1
|
Yang B, Hu S, Jiang Y, Xu L, Shu S, Zhang H. Advancements in Single-Cell RNA Sequencing Research for Neurological Diseases. Mol Neurobiol 2024; 61:8797-8819. [PMID: 38564138 DOI: 10.1007/s12035-024-04126-3] [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: 11/29/2023] [Accepted: 03/18/2024] [Indexed: 04/04/2024]
Abstract
Neurological diseases are a major cause of the global burden of disease. Although the mechanisms of the occurrence and development of neurological diseases are not fully clear, most of them are associated with cells mediating neuroinflammation. Yet medications and other therapeutic options to improve treatment are still very limited. Single-cell RNA sequencing (scRNA-seq), as a delightfully potent breakthrough technology, not only identifies various cell types and response states but also uncovers cell-specific gene expression changes, gene regulatory networks, intercellular communication, and cellular movement trajectories, among others, in different cell types. In this review, we describe the technology of scRNA-seq in detail and discuss and summarize the application of scRNA-seq in exploring neurological diseases, elaborating the corresponding specific mechanisms of the diseases as well as providing a reliable basis for new therapeutic approaches. Finally, we affirm that scRNA-seq promotes the development of the neuroscience field and enables us to have a deeper cellular understanding of neurological diseases in the future, which provides strong support for the treatment of neurological diseases and the improvement of patients' prognosis.
Collapse
Affiliation(s)
- Bingjie Yang
- Department of Neurology, The Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Shuqi Hu
- Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Neurology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang, China
| | - Yiru Jiang
- Department of Neurology, The Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Lei Xu
- Department of Neurology, Zhejiang Rongjun Hospital, Jiaxing, Zhejiang, China
| | - Song Shu
- Department of Neurology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang, China
| | - Hao Zhang
- Department of Neurology, The Fourth Clinical School of Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
- Department of Neurology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
2
|
Wang MY, Zhou Y, Li WL, Zhu LQ, Liu D. Friend or foe: Lactate in neurodegenerative diseases. Ageing Res Rev 2024; 101:102452. [PMID: 39127445 DOI: 10.1016/j.arr.2024.102452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 08/07/2024] [Indexed: 08/12/2024]
Abstract
Lactate, a byproduct of glycolysis, was considered as a metabolic waste until identified by studies on the Warburg effect. Increasing evidence elucidates that lactate functions as energy fuel, signaling molecule, and donor for protein lactylation. Altered lactate utilization is a common metabolic feature of the onset and progression of neurodegenerative diseases, such as Alzheimer's disease, multiple sclerosis, amyotrophic lateral sclerosis, Parkinson's disease and Huntington's disease. This review offers an overview of lactate metabolism from the perspective of production, transportation and clearance, and the role of lactate in neurodegenerative progression, as well as a summary of protein lactylation and the signaling function of lactate in neurodegenerative diseases. Besides, this review delves into the dual roles of changed lactate metabolism during neurodegeneration and explores prospective therapeutic methods targeting lactate. We propose that elucidating the correlation between lactate and neurodegeneration is pivotal for exploring innovative therapeutic interventions for neurodegenerative diseases.
Collapse
Affiliation(s)
- Ming-Yu Wang
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yang Zhou
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Wen-Lian Li
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Ling-Qiang Zhu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| | - Dan Liu
- Department of Medical Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| |
Collapse
|
3
|
Choi J, Beroncal EL, Chernega T, Brooks HJ, Kennedy JL, Fisher CE, Flint AJ, Herrmann N, Lanctôt KL, Mah L, Mulsant BH, Pollock BG, Rajji TK, Andreazza AC. Exploring mitochondrial blood-based and genetic markers in older adults with mild cognitive impairment and remitted major depressive disorder. Transl Psychiatry 2024; 14:457. [PMID: 39468012 PMCID: PMC11519657 DOI: 10.1038/s41398-024-03155-9] [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: 02/26/2024] [Revised: 10/06/2024] [Accepted: 10/09/2024] [Indexed: 10/30/2024] Open
Abstract
Mild cognitive impairment (MCI) is a prodromal stage in aging to possible progression to Alzheimer's disease and related dementia (ADRD), where co-occurrence of major depressive disorder (MDD) accelerates the progression. Metabolic and mitochondrial abnormalities in ADRD and other neurodegenerative disorders have been widely suggested, while possible mitochondrial dysfunction has been associated with etiopathology of both MCI and MDD. Hence, investigation of mitochondrial markers in MCI, MDD, and presence of both conditions is warranted. In total, 332 older adult participants were included: 168 with MCI, 108 with MCI plus remitted MDD (rMDD), and 56 with rMDD but without MCI. We measured plasma circulating mitochondrial DNA (ccf-mtDNA), lactate, and extracted nuclear mitochondrial encoded (NMt) single-nucleotide variants (SNVs) (n = 312). Non-parametric statistical tests on ccf-mtDNA and lactate levels were performed on the diagnosis, clinical and cardiometabolic variables. Binary sequence kernel association test (SKAT-O) and burden test were performed on NMt-SNV, adjusted for age, race, gender, type II diabetes, and APOE genotype. Lower level of lactate was observed in MCI (KW χ2 = 14.8, P = 0.0024), more specifically, significant differences of lower plasma lactate between MCI only and rMDD, but not between MCI+rMDD and MCI were found, suggesting potential roles in MCI driving lactate lower levels. While higher levels of ccf-mtDNA were observed in APOE-ε4 carrier (χ2 = 5.04, P = 0.05). This relationship was present only in MCI (P = 0.043) and MCI+rMDD groups (P = 0.023). No significant nuclear-encoded mitochondrial gene associations were observed with MCI or MDD. The results suggest decreased level of plasma lactate in individuals with MCI and MCI+rMDD, with inverse correlation with ccf-mtDNA, in addition to effect of APOE-ε4 in further increasing ccf-mtDNA specifically in participants with cognitive impairment. These findings contribute to a deeper understanding of the mitochondrial markers in MCI and MDD, warranting further research to explore the precise roles of mitochondrial abnormalities in the development and progression of MCI.
Collapse
Affiliation(s)
- Jaehyoung Choi
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Erika L Beroncal
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Timofei Chernega
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | | | - James L Kennedy
- Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Corinne E Fisher
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Keenan Research Centre for Biomedical Research, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON, Canada
| | - Alastair J Flint
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Centre for Mental Health, University Health Network, Toronto, ON, Canada
| | - Nathan Herrmann
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Krista L Lanctôt
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Linda Mah
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Department of Psychiatry, Baycrest (LM), Rotman Research Institute, Toronto, ON, Canada
| | - Benoit H Mulsant
- Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Bruce G Pollock
- Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Tarek K Rajji
- Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Toronto Dementia Research Alliance, Toronto, ON, Canada
| | - Ana C Andreazza
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada.
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
- Mitochondrial Innovation Initiative, MITO2i University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
4
|
Rajendrakumar AL, Arbeev KG, Bagley O, Duan M, Yashin AI, Ukraintseva S. APOE4 and Infectious Diseases Jointly Contribute to Brain Glucose Hypometabolism, a Biomarker of Alzheimer's Pathology: New Findings from the ADNI. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.13.24313582. [PMID: 39314962 PMCID: PMC11419198 DOI: 10.1101/2024.09.13.24313582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Background Impaired brain glucose metabolism is a preclinical feature of neurodegenerative diseases such as Alzheimer's disease (AD). Infections may promote AD-related pathology. Therefore, we investigated the interplay between infections and APOE4, a strong genetic risk factor for AD. Methods We analyzed data on 1,509 participants in the Alzheimer's Disease Neuroimaging Initiative (ADNI) database using multivariate linear regression models. The outcomes were rank-normalized hypometabolic convergence index (HCI), statistical regions of interest (SROI) for AD, and mild cognitive impairment (MCI). Marginal mean estimates for infections, stratified by APOE4 carrier status, were then computed. Results Prior infections were associated with greater HCI [β=0.15, 95% CI: 0.03, 0.27, p=0.01]. The combined effects of infections and APOE4 carriers on HCI levels were significantly greater than either variable alone. Among APOE4 carriers, the estimated marginal mean was 0.62, rising to 0.77, with infections (p<0.001), indicating an interaction effect. Carriers with multiple infections showed greater hypometabolism (higher HCI), with an estimate of 0.44 (p=0.01) compared to 0.11 (p=0.08) for those with a single infection, revealing a dose-response relationship. The estimates for the association of infections with SROI AD and SROI MCI were β=-0.01 (p=0.02) and β=-0.01 (p=0.04), respectively. Conclusion Our findings suggest that infections and APOE4 jointly contribute to brain glucose hypometabolism and AD pathology, supporting a "multi-hit" mechanism in AD development.
Collapse
Affiliation(s)
- Aravind Lathika Rajendrakumar
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, North Carolina, United States of America
| | - Konstantin G Arbeev
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, North Carolina, United States of America
| | - Olivia Bagley
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, North Carolina, United States of America
| | - Matt Duan
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, North Carolina, United States of America
| | - Anatoliy I Yashin
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, North Carolina, United States of America
| | - Svetlana Ukraintseva
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, North Carolina, United States of America
| |
Collapse
|
5
|
Carling GK, Fan L, Foxe NR, Norman K, Wong MY, Zhu D, Corona C, Razzoli A, Yu F, Yarahmady A, Ye P, Chen H, Huang Y, Amin S, Sereda R, Lopez-Lee C, Zacharioudakis E, Chen X, Xu J, Cheng F, Gavathiotis E, Cuervo AM, Holtzman DM, Mok SA, Sinha SC, Sidoli S, Ratan RR, Luo W, Gong S, Gan L. Alzheimer's disease-linked risk alleles elevate microglial cGAS-associated senescence and neurodegeneration in a tauopathy model. Neuron 2024:S0896-6273(24)00654-8. [PMID: 39353433 DOI: 10.1016/j.neuron.2024.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 07/02/2024] [Accepted: 09/04/2024] [Indexed: 10/04/2024]
Abstract
The strongest risk factors for late-onset sporadic Alzheimer's disease (AD) include the ε4 allele of apolipoprotein E (APOE), the R47H variant of triggering receptor expressed on myeloid cells 2 (TREM2), and female sex. Here, we combine APOE4 and TREM2R47H (R47H) in female P301S tauopathy mice to identify the pathways activated when AD risk is the strongest, thereby highlighting detrimental disease mechanisms. We find that R47H induces neurodegeneration in 9- to 10-month-old female APOE4 tauopathy mice. The combination of APOE4 and R47H (APOE4-R47H) worsened hyperphosphorylated tau pathology in the frontal cortex and amplified tauopathy-induced microglial cyclic guanosine monophosphate (GMP)-AMP synthase (cGAS)-stimulator of interferon genes (STING) signaling and downstream interferon response. APOE4-R47H microglia displayed cGAS- and BAX-dependent upregulation of senescence, showing association between neurotoxic signatures and implicating mitochondrial permeabilization in pathogenesis. By uncovering pathways enhanced by the strongest AD risk factors, our study points to cGAS-STING signaling and associated microglial senescence as potential drivers of AD risk.
