived iPSCs, a reliable in vitro model for the investigation of Alzheimer's disease. Rev Neurosci 2021; 32:379-402. [PMID: 33550785 DOI: 10.1515/revneuro-2020-0065] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 11/07/2020] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease and a common cause of dementia among elderly individuals. The disease is characterized by progressive cognitive decline, accumulation of senile amyloid plaques and neurofibrillary tangles, oxidative stress, and inflammation. Human-derived cell models of AD are scarce, and over the years, non-human-derived models have been developed to recapitulate clinical AD, investigate the disease's pathogenesis and develop therapies for the disease. Several pharmacological compounds have been developed for AD based on findings from non-human-derived cell models; however, these pharmacological compounds have failed at different phases of clinical trials. This necessitates the application of human-derived cell models, such as induced pluripotent stem cells (iPSCs) in their optimized form in AD mechanistic studies and preclinical drug testing. This review provides an overview of AD and iPSCs. The AD-relevant phenotypes of iPSC-derived AD brain cells and the usefulness of iPSCs in AD are highlighted. Finally, the various recommendations that have been made to enhance iPSC/AD modelling are discussed.
Collapse
Affiliation(s)
- Asiamah Ernest Amponsah
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, Hebei Province050017, China.,Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, Hebei Province050017, China
| | - Ruiyun Guo
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, Hebei Province050017, China.,Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, Hebei Province050017, China
| | - Desheng Kong
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, Hebei Province050017, China.,Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, Hebei Province050017, China
| | - Baofeng Feng
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, Hebei Province050017, China.,Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, Hebei Province050017, China
| | - Jingjing He
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, Hebei Province050017, China.,Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, Hebei Province050017, China
| | - Wei Zhang
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, Hebei Province050017, China.,Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, Hebei Province050017, China
| | - Xin Liu
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, Hebei Province050017, China.,Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, Hebei Province050017, China
| | - Xiaofeng Du
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, Hebei Province050017, China.,Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, Hebei Province050017, China
| | - Zhenhuan Ma
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, Hebei Province050017, China.,Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, Hebei Province050017, China
| | - Boxin Liu
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, Hebei Province050017, China.,Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, Hebei Province050017, China
| | - Jun Ma
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, Hebei Province050017, China.,Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, Hebei Province050017, China.,Human Anatomy Department, Hebei Medical University, Shijiazhuang, Hebei Province050017, China
| | - Huixian Cui
- Hebei Medical University-National University of Ireland Galway Stem Cell Research Center, Hebei Medical University, Shijiazhuang, Hebei Province050017, China.,Hebei Research Center for Stem Cell Medical Translational Engineering, Shijiazhuang, Hebei Province050017, China.,Human Anatomy Department, Hebei Medical University, Shijiazhuang, Hebei Province050017, China
| |
Collapse
|
26
|
Blue EE, Thornton TA, Kooperberg C, Liu S, Wactawski-Wende J, Manson J, Kuller L, Hayden K, Reiner AP. Non-coding variants in MYH11, FZD3, and SORCS3 are associated with dementia in women. Alzheimers Dement 2021; 17:215-225. [PMID: 32966694 PMCID: PMC7920533 DOI: 10.1002/alz.12181] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 07/17/2020] [Accepted: 08/03/2020] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Recent studies suggest that both sex-specific genetic risk factors and those shared between dementia and stroke are involved in dementia pathogenesis. METHODS We performed both single-variant and gene-based genome-wide association studies of >11,000 whole genome sequences from the Women's Health Initiative cohort to discover loci associated with dementia, with adjustment for age, ethnicity, stroke, and venous thromboembolism status. Evidence for prior evidence of association and differential gene expression in dementia-related tissues and samples was gathered for each locus. RESULTS Our multiethnic studies identified significant associations between variants within APOE, MYH11, FZD3, SORCS3, and GOLGA8B and risk of dementia. Ten genes implicated by these loci, including MYH11, FZD3, SORCS3, and GOLGA8B, were differentially expressed in the context of Alzheimer's disease. DISCUSSION Our association of MYH11, FZD3, SORCS3, and GOLGA8B with dementia is supported by independent functional studies in human subjects, model systems, and associations with shared risk factors for stroke and dementia.