Collapse
Affiliation(s)
- Gillian K Carling
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA; Neuroscience Graduate Program, Weill Cornell Medicine, New York, NY 10065, USA
| | - Li Fan
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Nessa R Foxe
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA; Neuroscience Graduate Program, Weill Cornell Medicine, New York, NY 10065, USA
| | - Kendra Norman
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Man Ying Wong
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Daphne Zhu
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Carlo Corona
- Burke Neurological Institute, Weill Cornell Medicine, White Plains, NY 10605, USA
| | - Agnese Razzoli
- Transfusion Medicine Unit, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia 42122, Italy; Clinical and Experimental PhD Program, University of Modena and Reggio Emilia, Modena 41121, Italy
| | - Fangmin Yu
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Allan Yarahmady
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Pearly Ye
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Hao Chen
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Yige Huang
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA; Biochemistry, Structural Biology, Cell Biology, Developmental Biology, and Molecular Biology Graduate Program, Weill Cornell Medicine, New York, NY 10065, USA
| | - Sadaf Amin
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Rebecca Sereda
- Department of Developmental and Molecular Biology, Institute for Aging Research, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| | - Chloe Lopez-Lee
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA; Neuroscience Graduate Program, Weill Cornell Medicine, New York, NY 10065, USA
| | - Emmanouil Zacharioudakis
- Department of Biochemistry, Department of Medicine, Montefiore Einstein Comprehensive Cancer Center, Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Xiaoying Chen
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jielin Xu
- Cleveland Clinic Genome Center and Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Feixiong Cheng
- Cleveland Clinic Genome Center and Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Evripidis Gavathiotis
- Department of Biochemistry, Department of Medicine, Montefiore Einstein Comprehensive Cancer Center, Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Ana Maria Cuervo
- Department of Developmental and Molecular Biology, Institute for Aging Research, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sue-Ann Mok
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Subhash C Sinha
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Simone Sidoli
- Department of Biochemistry, Department of Medicine, Montefiore Einstein Comprehensive Cancer Center, Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Rajiv R Ratan
- Burke Neurological Institute, Weill Cornell Medicine, White Plains, NY 10605, USA
| | - Wenjie Luo
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Shiaoching Gong
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Li Gan
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA.
| |
Collapse
|
6
|
Chen F, Zhao J, Meng F, He F, Ni J, Fu Y. The vascular contribution of apolipoprotein E to Alzheimer's disease. Brain 2024; 147:2946-2965. [PMID: 38748848 DOI: 10.1093/brain/awae156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 03/23/2024] [Accepted: 04/21/2024] [Indexed: 09/04/2024] Open
Abstract
Alzheimer's disease, the most prevalent form of dementia, imposes a substantial societal burden. The persistent inadequacy of disease-modifying drugs targeting amyloid plaques and neurofibrillary tangles suggests the contribution of alternative pathogenic mechanisms. A frequently overlooked aspect is cerebrovascular dysfunction, which may manifest early in the progression of Alzheimer's disease pathology. Mounting evidence underscores the pivotal role of the apolipoprotein E gene, particularly the apolipoprotein ε4 allele as the strongest genetic risk factor for late-onset Alzheimer's disease, in the cerebrovascular pathology associated with Alzheimer's disease. In this review, we examine the evidence elucidating the cerebrovascular impact of both central and peripheral apolipoprotein E on the pathogenesis of Alzheimer's disease. We present a novel three-hit hypothesis, outlining potential mechanisms that shed light on the intricate relationship among different pathogenic events. Finally, we discuss prospective therapeutics targeting the cerebrovascular pathology associated with apolipoprotein E and explore their implications for future research endeavours.
Collapse
Affiliation(s)
- Feng Chen
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jing Zhao
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, China
| | - Fanxia Meng
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Fangping He
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jie Ni
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Yuan Fu
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| |
Collapse
|
7
|
Johnson LA, Macauley SL. Alzheimer's and metabolism wed with IDO1. Science 2024; 385:826-827. [PMID: 39172856 DOI: 10.1126/science.adr5836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2024]
Abstract
Kynurenine pathway inhibition reverses deficits in Alzheimer's mouse models.
Collapse
Affiliation(s)
- Lance A Johnson
- Department of Physiology, University of Kentucky, Lexington, KY 40508 USA
- Sanders Brown Center on Aging,University of Kentucky, Lexington, KY 40508 USA
| | - Shannon L Macauley
- Department of Physiology, University of Kentucky, Lexington, KY 40508 USA
- Sanders Brown Center on Aging,University of Kentucky, Lexington, KY 40508 USA
- Department of Neuroscience, University of Kentucky, Lexington, KY 40508 USA
| |
Collapse
|
8
|
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
|
9
|
Ebright B, Duro MV, Chen K, Louie S, Yassine HN. Effects of APOE4 on omega-3 brain metabolism across the lifespan. Trends Endocrinol Metab 2024; 35:745-757. [PMID: 38609814 PMCID: PMC11321946 DOI: 10.1016/j.tem.2024.03.003] [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: 11/18/2023] [Revised: 03/09/2024] [Accepted: 03/14/2024] [Indexed: 04/14/2024]
Abstract
Omega-3 (n-3) polyunsaturated fatty acids (PUFAs), such as docosahexaenoic acid (DHA), have important roles in human nutrition and brain health by promoting neuronal functions, maintaining inflammatory homeostasis, and providing structural integrity. As Alzheimer's disease (AD) pathology progresses, DHA metabolism in the brain becomes dysregulated, the timing and extent of which may be influenced by the apolipoprotein E ε4 (APOE4) allele. Here, we discuss how maintaining adequate DHA intake early in life may slow the progression to AD dementia in cognitively normal individuals with APOE4, how recent advances in DHA brain imaging could offer insights leading to more personalized preventive strategies, and how alternative strategies targeting PUFA metabolism pathways may be more effective in mitigating disease progression in patients with existing AD dementia.
Collapse
Affiliation(s)
- Brandon Ebright
- Department of Clinical Pharmacy, Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Marlon V Duro
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Kai Chen
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Stan Louie
- Department of Clinical Pharmacy, Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Hussein N Yassine
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| |
Collapse
|
10
|
Budny V, Knöpfli Y, Meier D, Zürcher K, Bodenmann C, Peter SL, Müller T, Tardy M, Cortijo C, Tackenberg C. APOE4 Increases Energy Metabolism in APOE-Isogenic iPSC-Derived Neurons. Cells 2024; 13:1207. [PMID: 39056789 PMCID: PMC11274733 DOI: 10.3390/cells13141207] [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: 06/11/2024] [Revised: 07/08/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
The apolipoprotein E4 (APOE4) allele represents the major genetic risk factor for Alzheimer's disease (AD). In contrast, APOE2 is known to lower the AD risk, while APOE3 is defined as risk neutral. APOE plays a prominent role in the bioenergetic homeostasis of the brain, and early-stage metabolic changes have been detected in the brains of AD patients. Although APOE is primarily expressed by astrocytes in the brain, neurons have also been shown as source for APOE. However, the distinct roles of the three APOE isoforms in neuronal energy homeostasis remain poorly understood. In this study, we generated pure human neurons (iN cells) from APOE-isogenic induced pluripotent stem cells (iPSCs), expressing either APOE2, APOE3, APOE4, or carrying an APOE knockout (KO) to investigate APOE isoform-specific effects on neuronal energy metabolism. We showed that endogenously produced APOE4 enhanced mitochondrial ATP production in APOE-isogenic iN cells but not in the corresponding iPS cell line. This effect neither correlated with the expression levels of mitochondrial fission or fusion proteins nor with the intracellular or secreted levels of APOE, which were similar for APOE2, APOE3, and APOE4 iN cells. ATP production and basal respiration in APOE-KO iN cells strongly differed from APOE4 and more closely resembled APOE2 and APOE3 iN cells, indicating a gain-of-function mechanism of APOE4 rather than a loss-of-function. Taken together, our findings in APOE isogenic iN cells reveal an APOE genotype-dependent and neuron-specific regulation of oxidative energy metabolism.
Collapse
Affiliation(s)
- Vanessa Budny
- Institute for Regenerative Medicine, University of Zurich, 8952 Schlieren, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, 8057 Zurich, Switzerland
| | - Yannic Knöpfli
- Institute for Regenerative Medicine, University of Zurich, 8952 Schlieren, Switzerland
| | - Debora Meier
- Institute for Regenerative Medicine, University of Zurich, 8952 Schlieren, Switzerland
| | - Kathrin Zürcher
- Institute for Regenerative Medicine, University of Zurich, 8952 Schlieren, Switzerland
| | - Chantal Bodenmann
- Institute for Regenerative Medicine, University of Zurich, 8952 Schlieren, Switzerland
| | - Siri L. Peter
- Institute for Regenerative Medicine, University of Zurich, 8952 Schlieren, Switzerland
| | - Terry Müller
- Neurimmune AG, Wagistrasse 18, 8952 Schlieren, Switzerland
| | - Marie Tardy
- Neurimmune AG, Wagistrasse 18, 8952 Schlieren, Switzerland
| | - Cedric Cortijo
- Neurimmune AG, Wagistrasse 18, 8952 Schlieren, Switzerland
| | - Christian Tackenberg
- Institute for Regenerative Medicine, University of Zurich, 8952 Schlieren, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, 8057 Zurich, Switzerland
| |
Collapse
|
11
|
Zhang H, Zheng Q, Guo T, Zhang S, Zheng S, Wang R, Deng Q, Yang G, Zhang S, Tang L, Qi Q, Zhu L, Zhang XF, Luo H, Zhang X, Sun H, Gao Y, Zhang H, Zhou Y, Han A, Zhang CS, Xu H, Wang X. Metabolic reprogramming in astrocytes results in neuronal dysfunction in intellectual disability. Mol Psychiatry 2024; 29:1569-1582. [PMID: 35338313 DOI: 10.1038/s41380-022-01521-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 03/03/2022] [Accepted: 03/10/2022] [Indexed: 11/08/2022]
Abstract
Astrocyte aerobic glycolysis provides vital trophic support for central nervous system neurons. However, whether and how astrocytic metabolic dysregulation contributes to neuronal dysfunction in intellectual disability (ID) remain unclear. Here, we demonstrate a causal role for an ID-associated SNX27 mutation (R198W) in cognitive deficits involving reshaping astrocytic metabolism. We generated SNX27R196W (equivalent to human R198W) knock-in mice and found that they displayed deficits in synaptic function and learning behaviors. SNX27R196W resulted in attenuated astrocytic glucose uptake via GLUT1, leading to reduced lactate production and a switch from homeostatic to reactive astrocytes. Importantly, lactate supplementation or a ketogenic diet restored neuronal oxidative phosphorylation and reversed cognitive deficits in SNX27R196W mice. In summary, we illustrate a key role for astrocytic SNX27 in maintaining glucose supply and glycolysis and reveal that altered astrocytic metabolism disrupts the astrocyte-neuron interaction, which contributes to ID. Our work also suggests a feasible strategy for treating ID by restoring astrocytic metabolic function.
Collapse
Affiliation(s)
- Haibin Zhang
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Department of Neurosurgery, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
- Shenzhen Research Institute of Xiamen University, Shenzhen, Guangdong, 518057, China
| | - Qiuyang Zheng
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Department of Neurosurgery, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
- Shenzhen Research Institute of Xiamen University, Shenzhen, Guangdong, 518057, China
| | - Tiantian Guo
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Department of Neurosurgery, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Shijun Zhang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Shuang Zheng
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Department of Neurosurgery, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Ruimin Wang
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Department of Neurosurgery, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Qingfang Deng
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Department of Neurosurgery, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Guowei Yang
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Department of Neurosurgery, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Shuo Zhang
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Department of Neurosurgery, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Linxin Tang
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Department of Neurosurgery, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Qiuping Qi
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Department of Neurosurgery, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Lin Zhu
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Department of Neurosurgery, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Xiu-Fang Zhang
- Department of Pediatrics, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, 361102, China
| | - Hong Luo
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Department of Neurosurgery, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Xian Zhang
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Department of Neurosurgery, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Hao Sun
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Department of Neurosurgery, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Yue Gao
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Department of Neurosurgery, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Hongfeng Zhang
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Department of Neurosurgery, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Ying Zhou
- Department of Translational Medicine, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Aidong Han
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Chen-Song Zhang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Huaxi Xu
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Department of Neurosurgery, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Xin Wang
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Department of Neurosurgery, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China.