Collapse
Affiliation(s)
- Elizabeth E. Blue
- Division of Medical Genetics, University of Washington, Seattle, Washington, USA
| | - Timothy A. Thornton
- Department of Biostatistics, University of Washington, Seattle, Washington, USA
| | - Charles Kooperberg
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Simin Liu
- Department of Epidemiology, Brown University, Providence, Rhode Island, USA
- Department of Surgery, Brown University, Providence, Rhode Island, USA
- Department of Medicine, Brown University, Providence, Rhode Island, USA
| | - Jean Wactawski-Wende
- Department of Epidemiology and Environmental Health, University at Buffalo, Buffalo, New York, USA
| | - JoAnn Manson
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Lew Kuller
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Kathleen Hayden
- Department of Social Science and Health Policy, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Alexander P. Reiner
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
27
|
Weigand AJ, Thomas KR, Bangen KJ, Eglit GML, Delano-Wood L, Gilbert PE, Brickman AM, Bondi MW. APOE interacts with tau PET to influence memory independently of amyloid PET in older adults without dementia. Alzheimers Dement 2021; 17:61-69. [PMID: 32886451 DOI: 10.1002/alz.12173] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 07/11/2020] [Accepted: 07/14/2020] [Indexed: 02/03/2023]
Abstract
INTRODUCTION Apolipoprotein E (APOE) interacts with Alzheimer's disease pathology to promote disease progression. We investigated the moderating effect of APOE on independent associations of amyloid and tau positron emission tomography (PET) with cognition. METHODS For 297 nondemented older adults from the Alzheimer's Disease Neuroimaging Initiative, regression equations modeled associations between cognition and (1) cortical amyloid beta (Aβ) PET levels adjusting for tau and (2) medial temporal lobe (MTL) tau PET levels adjusting for Aβ, including interactions with APOE ε4-carrier status. RESULTS Adjusting for tau PET, Aβ was not associated with cognition and did not interact with APOE. In contrast, adjusting for Aβ PET, MTL tau was associated with all cognitive domains. Further, there was a stronger moderating effect of APOE on MTL tau and memory associations in ε4-carriers, even among Aβ-negative individuals. DISCUSSION Findings suggest that APOE may interact with tau independently of Aβ and that elevated MTL tau confers negative cognitive consequences in Aβ-negative ε4 carriers.
Collapse
Affiliation(s)
- Alexandra J Weigand
- San Diego State University/University of California, San Diego Joint Doctoral Program, San Diego
| | - Kelsey R Thomas
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA
- Department of Psychiatry, University of California, San Diego, California, USA
| | - Katherine J Bangen
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA
- Department of Psychiatry, University of California, San Diego, California, USA
| | - Graham M L Eglit
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA
| | - Lisa Delano-Wood
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA
- Department of Psychiatry, University of California, San Diego, California, USA
| | - Paul E Gilbert
- Department of Psychology, San Diego State University, California, USA
| | - Adam M Brickman
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Mark W Bondi
- Veterans Affairs San Diego Healthcare System, San Diego, California, USA
- Department of Psychiatry, University of California, San Diego, California, USA
| |
Collapse
|
28
|
Calvo-Rodriguez M, Bacskai BJ. Mitochondria and Calcium in Alzheimer's Disease: From Cell Signaling to Neuronal Cell Death. Trends Neurosci 2020; 44:136-151. [PMID: 33160650 DOI: 10.1016/j.tins.2020.10.004] [Citation(s) in RCA: 132] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 09/03/2020] [Accepted: 10/07/2020] [Indexed: 02/06/2023]
Abstract
Mitochondrial dysfunction has been implicated in the pathogenesis of almost all neurological diseases, including Alzheimer's disease (AD). Historically, a primary focus in this context has been the link between mitochondrial dynamics and amyloid β toxicity. Recent evidence suggests that dysregulation of mitochondrial calcium homeostasis is also related to tau and other risk factors in AD, although an ongoing challenge in the field is that data collected from different models or experimental settings have not always been consistent. We examine recent literature on mitochondrial dysregulation in AD, with special emphasis on mitochondrial calcium. We include data from in vitro systems, genetic animal models, and AD-derived human tissue, and discuss whether mitochondrial calcium transporters should be proposed as therapeutic candidates for the development of neuroprotective drugs against AD.
Collapse
Affiliation(s)
- Maria Calvo-Rodriguez
- Alzheimer Research Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, 114 16th Street, Charlestown, MA 02129, USA
| | - Brian J Bacskai
- Alzheimer Research Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, 114 16th Street, Charlestown, MA 02129, USA.
| |
Collapse
|