- Shenzhen Research Institute of Xiamen University, Shenzhen, Guangdong, 518057, China.
| |
Collapse
|
12
|
Foley KE, Wilcock DM. Three major effects of APOE ε4 on Aβ immunotherapy induced ARIA. Front Aging Neurosci 2024; 16:1412006. [PMID: 38756535 PMCID: PMC11096466 DOI: 10.3389/fnagi.2024.1412006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 04/23/2024] [Indexed: 05/18/2024] Open
Abstract
The targeting of amyloid-beta (Aβ) plaques therapeutically as one of the primary causes of Alzheimer's disease (AD) dementia has been an ongoing effort spanning decades. While some antibodies are extremely promising and have been moved out of clinical trials and into the clinic, most of these treatments show similar adverse effects in the form of cerebrovascular damage known as amyloid-related imaging abnormalities (ARIA). The two categories of ARIA are of major concern for patients, families, and prescribing physicians, with ARIA-E presenting as cerebral edema, and ARIA-H as cerebral hemorrhages (micro- and macro-). From preclinical and clinical trials, it has been observed that the greatest genetic risk factor for AD, APOEε4, is also a major risk factor for anti-Aβ immunotherapy-induced ARIA. APOEε4 carriers represent a large population of AD patients, and, therefore, limits the broad adoption of these therapies across the AD population. In this review we detail three hypothesized mechanisms by which APOEε4 influences ARIA risk: (1) reduced cerebrovascular integrity, (2) increased neuroinflammation and immune dysregulation, and (3) elevated levels of CAA. The effects of APOEε4 on ARIA risk is clear, however, the underlying mechanisms require more research.
Collapse
Affiliation(s)
- Kate E. Foley
- Stark Neurosciences Research Institute, Indiana University, Indianapolis, IN, United States
- Department of Neurology, School of Medicine, Indiana University, Indianapolis, IN, United States
| | - Donna M. Wilcock
- Stark Neurosciences Research Institute, Indiana University, Indianapolis, IN, United States
- Department of Neurology, School of Medicine, Indiana University, Indianapolis, IN, United States
| |
Collapse
|
13
|
Wu L, Guo X, Gao Y, Yu W, Qin W, Kuang H, Su Y. Untargeted metabolomics reveals intervention effects of wine-processed Schisandra chinensis polysaccharide on Alzheimer's disease mice. Int J Biol Macromol 2024; 267:130804. [PMID: 38565361 DOI: 10.1016/j.ijbiomac.2024.130804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 03/02/2024] [Accepted: 03/10/2024] [Indexed: 04/04/2024]
Abstract
Schisandra chinensis (Turcz.) Baill (SC) is a traditional sedative in China, with wide applications for treating various neurological disorders. Its polysaccharide component has been gaining increased attention for its potential in nerve protection. While raw SC is the primary focus of current research, its processed products are primarily utilized as clinical medicines. Notably, limited research exists on the mechanisms underlying the effects of wine-processed Schisandra chinensis polysaccharide (WSCP) in Alzheimer's Disease (AD). Therefore, this study seeks to assess the therapeutic impact of WSCP on AD mice and investigate the underlying mechanisms through biochemical and metabolomics analyses. The results demonstrate that WSCP exerts significant therapeutic effects on AD mice by enhancing learning and memory abilities, mitigating hippocampal neuronal damage, reducing abnormal amyloid-beta (Aβ) deposition, and attenuating hyperphosphorylation of Tau. Biochemical analysis revealed that WSCP can increase SOD content and decrease MDA, IL-6, and TNF-α content in AD mice. Furthermore, serum metabolomic results showed that WSCP intervention can reverse metabolic disorders in AD mice. 43 endogenous metabolites were identified as potential biomarkers for WSCP treatment of AD, and the major metabolic pathways were Ala, Glu and Asp metabolism, TCA cycle. Overall, these findings will provide a basis for further development of WSCP.
Collapse
Affiliation(s)
- Lun Wu
- Institute of Traditional Chinese Medicine, Heilongjiang University Of Chinese Medicine, Heilongjiang 150040, China
| | - Xingyu Guo
- School of Pharmacy, Heilongjiang University Of Chinese Medicine, Heilongjiang 150040, China
| | - Yue Gao
- School of Pharmacy, Heilongjiang University Of Chinese Medicine, Heilongjiang 150040, China
| | - Wenting Yu
- School of Pharmacy, Heilongjiang University Of Chinese Medicine, Heilongjiang 150040, China
| | - Wen Qin
- School of Pharmacy, Heilongjiang University Of Chinese Medicine, Heilongjiang 150040, China
| | - Haixue Kuang
- School of Pharmacy, Heilongjiang University Of Chinese Medicine, Heilongjiang 150040, China
| | - Yang Su
- School of Pharmacy, Heilongjiang University Of Chinese Medicine, Heilongjiang 150040, China.
| |
Collapse
|
14
|
Zhang X, Chen C, Liu Y. Navigating the metabolic maze: anomalies in fatty acid and cholesterol processes in Alzheimer's astrocytes. Alzheimers Res Ther 2024; 16:63. [PMID: 38521950 PMCID: PMC10960454 DOI: 10.1186/s13195-024-01430-x] [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: 11/27/2023] [Accepted: 03/13/2024] [Indexed: 03/25/2024]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia, and its underlying mechanisms have been a subject of great interest. The mainstream theory of AD pathology suggests that the disease is primarily associated with tau protein and amyloid-beta (Aβ). However, an increasing body of research has revealed that abnormalities in lipid metabolism may be an important event throughout the pathophysiology of AD. Astrocytes, as important members of the lipid metabolism network in the brain, play a significant role in this event. The study of abnormal lipid metabolism in astrocytes provides a new perspective for understanding the pathogenesis of AD. This review focuses on the abnormal metabolism of fatty acids (FAs) and cholesterol in astrocytes in AD, and discusses it from three perspectives: lipid uptake, intracellular breakdown or synthesis metabolism, and efflux transport. We found that, despite the accumulation of their own fatty acids, astrocytes cannot efficiently uptake fatty acids from neurons, leading to fatty acid accumulation within neurons and resulting in lipotoxicity. In terms of cholesterol metabolism, astrocytes exhibit a decrease in endogenous synthesis due to the accumulation of exogenous cholesterol. Through a thorough investigation of these metabolic abnormalities, we can provide new insights for future therapeutic strategies by literature review to navigate this complex metabolic maze and bring hope to patients with Alzheimer's disease.
Collapse
Affiliation(s)
- Xiaoyu Zhang
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Chuanying Chen
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
- School of Traditional Chinese Medicine, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Yi Liu
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| |
Collapse
|
15
|
Reyes-Reyes EM, Brown J, Trial MD, Chinnasamy D, Wiegand JP, Bradford D, Brinton RD, Rodgers KE. Vivaria housing conditions expose sex differences in brain oxidation, microglial activation, and immune system states in aged hAPOE4 mice. Exp Brain Res 2024; 242:543-557. [PMID: 38206365 PMCID: PMC10894770 DOI: 10.1007/s00221-023-06763-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 12/04/2023] [Indexed: 01/12/2024]
Abstract
Apolipoprotein E ε4 allele (APOE4) is the predominant genetic risk factor for late-onset Alzheimer's disease (AD). APOE4 mouse models have provided advances in the understanding of disease pathogenesis, but unaccounted variables like rodent housing status may hinder translational outcomes. Non-sterile aspects like food and bedding can be major sources of changes in rodent microflora. Alterations in intestinal microbial ecology can cause mucosal barrier impairment and increase pro-inflammatory signals. The present study examined the role of sterile and non-sterile food and housing on redox indicators and the immune status of humanized-APOE4 knock-in mice (hAPOe4). hAPOE4 mice were housed under sterile conditions until 22 months of age, followed by the transfer of a cohort of mice to non-sterile housing for 2 months. At 24 months of age, the redox/immunologic status was evaluated by flow cytometry/ELISA. hAPOE4 females housed under non-sterile conditions exhibited: (1) higher neuronal and microglial oxygen radical production and (2) lower CD68+ microglia (brain) and CD8+ T cells (periphery) compared to sterile-housed mice. In contrast, hAPOE4 males in non-sterile housing exhibited: (1) higher MHCII+ microglia and CD11b+CD4+ T cells (brain) and (2) higher CD11b+CD4+ T cells and levels of lipopolysaccharide-binding protein and inflammatory cytokines in the periphery relative to sterile-housed mice. This study demonstrated that sterile vs. non-sterile housing conditions are associated with the activation of redox and immune responses in the brain and periphery in a sex-dependent manner. Therefore, housing status may contribute to variable outcomes in both the brain and periphery.
Collapse
Affiliation(s)
- E M Reyes-Reyes
- Center for Innovation in Brain Science, University of Arizona, 1230 N. Cherry Ave, PO Box 210242, Tucson, AZ, 85721-0242, USA
| | - J Brown
- Center for Innovation in Brain Science, University of Arizona, 1230 N. Cherry Ave, PO Box 210242, Tucson, AZ, 85721-0242, USA
| | - M D Trial
- Center for Innovation in Brain Science, University of Arizona, 1230 N. Cherry Ave, PO Box 210242, Tucson, AZ, 85721-0242, USA
| | - D Chinnasamy
- Center for Innovation in Brain Science, University of Arizona, 1230 N. Cherry Ave, PO Box 210242, Tucson, AZ, 85721-0242, USA
| | - J P Wiegand
- Center for Innovation in Brain Science, University of Arizona, 1230 N. Cherry Ave, PO Box 210242, Tucson, AZ, 85721-0242, USA
| | - D Bradford
- Center for Innovation in Brain Science, University of Arizona, 1230 N. Cherry Ave, PO Box 210242, Tucson, AZ, 85721-0242, USA
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - R D Brinton
- Center for Innovation in Brain Science, University of Arizona, 1230 N. Cherry Ave, PO Box 210242, Tucson, AZ, 85721-0242, USA
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - K E Rodgers
- Center for Innovation in Brain Science, University of Arizona, 1230 N. Cherry Ave, PO Box 210242, Tucson, AZ, 85721-0242, USA.
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
16
|
Constantino NJ, Carroll CM, Williams HC, Yuede CM, Sheehan PW, Andy Snipes J, Musiek ES, Johnson LA, Macauley SL. Kir6.2-K ATP channels alter glycolytic flux to modulate cortical activity, arousal, and sleep-wake homeostasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.23.581817. [PMID: 38464274 PMCID: PMC10925108 DOI: 10.1101/2024.02.23.581817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Metabolism plays an important role in the maintenance of vigilance states (e.g. wake, NREM, and REM). Brain lactate fluctuations are a biomarker of sleep. Increased interstitial fluid (ISF) lactate levels are necessary for arousal and wake-associated behaviors, while decreased ISF lactate is required for sleep. ATP-sensitive potassium (K ATP ) channels couple glucose-lactate metabolism with neuronal excitability. Therefore, we explored how deletion of neuronal K ATP channel activity (Kir6.2-/- mice) affected the relationship between glycolytic flux, neuronal activity, and sleep/wake homeostasis. Kir6.2-/- mice shunt glucose towards glycolysis, reduce neurotransmitter synthesis, dampen cortical EEG activity, and decrease arousal. Kir6.2-/- mice spent more time awake at the onset of the light period due to altered ISF lactate dynamics. Together, we show that Kir6.2-K ATP channels act as metabolic sensors to gate arousal by maintaining the metabolic stability of each vigilance state and providing the metabolic flexibility to transition between states. Highlights Glycolytic flux is necessary for neurotransmitter synthesis. In its absence, neuronal activity is compromised causing changes in arousal and vigilance states despite sufficient energy availability. With Kir6.2-K ATP channel deficiency, the ability to both maintain and shift between different vigilance states is compromised due to changes in glucose utilization. Kir6.2-K ATP channels are metabolic sensors under circadian control that gate arousal and sleep/wake transitions.
Collapse
|
17
|
Blumenfeld J, Yip O, Kim MJ, Huang Y. Cell type-specific roles of APOE4 in Alzheimer disease. Nat Rev Neurosci 2024; 25:91-110. [PMID: 38191720 PMCID: PMC11073858 DOI: 10.1038/s41583-023-00776-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/28/2023] [Indexed: 01/10/2024]
Abstract
The ɛ4 allele of the apolipoprotein E gene (APOE), which translates to the APOE4 isoform, is the strongest genetic risk factor for late-onset Alzheimer disease (AD). Within the CNS, APOE is produced by a variety of cell types under different conditions, posing a challenge for studying its roles in AD pathogenesis. However, through powerful advances in research tools and the use of novel cell culture and animal models, researchers have recently begun to study the roles of APOE4 in AD in a cell type-specific manner and at a deeper and more mechanistic level than ever before. In particular, cutting-edge omics studies have enabled APOE4 to be studied at the single-cell level and have allowed the identification of critical APOE4 effects in AD-vulnerable cellular subtypes. Through these studies, it has become evident that APOE4 produced in various types of CNS cell - including astrocytes, neurons, microglia, oligodendrocytes and vascular cells - has diverse roles in AD pathogenesis. Here, we review these scientific advances and propose a cell type-specific APOE4 cascade model of AD. In this model, neuronal APOE4 emerges as a crucial pathological initiator and driver of AD pathogenesis, instigating glial responses and, ultimately, neurodegeneration. In addition, we provide perspectives on future directions for APOE4 research and related therapeutic developments in the context of AD.
Collapse
Affiliation(s)
- Jessica Blumenfeld
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Oscar Yip
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Min Joo Kim
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Yadong Huang
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA.
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA.
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA.
- Gladstone Center for Translational Advancement, Gladstone Institutes, San Francisco, CA, USA.
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA.
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
18
|
Carling GK, Fan L, Foxe NR, Norman K, Ye P, Wong MY, Zhu D, Yu F, Xu J, Yarahmady A, Chen H, Huang Y, Amin S, Zacharioudakis E, Chen X, Holtzman DM, Mok SA, Gavathiotis E, Sinha SC, Cheng F, Luo W, Gong S, Gan L. Alzheimer's disease-linked risk alleles elevate microglial cGAS-associated senescence and neurodegeneration in a tauopathy model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.24.577107. [PMID: 38328219 PMCID: PMC10849737 DOI: 10.1101/2024.01.24.577107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
The strongest risk factors for Alzheimer's disease (AD) include the χ4 allele of apolipoprotein E (APOE), the R47H variant of triggering receptor expressed on myeloid cells 2 (TREM2), and female sex. Here, we combine APOE4 and TREM2R47H ( R47H ) in female P301S tauopathy mice to identify the pathways activated when AD risk is the strongest, thereby highlighting disease-causing mechanisms. We find that the R47H variant induces neurodegeneration in female APOE4 mice without impacting hippocampal tau load. The combination of APOE4 and R47H amplified tauopathy-induced cell-autonomous microglial cGAS-STING signaling and type-I interferon response, and interferon signaling converged across glial cell types in the hippocampus. APOE4-R47H microglia displayed cGAS- and BAX-dependent upregulation of senescence, showing association between neurotoxic signatures and implicating mitochondrial permeabilization in pathogenesis. By uncovering pathways enhanced by the strongest AD risk factors, our study points to cGAS-STING signaling and associated microglial senescence as potential drivers of AD risk.
Collapse
|
19
|
Han Y, Huang C, Pan Y, Gu X. Single Cell Sequencing Technology and Its Application in Alzheimer's Disease. J Alzheimers Dis 2024; 97:1033-1050. [PMID: 38217599 DOI: 10.3233/jad-230861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2024]
Abstract
Alzheimer's disease (AD) involves degeneration of cells in the brain. Due to insidious onset and slow progression, AD is often not diagnosed until it gets progressed to a more severe stage. The diagnosis and treatment of AD has been a challenge. In recent years, high-throughput sequencing technologies have exhibited advantages in exploring the pathogenesis of diseases. However, the types of cells of the central nervous system are complex and traditional bulk sequencing cannot reflect their heterogeneity. Single-cell sequencing technology enables study at the individual cell level and has an irreplaceable advantage in the study of complex diseases. In recent years, this field has expanded rapidly and several types of single-cell sequencing technologies have emerged, including transcriptomics, epigenomics, genomics and proteomics. This review article provides an overview of these single-cell sequencing technologies and their application in AD.
Collapse
Affiliation(s)
- Yuru Han
- Shanghai Key Laboratory of Molecular Imaging, Zhoupu Hospital, Shanghai University of Medicine and Health Sciences, Shanghai, China
- School of Pharmacy, Shanghai University of Medicine & Health Sciences, Shanghai, China
- School of Health Sciences and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Congying Huang
- Shanghai Key Laboratory of Molecular Imaging, Zhoupu Hospital, Shanghai University of Medicine and Health Sciences, Shanghai, China
- School of Pharmacy, Shanghai University of Medicine & Health Sciences, Shanghai, China
- School of Health Sciences and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Yuhui Pan
- Center for Disease Control and Prevention of Harbin, Harbin, China
| | - Xuefeng Gu
- Shanghai Key Laboratory of Molecular Imaging, Zhoupu Hospital, Shanghai University of Medicine and Health Sciences, Shanghai, China
- School of Pharmacy, Shanghai University of Medicine & Health Sciences, Shanghai, China
- School of Health Sciences and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| |
Collapse
|
20
|
Burke BI, Goh J, Albathi FA, Valentino TR, Nolt GL, Joshi JK, Dungan CM, Johnson LA, Wen Y, Ismaeel A, McCarthy JJ. ApoE isoform does not influence skeletal muscle regeneration in adult mice. Front Physiol 2023; 14:1302695. [PMID: 38074327 PMCID: PMC10702509 DOI: 10.3389/fphys.2023.1302695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 11/10/2023] [Indexed: 02/12/2024] Open
Abstract
Introduction: Apolipoprotein E (ApoE) has been shown to be necessary for proper skeletal muscle regeneration. Consistent with this finding, single-cell RNA-sequencing analyses of skeletal muscle stem cells (MuSCs) revealed that Apoe is a top marker of quiescent MuSCs that is downregulated upon activation. The purpose of this study was to determine if muscle regeneration is altered in mice which harbor one of the three common human ApoE isoforms, referred to as ApoE2, E3 and E4. Methods: Histomorphometric analyses were employed to assess muscle regeneration in ApoE2, E3, and E4 mice after 14 days of recovery from barium chloride-induced muscle damage in vivo, and primary MuSCs were isolated to assess proliferation and differentiation of ApoE2, E3, and E4 MuSCs in vitro. Results: There was no difference in the basal skeletal muscle phenotype of ApoE isoforms as evaluated by section area, myofiber cross-sectional area (CSA), and myonuclear and MuSC abundance per fiber. Although there were no differences in fiber-type frequency in the soleus, Type IIa relative frequency was significantly lower in plantaris muscles of ApoE4 mice compared to ApoE3. Moreover, ApoE isoform did not influence muscle regeneration as assessed by fiber frequency, fiber CSA, and myonuclear and MuSC abundance. Finally, there were no differences in the proliferative capacity or myogenic differentiation potential of MuSCs between any ApoE isoform. Discussion: Collectively, these data indicate nominal effects of ApoE isoform on the ability of skeletal muscle to regenerate following injury or the in vitro MuSC phenotype.
Collapse
Affiliation(s)
- Benjamin I. Burke
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, United States
- Center for Muscle Biology, University of Kentucky, Lexington, KY, United States
| | - Jensen Goh
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, United States
- Center for Muscle Biology, University of Kentucky, Lexington, KY, United States
| | - Fatmah A. Albathi
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, United States
- Center for Muscle Biology, University of Kentucky, Lexington, KY, United States
| | | | - Georgia L. Nolt
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, United States
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States
| | - Jai K. Joshi
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, United States
- Center for Muscle Biology, University of Kentucky, Lexington, KY, United States
| | - Cory M. Dungan
- Department of Health, Human Performance, and Recreation, Robbins College of Health and Human Sciences, Baylor University, Waco, TX, United States
| | - Lance A. Johnson
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, United States
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States
| | - Yuan Wen
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, United States
- Center for Muscle Biology, University of Kentucky, Lexington, KY, United States
- Division of Biomedical Informatics, Department of Internal Medicine, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Ahmed Ismaeel
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, United States
- Center for Muscle Biology, University of Kentucky, Lexington, KY, United States
| | - John J. McCarthy
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, United States
- Center for Muscle Biology, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
21
|
Lee H, Cho S, Kim MJ, Park YJ, Cho E, Jo YS, Kim YS, Lee JY, Thoudam T, Woo SH, Lee SI, Jeon J, Lee YS, Suh BC, Yoon JH, Go Y, Lee IK, Seo J. ApoE4-dependent lysosomal cholesterol accumulation impairs mitochondrial homeostasis and oxidative phosphorylation in human astrocytes. Cell Rep 2023; 42:113183. [PMID: 37777962 DOI: 10.1016/j.celrep.2023.113183] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 08/07/2023] [Accepted: 09/13/2023] [Indexed: 10/03/2023] Open
Abstract
Recent developments in genome sequencing have expanded the knowledge of genetic factors associated with late-onset Alzheimer's disease (AD). Among them, genetic variant ε4 of the APOE gene (APOE4) confers the greatest disease risk. Dysregulated glucose metabolism is an early pathological feature of AD. Using isogenic ApoE3 and ApoE4 astrocytes derived from human induced pluripotent stem cells, we find that ApoE4 increases glycolytic activity but impairs mitochondrial respiration in astrocytes. Ultrastructural and autophagy flux analyses show that ApoE4-induced cholesterol accumulation impairs lysosome-dependent removal of damaged mitochondria. Acute treatment with cholesterol-depleting agents restores autophagic activity, mitochondrial dynamics, and associated proteomes, and extended treatment rescues mitochondrial respiration in ApoE4 astrocytes. Taken together, our study provides a direct link between ApoE4-induced lysosomal cholesterol accumulation and abnormal oxidative phosphorylation.
Collapse
Affiliation(s)
- Hyein Lee
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science & Technology, Daegu 42988, South Korea
| | - Sukhee Cho
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science & Technology, Daegu 42988, South Korea
| | - Mi-Jin Kim
- Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu 41944, South Korea
| | - Yeo Jin Park
- Korean Medicine Life Science, University of Science and Technology, Daejeon 34054, South Korea; Korean Medicine-Application Center, Korea Institute of Oriental Medicine, Daegu 41062, South Korea
| | - Eunji Cho
- Neurodegenerative Disease Research Group, Korea Brain Research Institute, Daegu 41062, South Korea
| | - Yeon Suk Jo
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science & Technology, Daegu 42988, South Korea; Neurodegenerative Disease Research Group, Korea Brain Research Institute, Daegu 41062, South Korea
| | - Yong-Seok Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science & Technology, Daegu 42988, South Korea
| | - Jung Yi Lee
- Leading-Edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University School of Medicine, Daegu 41944, South Korea
| | - Themis Thoudam
- Research Institute of Aging and Metabolism, Kyungpook National University School of Medicine, Daegu 41944, South Korea
| | - Seung-Hwa Woo
- Department of New Biology, Daegu Gyeongbuk Institute of Science & Technology, Daegu 42988, South Korea
| | - Se-In Lee
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science & Technology, Daegu 42988, South Korea
| | - Juyeong Jeon
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science & Technology, Daegu 42988, South Korea
| | - Young-Sam Lee
- Department of New Biology, Daegu Gyeongbuk Institute of Science & Technology, Daegu 42988, South Korea
| | - Byung-Chang Suh
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science & Technology, Daegu 42988, South Korea
| | - Jong Hyuk Yoon
- Neurodegenerative Disease Research Group, Korea Brain Research Institute, Daegu 41062, South Korea
| | - Younghoon Go
- Korean Medicine-Application Center, Korea Institute of Oriental Medicine, Daegu 41062, South Korea.
| | - In-Kyu Lee
- Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu 41944, South Korea; Leading-Edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University School of Medicine, Daegu 41944, South Korea; Research Institute of Aging and Metabolism, Kyungpook National University School of Medicine, Daegu 41944, South Korea.
| | - Jinsoo Seo
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science & Technology, Daegu 42988, South Korea.
| |
Collapse
|
22
|
Sun YY, Wang Z, Huang HC. Roles of ApoE4 on the Pathogenesis in Alzheimer's Disease and the Potential Therapeutic Approaches. Cell Mol Neurobiol 2023; 43:3115-3136. [PMID: 37227619 PMCID: PMC10211310 DOI: 10.1007/s10571-023-01365-1] [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: 02/14/2023] [Accepted: 05/17/2023] [Indexed: 05/26/2023]
Abstract
The Apolipoprotein E ε4 (ApoE ε4) allele, encoding ApoE4, is the strongest genetic risk factor for late-onset Alzheimer's disease (LOAD). Emerging epidemiological evidence indicated that ApoE4 contributes to AD through influencing β-amyloid (Aβ) deposition and clearance. However, the molecular mechanisms of ApoE4 involved in AD pathogenesis remains unclear. Here, we introduced the structure and functions of ApoE isoforms, and then we reviewed the potential mechanisms of ApoE4 in the AD pathogenesis, including the effect of ApoE4 on Aβ pathology, and tau phosphorylation, oxidative stress; synaptic function, cholesterol transport, and mitochondrial dysfunction; sleep disturbances and cerebrovascular integrity in the AD brains. Furthermore, we discussed the available strategies for AD treatments that target to ApoE4. In general, this review overviews the potential roles of ApoE4 in the AD development and suggests some therapeutic approaches for AD. ApoE4 is genetic risk of AD. ApoE4 is involved in the AD pathogenesis. Aβ deposition, NFT, oxidative stress, abnormal cholesterol, mitochondrial dysfunction and neuroinflammation could be observed in the brains with ApoE4. Targeting the interaction of ApoE4 with the AD pathology is available strategy for AD treatments.
Collapse
Affiliation(s)
- Yu-Ying Sun
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, 100191 China
- Key Laboratory of Natural Products Development and Innovative Drug Research, Beijing Union University, Beijing, 100023 China
| | - Zhun Wang
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, 100191 China
- Key Laboratory of Natural Products Development and Innovative Drug Research, Beijing Union University, Beijing, 100023 China
| | - Han-Chang Huang
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, 100191 China
- Key Laboratory of Natural Products Development and Innovative Drug Research, Beijing Union University, Beijing, 100023 China
| |
Collapse
|
23
|
Litke R, Vicari J, Huang BT, Shapiro L, Roh KH, Silver A, Talreja P, Palacios N, Yoon Y, Kellner C, Kaniskan H, Vangeti S, Jin J, Ramos-Lopez I, Mobbs C. Novel small molecules inhibit proteotoxicity and inflammation: Mechanistic and therapeutic implications for Alzheimer's Disease, healthspan and lifespan- Aging as a consequence of glycolysis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.12.544352. [PMID: 37398396 PMCID: PMC10312632 DOI: 10.1101/2023.06.12.544352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Inflammation drives many age-related, especially neurological, diseases, and likely mediates age-related proteotoxicity. For example, dementia due to Alzheimer's Disease (AD), cerebral vascular disease, many other neurodegenerative conditions is increasingly among the most devastating burdens on the American (and world) health system and threatens to bankrupt the American health system as the population ages unless effective treatments are developed. Dementia due to either AD or cerebral vascular disease, and plausibly many other neurodegenerative and even psychiatric conditions, is driven by increased age-related inflammation, which in turn appears to mediate Abeta and related proteotoxic processes. The functional significance of inflammation during aging is also supported by the fact that Humira, which is simply an antibody to the pro-inflammatory cytokine TNF-a, is the best-selling drug in the world by revenue. These observations led us to develop parallel high-throughput screens to discover small molecules which inhibit age-related Abeta proteotoxicity in a C. elegans model of AD AND LPS-induced microglial TNF-a. In the initial screen of 2560 compounds (Microsource Spectrum library) to delay Abeta proteotoxicity, the most protective compounds were, in order, phenylbutyrate, methicillin, and quetiapine, which belong to drug classes (HDAC inhibitors, beta lactam antibiotics, and tricyclic antipsychotics, respectably) already robustly implicated as promising to protect in neurodegenerative diseases, especially AD. RNAi and chemical screens indicated that the protective effects of HDAC inhibitors to reduce Abeta proteotoxicity are mediated by inhibition of HDAC2, also implicated in human AD, dependent on the HAT Creb binding protein (Cbp), which is also required for the protective effects of both dietary restriction and the daf-2 mutation (inactivation of IGF-1 signaling) during aging. In addition to methicillin, several other beta lactam antibiotics also delayed Abeta proteotoxicity and reduced microglial TNF-a. In addition to quetiapine, several other tricyclic antipsychotic drugs also delayed age-related Abeta proteotoxicity and increased microglial TNF-a, leading to the synthesis of a novel congener, GM310, which delays Abeta as well as Huntingtin proteotoxicity, inhibits LPS-induced mouse and human microglial and monocyte TNF-a, is highly concentrated in brain after oral delivery with no apparent toxicity, increases lifespan, and produces molecular responses highly similar to those produced by dietary restriction, including induction of Cbp inhibition of inhibitors of Cbp, and genes promoting a shift away from glycolysis and toward metabolism of alternate (e.g., lipid) substrates. GM310, as well as FDA-approved tricyclic congeners, prevented functional impairments and associated increase in TNF-a in a mouse model of stroke. Robust reduction of glycolysis by GM310 was functionally corroborated by flux analysis, and the glycolytic inhibitor 2-DG inhibited microglial TNF-a and other markers of inflammation, delayed Abeta proteotoxicity, and increased lifespan. These results support the value of phenotypic screens to discover drugs to treat age-related, especially neurological and even psychiatric diseases, including AD and stroke, and to clarify novel mechanisms driving neurodegeneration (e.g., increased microglial glycolysis drives neuroinflammation and subsequent neurotoxicity) suggesting novel treatments (selective inhibitors of microglial glycolysis).
Collapse
|
24
|
Cho J, Alexander KL, Ferrell JL, Johnson LA, Estus S, D’Orazio SEF. Apolipoprotein E genotype affects innate susceptibility to Listeria monocytogenes infection in aged male mice. Infect Immun 2023; 91:e0025123. [PMID: 37594272 PMCID: PMC10501219 DOI: 10.1128/iai.00251-23] [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: 06/29/2023] [Accepted: 06/29/2023] [Indexed: 08/19/2023] Open
Abstract
Apolipoprotein E (ApoE) is a lipid transport protein that is hypothesized to suppress proinflammatory cytokine production, particularly after stimulation with Toll-like receptor (TLR) ligands such as lipopolysaccharide (LPS). Studies using transgenic ApoE human replacement mice (APOE) expressing one of three different allelic variants suggest that there is a hierarchy in terms of responsiveness to proinflammatory stimuli such as APOE4/E4 > APOE3/E3 > APOE2/E2. In this study, we test the hypothesis that APOE genotype can also predict susceptibility to infection with the facultative intracellular gram-positive bacterium Listeria monocytogenes. We found that bone-marrow-derived macrophages isolated from aged APOE4/E4 mice expressed elevated levels of nitric oxide synthase 2 and were highly resistant to in vitro infection with L. monocytogenes compared to APOE3/E3 and APOE2/E2 mice. However, we did not find statistically significant differences in cytokine or chemokine output from either macrophages or whole splenocytes isolated from APOE2/E2, APOE3/E3, or APOE4/E4 mice following L. monocytogenes infection. In vivo, overall susceptibility to foodborne listeriosis also did not differ by APOE genotype in either young (2 mo old) or aged (15 mo old) C57BL/6 mice. However, we observed a sex-dependent susceptibility to infection in aged APOE2/E2 male mice and a sex-dependent resistance to infection in aged APOE4/E4 male mice that was not present in female mice. Thus, these results suggest that APOE genotype does not play an important role in innate resistance to infection with L. monocytogenes but may be linked to sex-dependent changes that occur during immune senescence.
Collapse
Affiliation(s)
- Jooyoung Cho
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Katie L. Alexander
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Jessica L. Ferrell
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Lance A. Johnson
- Department of Physiology, University of Kentucky College of Medicine, Lexington, Kentucky, USA
- Sanders Brown Center on Aging, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Steven Estus
- Department of Physiology, University of Kentucky College of Medicine, Lexington, Kentucky, USA
- Sanders Brown Center on Aging, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Sarah E. F. D’Orazio
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| |
Collapse
|
25
|
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
|
26
|
Abyadeh M, Gupta V, Paulo JA, Sheriff S, Shadfar S, Fitzhenry M, Amirkhani A, Gupta V, Salekdeh GH, Haynes PA, Graham SL, Mirzaei M. Apolipoprotein ε in Brain and Retinal Neurodegenerative Diseases. Aging Dis 2023; 14:1311-1330. [PMID: 37199411 PMCID: PMC10389820 DOI: 10.14336/ad.2023.0312-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 03/12/2023] [Indexed: 05/19/2023] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia that remains incurable and has become a major medical, social, and economic challenge worldwide. AD is characterized by pathological hallmarks of senile plaques (SP) and neurofibrillary tangles (NFTs) that damage the brain up to twenty years before a clinical diagnosis is made. Interestingly these pathological features have also been observed in retinal neurodegenerative diseases including age related macular degeneration (ARMD), glaucoma and diabetic retinopathy (DR). An association of AD with these diseases has been suggested in epidemiological studies and several common pathological events and risk factors have been identified between these diseases. The E4 allele of Apolipoprotein E (APOE) is a well-established genetic risk factor for late onset AD. The ApoE ε4 allele is also associated with retinal neurodegenerative diseases however in contrast to AD, it is considered protective in AMD, likewise ApoE E2 allele, which is a protective factor for AD, has been implicated as a risk factor for AMD and glaucoma. This review summarizes the evidence on the effects of ApoE in retinal neurodegenerative diseases and discusses the overlapping molecular pathways in AD. The involvement of ApoE in regulating amyloid beta (Aβ) and tau pathology, inflammation, vascular integrity, glucose metabolism and vascular endothelial growth factor (VEGF) signaling is also discussed.
Collapse
Affiliation(s)
| | - Vivek Gupta
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW 2109, Australia.
| | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA.
| | - Samran Sheriff
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW 2109, Australia.
| | - Sina Shadfar
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW 2109, Australia.
| | - Matthew Fitzhenry
- Australian Proteome Analysis Facility, Macquarie University, Macquarie Park, NSW 2113, Australia.
| | - Ardeshir Amirkhani
- Australian Proteome Analysis Facility, Macquarie University, Macquarie Park, NSW 2113, Australia.
| | - Veer Gupta
- School of Medicine, Deakin University, VIC, Australia.
| | - Ghasem H Salekdeh
- School of Natural Sciences, Macquarie University, Macquarie Park, NSW 2109, Australia.
| | - Paul A Haynes
- School of Natural Sciences, Macquarie University, Macquarie Park, NSW 2109, Australia.
| | - Stuart L Graham
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW 2109, Australia.
| | - Mehdi Mirzaei
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Macquarie Park, North Ryde, Sydney, NSW 2109, Australia.
| |
Collapse
|
27
|
Fleeman RM, Kuhn MK, Chan DC, Proctor EA. Apolipoprotein E ε4 modulates astrocyte neuronal support functions in the presence of amyloid-β. J Neurochem 2023; 165:536-549. [PMID: 36762973 PMCID: PMC10903110 DOI: 10.1111/jnc.15781] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 01/16/2023] [Accepted: 02/01/2023] [Indexed: 02/11/2023]
Abstract
Apolipoprotein E (APOE) is a lipid transporter produced predominantly by astrocytes in the brain. The ε4 variant of APOE (APOE4) is the strongest and most common genetic risk factor for Alzheimer's disease (AD). Although the molecular mechanisms of this increased risk are unclear, APOE4 is known to alter immune signaling and lipid and glucose metabolism. Astrocytes provide various forms of support to neurons, including regulating neuronal metabolism and immune responses through cytokine signaling. Changes in astrocyte function because of APOE4 may therefore decrease neuronal support, leaving neurons more vulnerable to stress and disease insults. To determine whether APOE4 alters astrocyte neuronal support functions, we measured glycolytic and oxidative metabolism of neurons treated with conditioned media from APOE4 or APOE3 (the common, risk-neutral variant) primary astrocyte cultures. We found that APOE4 neurons treated with conditioned media from resting APOE4 astrocytes had similar metabolism to APOE3 neurons treated with media from resting APOE3 astrocytes, but treatment with astrocytic conditioned media from astrocytes challenged with amyloid-β (Aβ), a key pathological protein in AD, caused APOE4 neurons to increase their basal mitochondrial and glycolytic metabolic rates more than APOE3 neurons. These changes were not because of differences in astrocytic lactate production or glucose utilization, but instead correlated with increased glycolytic ATP production and a lack of cytokine secretion in response to Aβ. Additionally, we identified that astrocytic cytokine signatures could predict basal metabolism of neurons treated with the astrocytic conditioned media. Together, these findings suggest that in the presence of Aβ, APOE4 astrocytes alter immune and metabolic functions that result in a compensatory increase in neuronal metabolic stress.
Collapse
Affiliation(s)
- Rebecca M. Fleeman
- Department of Neurosurgery, Penn State College of Medicine, Hershey, Pennsylvania, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Madison K. Kuhn
- Department of Neurosurgery, Penn State College of Medicine, Hershey, Pennsylvania, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, Pennsylvania, USA
- Department of Biomedical Engineering, Pennsylvania State University, University Park, State College, Pennsylvania, USA
- Center for Neural Engineering, Pennsylvania State University, University Park, State College, Pennsylvania, USA
| | - Dennis C. Chan
- Department of Neurosurgery, Penn State College of Medicine, Hershey, Pennsylvania, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, Pennsylvania, USA
- Department of Biomedical Engineering, Pennsylvania State University, University Park, State College, Pennsylvania, USA
- Center for Neural Engineering, Pennsylvania State University, University Park, State College, Pennsylvania, USA
| | - Elizabeth A. Proctor
- Department of Neurosurgery, Penn State College of Medicine, Hershey, Pennsylvania, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, Pennsylvania, USA
- Department of Biomedical Engineering, Pennsylvania State University, University Park, State College, Pennsylvania, USA
- Center for Neural Engineering, Pennsylvania State University, University Park, State College, Pennsylvania, USA
- Department of Engineering Science & Mechanics, Pennsylvania State University, University Park, State College, Pennsylvania, USA
| |
Collapse
|
28
|
Hanson AJ, Banks WA, Bettcher LF, Pepin R, Raftery D, Navarro SL, Craft S. Cerebrospinal Fluid Metabolomics: Pilot Study of Using Metabolomics to Assess Diet and Metabolic Interventions in Alzheimer's Disease and Mild Cognitive Impairment. Metabolites 2023; 13:569. [PMID: 37110227 PMCID: PMC10145981 DOI: 10.3390/metabo13040569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/17/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
Brain glucose hypometabolism is an early sign of Alzheimer's disease (AD), and interventions which offset this deficit, such as ketogenic diets, show promise as AD therapeutics. Conversely, high-fat feeding may exacerbate AD risk. We analyzed the metabolomic profile of cerebrospinal fluid (CSF) in a pilot study of older adults who underwent saline and triglyceride (TG) infusions. Older adults (12 cognitively normal (CN), age 65.3 ± 8.1, and 9 with cognitive impairment (CI), age 70.9 ± 8.6) underwent a 5 h TG or saline infusion on different days using a random crossover design; CSF was collected at the end of infusion. Aqueous metabolites were measured using a targeted mass spectroscopy (MS) platform focusing on 215 metabolites from over 35 different metabolic pathways. Data were analyzed using MetaboAnalyst 4.0 and SAS. Of the 215 targeted metabolites, 99 were detectable in CSF. Only one metabolite significantly differed by treatment: the ketone body 3-hydroxybutyrate (HBA). Post hoc analyses showed that HBA levels were associated with age and markers of metabolic syndrome and demonstrated different correlation patterns for the two treatments. When analyzed by cognitive diagnosis group, TG-induced increases in HBA were over 3 times higher for those with cognitive impairment (change score CN +9.8 uM ± 8.3, CI +32.4 ± 7.4, p = 0.0191). Interestingly, individuals with cognitive impairment had higher HBA levels after TG infusion than those with normal cognition. These results suggest that interventions that increase plasma ketones may lead to higher brain ketones in groups at risk for AD and should be confirmed in larger intervention studies.
Collapse
Affiliation(s)
- Angela J. Hanson
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA 98104, USA
| | - William A. Banks
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA 98104, USA
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98102, USA
| | - Lisa F. Bettcher
- Department of Anesthesiology and Pain Medicine, Northwest Metabolomics Research Center, University of Washington, Seattle, WA 98109, USA
| | - Robert Pepin
- Department of Anesthesiology and Pain Medicine, Northwest Metabolomics Research Center, University of Washington, Seattle, WA 98109, USA
| | - Daniel Raftery
- Department of Anesthesiology and Pain Medicine, Northwest Metabolomics Research Center, University of Washington, Seattle, WA 98109, USA
| | - Sandi L. Navarro
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Suzanne Craft
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27109, USA
| |
Collapse
|
29
|
Nwadiugwu M, Shen H, Deng HW. Potential Molecular Mechanisms of Alzheimer's Disease from Genetic Studies. BIOLOGY 2023; 12:biology12040602. [PMID: 37106802 PMCID: PMC10136191 DOI: 10.3390/biology12040602] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/09/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023]
Abstract
The devastating effects of Alzheimer's disease (AD) are yet to be ameliorated due to the absence of curative treatment options. AD is an aging-related disease that affects cognition, and molecular imbalance is one of its hallmarks. There is a need to identify common causes of molecular imbalance in AD and their potential mechanisms for continuing research. A narrative synthesis of molecular mechanisms in AD from primary studies that employed single-cell sequencing (scRNA-seq) or spatial genomics was conducted using Embase and PubMed databases. We found that differences in molecular mechanisms in AD could be grouped into four key categories: sex-specific features, early-onset features, aging, and immune system pathways. The reported causes of molecular imbalance were alterations in bile acid (BA) synthesis, PITRM1, TREM2, olfactory mucosa (OM) cells, cholesterol catabolism, NFkB, double-strand break (DSB) neuronal damage, P65KD silencing, tau and APOE expression. What changed from previous findings in contrast to results obtained were explored to find potential factors for AD-modifying investigations.
Collapse
Affiliation(s)
- Martin Nwadiugwu
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Hui Shen
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Hong-Wen Deng
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA 70112, USA
| |
Collapse
|
30
|
Tian JS, Zhao YH, Ling-Hu T, Wu WZ, Wang XX, Ji C, Zhao WD, Han YM, Qin XM. A novel insight for high-rate and low-efficiency glucose metabolism in depression through stable isotope-resolved metabolomics in CUMS-induced rats. J Affect Disord 2023; 331:121-129. [PMID: 36948469 DOI: 10.1016/j.jad.2023.03.061] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 03/12/2023] [Accepted: 03/18/2023] [Indexed: 03/24/2023]
Abstract
BACKGROUND Existing research has suggested that depression results in disorders of glucose metabolism in the organism which causing insufficient energy supply. However, the overall changes in glucose metabolism that arise from depression have not been clarified. METHODS In this study, the depression-like behavior in chronically unpredictable mild stressed rats was investigated, and the fate of glucose was tracked through isotope tracing and mass spectrometry, with a focus on metabolite changes in cecal contents. RESULTS As indicated by the results, the isotopic results of cecal contents can indicate the metabolic end of the organism. Moreover, the TCA cycle activity was notably reduced, and the gluconeogenesis pathway was abnormally up-regulated in the CUMS-induced rats. The organism expedited other glucose metabolism pathways to make up for the insufficiency of energy. As a result, the activity of the inefficient glycolysis pathway was increased. LIMITATIONS Existing research has only investigated the metabolism of 13C-glucose, and lipids and proteins have been rarely explored. CONCLUSIONS The chronic unpredictable mild stress can inhibit the entry of pyruvate into mitochondria in SD rats, such that the activity of TCA is reduced, and insufficient energy supply is caused. The organism is capable of expediting other glucose metabolism rate pathways to make up for the insufficiency of energy, whereas it still cannot compensate for the loss of energy. As a result, CUMS-induced rats exhibited high-rate and low-efficiency glucose metabolism.
Collapse
Affiliation(s)
- Jun-Sheng Tian
- Modern Research Center for Traditional Chinese Medicine of Shanxi University, No.92, Wucheng Road, Taiyuan 030006, China; Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Shanxi University, Taiyuan 030006, China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan 030006, China
| | - Yun-Hao Zhao
- Modern Research Center for Traditional Chinese Medicine of Shanxi University, No.92, Wucheng Road, Taiyuan 030006, China
| | - Ting Ling-Hu
- Modern Research Center for Traditional Chinese Medicine of Shanxi University, No.92, Wucheng Road, Taiyuan 030006, China
| | - Wen-Ze Wu
- Modern Research Center for Traditional Chinese Medicine of Shanxi University, No.92, Wucheng Road, Taiyuan 030006, China
| | - Xian-Xian Wang
- Modern Research Center for Traditional Chinese Medicine of Shanxi University, No.92, Wucheng Road, Taiyuan 030006, China
| | - Cui Ji
- School of Physical Education, Shanxi University, Taiyuan 030006, China
| | - Wei-di Zhao
- School of Physical Education, Shanxi University, Taiyuan 030006, China
| | - Yu-Mei Han
- School of Physical Education, Shanxi University, Taiyuan 030006, China
| | - Xue-Mei Qin
- Modern Research Center for Traditional Chinese Medicine of Shanxi University, No.92, Wucheng Road, Taiyuan 030006, China; Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Shanxi University, Taiyuan 030006, China; The Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan 030006, China.
| |
Collapse
|
31
|
Fleeman RM, Snyder AM, Kuhn MK, Chan DC, Smith GC, Crowley NA, Arnold AC, Proctor EA. Predictive link between systemic metabolism and cytokine signatures in the brain of apolipoprotein E ε4 mice. Neurobiol Aging 2023; 123:154-169. [PMID: 36572594 PMCID: PMC9892258 DOI: 10.1016/j.neurobiolaging.2022.11.015] [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: 06/29/2022] [Revised: 11/23/2022] [Accepted: 11/25/2022] [Indexed: 12/12/2022]
Abstract
The ε4 variant of apolipoprotein E (APOE) is the strongest and most common genetic risk factor for Alzheimer's disease (AD). While the mechanism of conveyed risk is incompletely understood, promotion of inflammation, dysregulated metabolism, and protein misfolding and aggregation are contributors to accelerating disease. Here we determined the concurrent effects of systemic metabolic changes and brain inflammation in young (3-month-old) and aged (18-month-old) male and female mice carrying the APOE4 gene. Using functional metabolic assays alongside multivariate modeling of hippocampal cytokine levels, we found that brain cytokine signatures are predictive of systemic metabolic outcomes, independent of AD proteinopathies. Male and female mice each produce different cytokine signatures as they age and as their systemic metabolic phenotype declines, and these signatures are APOE genotype dependent. Ours is the first study to identify a quantitative and predictive link between systemic metabolism and specific pathological cytokine signatures in the brain. Our results highlight the effects of APOE4 beyond the brain and suggest the potential for bi-directional influence of risk factors in the brain and periphery.
Collapse
Affiliation(s)
- Rebecca M Fleeman
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA; Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
| | - Amanda M Snyder
- Department of Neurology, Penn State College of Medicine, Hershey, PA, USA
| | - Madison K Kuhn
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA; Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA; Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, USA; Center for Neural Engineering, Pennsylvania State University, University Park, PA, USA
| | - Dennis C Chan
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA; Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA; Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, USA; Center for Neural Engineering, Pennsylvania State University, University Park, PA, USA
| | - Grace C Smith
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, USA; Department of Biology, Pennsylvania State University, University Park, PA, USA
| | - Nicole A Crowley
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, USA; Center for Neural Engineering, Pennsylvania State University, University Park, PA, USA; Department of Biology, Pennsylvania State University, University Park, PA, USA
| | - Amy C Arnold
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA, USA
| | - Elizabeth A Proctor
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA; Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA; Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, USA; Center for Neural Engineering, Pennsylvania State University, University Park, PA, USA; Department of Engineering Science & Mechanics, Pennsylvania State University, University Park, PA, USA.
| |
Collapse
|
32
|
Lee S, Williams HC, Gorman AA, Devanney NA, Harrison DA, Walsh AE, Goulding DS, Tuck T, Schwartz JL, Zajac DJ, Macauley SL, Estus S, Julia TCW, Johnson LA, Morganti JM. APOE4 drives transcriptional heterogeneity and maladaptive immunometabolic responses of astrocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.06.527204. [PMID: 36798317 PMCID: PMC9934552 DOI: 10.1101/2023.02.06.527204] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Apolipoprotein E4 (APOE4) is the strongest risk allele associated with the development of late onset Alzheimer's disease (AD). Across the CNS, astrocytes are the predominant expressor of APOE while also being critical mediators of neuroinflammation and cerebral metabolism. APOE4 has been consistently linked with dysfunctional inflammation and metabolic processes, yet insights into the molecular constituents driving these responses remain unclear. Utilizing complementary approaches across humanized APOE mice and isogenic human iPSC astrocytes, we demonstrate that ApoE4 alters the astrocyte immunometabolic response to pro-inflammatory stimuli. Our findings show that ApoE4-expressing astrocytes acquire distinct transcriptional repertoires at single-cell and spatially-resolved domains, which are driven in-part by preferential utilization of the cRel transcription factor. Further, inhibiting cRel translocation in ApoE4 astrocytes abrogates inflammatory-induced glycolytic shifts and in tandem mitigates production of multiple pro-inflammatory cytokines. Altogether, our findings elucidate novel cellular underpinnings by which ApoE4 drives maladaptive immunometabolic responses of astrocytes.
Collapse
Affiliation(s)
- Sangderk Lee
- Sanders-Brown Center on Aging, University of Kentucky College of Medicine, Lexington, KY
| | - Holden C. Williams
- Sanders-Brown Center on Aging, University of Kentucky College of Medicine, Lexington, KY
- Department of Physiology, University of Kentucky College of Medicine, Lexington, KY
| | - Amy A. Gorman
- Sanders-Brown Center on Aging, University of Kentucky College of Medicine, Lexington, KY
| | - Nicholas A. Devanney
- Sanders-Brown Center on Aging, University of Kentucky College of Medicine, Lexington, KY
- Department of Biology, University of Kentucky College of Arts and Sciences, Lexington, KY
| | | | - Adeline E. Walsh
- Department of Biology, University of Kentucky College of Arts and Sciences, Lexington, KY
| | - Danielle S. Goulding
- Sanders-Brown Center on Aging, University of Kentucky College of Medicine, Lexington, KY
| | - Tony Tuck
- Boston University, Chobanian & Avedisian School of Medicine, Boston, MA
| | - James L. Schwartz
- Sanders-Brown Center on Aging, University of Kentucky College of Medicine, Lexington, KY
| | - Diana J. Zajac
- Sanders-Brown Center on Aging, University of Kentucky College of Medicine, Lexington, KY
- Department of Biology, University of Kentucky College of Arts and Sciences, Lexington, KY
| | - Shannon L. Macauley
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC
| | - Steven Estus
- Sanders-Brown Center on Aging, University of Kentucky College of Medicine, Lexington, KY
- Department of Biology, University of Kentucky College of Arts and Sciences, Lexington, KY
| | - TCW Julia
- Boston University, Chobanian & Avedisian School of Medicine, Boston, MA
| | - Lance A. Johnson
- Sanders-Brown Center on Aging, University of Kentucky College of Medicine, Lexington, KY
- Department of Biology, University of Kentucky College of Arts and Sciences, Lexington, KY
| | - Josh M. Morganti
- Sanders-Brown Center on Aging, University of Kentucky College of Medicine, Lexington, KY
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY
| |
Collapse
|
33
|
Zhang X, Wu L, Swerdlow RH, Zhao L. Opposing Effects of ApoE2 and ApoE4 on Glycolytic Metabolism in Neuronal Aging Supports a Warburg Neuroprotective Cascade against Alzheimer's Disease. Cells 2023; 12:410. [PMID: 36766752 PMCID: PMC9914046 DOI: 10.3390/cells12030410] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 01/20/2023] [Accepted: 01/23/2023] [Indexed: 01/27/2023] Open
Abstract
Apolipoprotein E4 (ApoE4) is the most recognized genetic risk factor for late-onset Alzheimer's disease (LOAD), whereas ApoE2 reduces the risk for LOAD. The underlying mechanisms are unclear but may include effects on brain energy metabolism. Here, we used neuro-2a (N2a) cells that stably express human ApoE isoforms (N2a-hApoE), differentiated N2a-hApoE neuronal cells, and humanized ApoE knock-in mouse models to investigate relationships among ApoE isoforms, glycolytic metabolism, and neuronal health and aging. ApoE2-expressing cells retained robust hexokinase (HK) expression and glycolytic activity, whereas these endpoints progressively declined with aging in ApoE4-expressing cells. These divergent ApoE2 and ApoE4 effects on glycolysis directly correlated with markers of cellular wellness. Moreover, ApoE4-expressing cells upregulated phosphofructokinase and pyruvate kinase with the apparent intent of compensating for the HK-dependent glycolysis reduction. The introduction of ApoE2 increased HK levels and glycolysis flux in ApoE4 cells. PI3K/Akt signaling was distinctively regulated by ApoE isoforms but was only partially responsible for the ApoE-mediated effects on HK. Collectively, our findings indicate that human ApoE isoforms differentially modulate neuronal glycolysis through HK regulation, with ApoE2 upregulating and ApoE4 downregulating, which markedly impacts neuronal health during aging. These findings lend compelling support to the emerging inverse-Warburg theory of AD and highlight a therapeutic opportunity for bolstering brain glycolytic resilience to prevent and treat AD.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA
| | - Long Wu
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA
| | - Russell H. Swerdlow
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Liqin Zhao
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA
- Neuroscience Graduate Program, University of Kansas, Lawrence, KS 66045, USA
| |
Collapse
|
34
|
Macyczko JR, Wang N, Zhao J, Ren Y, Lu W, Ikezu TC, Zhao N, Liu CC, Bu G, Li Y. Suppression of Wnt/β-Catenin Signaling Is Associated with Downregulation of Wnt1, PORCN, and Rspo2 in Alzheimer's Disease. Mol Neurobiol 2023; 60:26-35. [PMID: 36215026 PMCID: PMC9795414 DOI: 10.1007/s12035-022-03065-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 10/03/2022] [Indexed: 12/30/2022]
Abstract
Wnt and R-spondin (Rspo) proteins are two major types of endogenous Wnt/β-catenin signaling agonists. While Wnt/β-catenin signaling is greatly diminished in Alzheimer's disease (AD), it remains to be elucidated whether the inhibition of this pathway is associated with dysregulation of Wnt and Rspo proteins. By analyzing temporal cortex RNA-seq data of the human postmortem brain samples, we found that WNT1 and RRPO2 were significantly downregulated in human AD brains. In addition, the expression of Wnt acyltransferase porcupine (PORCN), which is essential for Wnt maturation and secretion, was greatly deceased in these human AD brains. Interestingly, the lowest levels of WNT1, PORCN, and RSPO2 expression were found in human AD brains carrying two copies of APOE4 allele, the strongest genetic risk factor of late-onset AD. Importantly, there were positive correlations among the levels of WNT1, PORCN, and RSPO2 expression in human AD brains. Supporting observations in humans, Wnt1, PORCN, and Rspo2 were downregulated and Wnt/β-catenin signaling was diminished in the 5xFAD amyloid model mice. In human APOE-targeted replacement mice, downregulation of WNT1, PORCN, and RSPO2 expression was positively associated with aging and APOE4 genotype. Finally, WNT1 and PORCN expression and Wnt/β-catenin signaling were inhibited in human APOE4 iPSC-derived astrocytes when compared to the isogenic APOE3 iPSC-derived astrocytes. Altogether, our findings suggest that the dysregulations of Wnt1, PORCN, and Rspo2 could be coordinated together to diminish Wnt/β-catenin signaling in aging- and APOE4-dependent manners in the AD brain.
Collapse
Affiliation(s)
- Jesse R Macyczko
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Na Wang
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Jing Zhao
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
- Center for Regenerative Medicine, Neuroregeneration Laboratory, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Yingxue Ren
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Wenyan Lu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
- Center for Regenerative Medicine, Neuroregeneration Laboratory, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Tadafumi C Ikezu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Na Zhao
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Chia-Chen Liu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
- Center for Regenerative Medicine, Neuroregeneration Laboratory, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Yonghe Li
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA.
| |
Collapse
|
35
|
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
|
36
|
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
|
37
|
Linghu T, Zhao Y, Wu W, Gao Y, Tian J, Qin X. Novel targets for ameliorating energy metabolism disorders in depression through stable isotope-resolved metabolomics. BIOCHIMICA ET BIOPHYSICA ACTA. BIOENERGETICS 2022; 1863:148578. [PMID: 35640666 DOI: 10.1016/j.bbabio.2022.148578] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 04/28/2022] [Accepted: 05/25/2022] [Indexed: 06/15/2023]
Abstract
The severe harm of depression to human health and life has attracted global attention, but the exact mechanism is not yet known due to the complicated pathogenesis. The existing antidepressants are far from ideal, indicating it is urgently needed to seek safe and effective drugs from a unique perspective. Based on the hypothesis of "mitochondrial dysfunction" proposed recently, we attempt to focus on the substrates supply of energy metabolism. We applied stable isotope-resolved metabolomics, and revealed that significantly decreased TCA cycle and abnormally increased gluconeogenesis pathway in CUMS rats. Pyruvate dehydrogenase (PDH) and pyruvate carboxylase (PC) maybe the key metabolic enzymes. This metabolic reprogramming was confirmed through ELISA assays and Western blot analysis. To explore the causes of substrates supply disorder in depression, we conducted the mitochondrial structure-function evaluation. Interestingly, the levels of the mitochondrial pyruvate carrier (MPC) decreased significantly, which is essential for the entry of pyruvic acid into the TCA cycle. Together, MPC, PDH and PC are expected to become potential novel therapeutic targets for treating depressive disorders. This research provides a unique insight for re-cognizing the pathological mechanisms of depression, the novel targets for development of ideal antidepressants, as well as a paradigm for deciphering abnormal metabolic pathways in other metabolic diseases.
Collapse
Affiliation(s)
- Ting Linghu
- Modern Research Center for Traditional Chinese Medicine, the Institute for Biomedicine and Health, the Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, the Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Shanxi University, Taiyuan 030006, China
| | - Yunhao Zhao
- Modern Research Center for Traditional Chinese Medicine, the Institute for Biomedicine and Health, the Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, the Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Shanxi University, Taiyuan 030006, China
| | - Wenze Wu
- Modern Research Center for Traditional Chinese Medicine, the Institute for Biomedicine and Health, the Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, the Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Shanxi University, Taiyuan 030006, China
| | - Yao Gao
- Department of Psychiatry, First Hospital/First Clinical Medical College of Shanxi Medical University, Taiyuan 030001, China
| | - Junsheng Tian
- Modern Research Center for Traditional Chinese Medicine, the Institute for Biomedicine and Health, the Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, the Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Shanxi University, Taiyuan 030006, China.
| | - Xuemei Qin
- Modern Research Center for Traditional Chinese Medicine, the Institute for Biomedicine and Health, the Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, the Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Shanxi University, Taiyuan 030006, China.
| |
Collapse
|
38
|
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
|
39
|
Turner AK, Shaw BC, Simpson JF, Estus S. Identification and Quantitation of Novel ABI3 Isoforms Relative to Alzheimer’s Disease Genetics and Neuropathology. Genes (Basel) 2022; 13:genes13091607. [PMID: 36140776 PMCID: PMC9498898 DOI: 10.3390/genes13091607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 08/31/2022] [Accepted: 09/02/2022] [Indexed: 11/17/2022] Open
Abstract
Elucidating the actions of genetic polymorphisms associated with the risk of Alzheimer’s disease (AD) may provide novel insights into underlying mechanisms. Two polymorphisms have implicated ABI3 as a modulator of AD risk. Here, we sought to identify ABI3 isoforms expressed in human AD and non-AD brain, quantify the more abundant isoforms as a function of AD genetics and neuropathology, and provide an initial in vitro characterization of the proteins produced by these novel isoforms. We report that ABI3 expression is increased with AD neuropathology but not associated with AD genetics. Single-cell RNAseq of APP/PS1 mice showed that Abi3 is primarily expressed by microglia, including disease-associated microglia. In human brain, several novel ABI3 isoforms were identified, including isoforms with partial or complete loss of exon 6. Expression of these isoforms correlated tightly with total ABI3 expression but were not influenced by AD genetics. Lastly, we performed an initial characterization of these isoforms in transfected cells and found that, while full-length ABI3 was expressed in a dispersed punctate fashion within the cytosol, isoforms lacking most or all of exon six tended to form extensive protein aggregates. In summary, ABI3 expression is restricted to microglia, is increased with Alzheimer’s neuropathology, and includes several isoforms that display a variable tendency to aggregate when expressed in vitro.
Collapse
|
40
|
Andersen JV, Schousboe A, Verkhratsky A. Astrocyte energy and neurotransmitter metabolism in Alzheimer's disease: integration of the glutamate/GABA-glutamine cycle. Prog Neurobiol 2022; 217:102331. [PMID: 35872221 DOI: 10.1016/j.pneurobio.2022.102331] [Citation(s) in RCA: 79] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 07/14/2022] [Accepted: 07/19/2022] [Indexed: 02/06/2023]
Abstract
Astrocytes contribute to the complex cellular pathology of Alzheimer's disease (AD). Neurons and astrocytes function in close collaboration through neurotransmitter recycling, collectively known as the glutamate/GABA-glutamine cycle, which is essential to sustain neurotransmission. Neurotransmitter recycling is intimately linked to astrocyte energy metabolism. In the course of AD, astrocytes undergo extensive metabolic remodeling, which may profoundly affect the glutamate/GABA-glutamine cycle. The consequences of altered astrocyte function and metabolism in relation to neurotransmitter recycling are yet to be comprehended. Metabolic alterations of astrocytes in AD deprive neurons of metabolic support, thereby contributing to synaptic dysfunction and neurodegeneration. In addition, several astrocyte-specific components of the glutamate/GABA-glutamine cycle, including glutamine synthesis and synaptic neurotransmitter uptake, are perturbed in AD. Integration of the complex astrocyte biology within the context of AD is essential for understanding the fundamental mechanisms of the disease, while restoring astrocyte metabolism may serve as an approach to arrest or even revert clinical progression of AD.
Collapse
Affiliation(s)
- Jens V Andersen
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark.
| | - Arne Schousboe
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK; Achucarro Center for Neuroscience, IKERBASQUE, 48011 Bilbao, Spain; Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, LT-01102 Vilnius, Lithuania.
| |
Collapse
|
41
|
Skowronska-Krawczyk D, Narayan P, Tessarz P. Editorial: Molecular Role of Lipids in Aging. FRONTIERS IN AGING 2022; 3:946884. [PMID: 35821810 PMCID: PMC9261349 DOI: 10.3389/fragi.2022.946884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 05/23/2022] [Indexed: 11/13/2022]
Affiliation(s)
- Dorota Skowronska-Krawczyk
- Department of Physiology and Biophysics, Department of Ophthalmology, Center for Translational Vision Research, School of Medicine, University of California Irvine, Irvine, CA, United States
| | - Priyanka Narayan
- Genetics and Biochemistry Branch, National Institute of Diabetes and Digestive and Kidney Diseases and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD, United States
| | - Peter Tessarz
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| |
Collapse
|
42
|
Factors Influencing Alzheimer's Disease Risk: Whether and How They are Related to the APOE Genotype. Neurosci Bull 2022; 38:809-819. [PMID: 35149974 PMCID: PMC9276873 DOI: 10.1007/s12264-021-00814-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 11/25/2021] [Indexed: 11/21/2022] Open
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disease featuring progressive cognitive impairment. Although the etiology of late-onset AD remains unclear, the close association of AD with apolipoprotein E (APOE), a gene that mainly regulates lipid metabolism, has been firmly established and may shed light on the exploration of AD pathogenesis and therapy. However, various confounding factors interfere with the APOE-related AD risk, raising questions about our comprehension of the clinical findings concerning APOE. In this review, we summarize the most debated factors interacting with the APOE genotype and AD pathogenesis, depict the extent to which these factors relate to APOE-dependent AD risk, and discuss the possible underlying mechanisms.
Collapse
|
43
|
Troutwine BR, Hamid L, Lysaker CR, Strope TA, Wilkins HM. Apolipoprotein E and Alzheimer's disease. Acta Pharm Sin B 2022; 12:496-510. [PMID: 35256931 PMCID: PMC8897057 DOI: 10.1016/j.apsb.2021.10.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/30/2021] [Accepted: 10/07/2021] [Indexed: 12/14/2022] Open
Abstract
Genetic variation in apolipoprotein E (APOE) influences Alzheimer's disease (AD) risk. APOE ε4 alleles are the strongest genetic risk factor for late onset sporadic AD. The AD risk is dose dependent, as those carrying one APOE ε4 allele have a 2-3-fold increased risk, while those carrying two ε4 alleles have a 10-15-fold increased risk. Individuals carrying APOE ε2 alleles have lower AD risk and those carrying APOE ε3 alleles have neutral risk. APOE is a lipoprotein which functions in lipid transport, metabolism, and inflammatory modulation. Isoform specific effects of APOE within the brain include alterations to Aβ, tau, neuroinflammation, and metabolism. Here we review the association of APOE with AD, the APOE isoform specific effects within brain and periphery, and potential therapeutics.
Collapse
Affiliation(s)
- Benjamin R. Troutwine
- Department of Neurology University of Kansas Medical Center, Kansas City, KS 66160, USA
- University of Kansas Alzheimer's Disease Center, Kansas City, KS 66160, USA
| | - Laylan Hamid
- University of Kansas Alzheimer's Disease Center, Kansas City, KS 66160, USA
| | - Colton R. Lysaker
- University of Kansas Alzheimer's Disease Center, Kansas City, KS 66160, USA
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Taylor A. Strope
- University of Kansas Alzheimer's Disease Center, Kansas City, KS 66160, USA
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Heather M. Wilkins
- Department of Neurology University of Kansas Medical Center, Kansas City, KS 66160, USA
- University of Kansas Alzheimer's Disease Center, Kansas City, KS 66160, USA
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
44
|
Barthelson K, Newman M, Lardelli M. Brain transcriptomes of zebrafish and mouse Alzheimer's disease knock-in models imply early disrupted energy metabolism. Dis Model Mech 2021; 15:273566. [PMID: 34842276 PMCID: PMC8807579 DOI: 10.1242/dmm.049187] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 11/17/2021] [Indexed: 11/21/2022] Open
Abstract
Energy production is the most fundamentally important cellular activity supporting all other functions, particularly in highly active organs, such as brains. Here, we summarise transcriptome analyses of young adult (pre-disease) brains from a collection of 11 early-onset familial Alzheimer's disease (EOFAD)-like and non-EOFAD-like mutations in three zebrafish genes. The one cellular activity consistently predicted as affected by only the EOFAD-like mutations is oxidative phosphorylation, which produces most of the energy of the brain. All the mutations were predicted to affect protein synthesis. We extended our analysis to knock-in mouse models of APOE alleles and found the same effect for the late onset Alzheimer's disease risk allele ε4. Our results support a common molecular basis for the initiation of the pathological processes leading to both early and late onset forms of Alzheimer's disease, and illustrate the utility of zebrafish and knock-in single EOFAD mutation models for understanding the causes of this disease. Summary: Young adult zebrafish mutants and a mouse model of a genetic variant promoting early- and late-onset Alzheimer's disease, respectively, share changes in brain gene expression, indicating disturbance of oxidative phosphorylation.
Collapse
Affiliation(s)
- Karissa Barthelson
- Alzheimer's Disease Genetics Laboratory, School of Biological Sciences, University of Adelaide, North Terrace, Adelaide, SA 5005, Australia
| | - Morgan Newman
- Alzheimer's Disease Genetics Laboratory, School of Biological Sciences, University of Adelaide, North Terrace, Adelaide, SA 5005, Australia
| | - Michael Lardelli
- Alzheimer's Disease Genetics Laboratory, School of Biological Sciences, University of Adelaide, North Terrace, Adelaide, SA 5005, Australia
| |
Collapse
|