1
|
Zhu D, Wu JZ, Griffin PT, Samuelson BA, Sinclair DA, Kane AE. Metabolomics biomarkers of frailty: a longitudinal study of aging female and male mice. NPJ AGING 2025; 11:40. [PMID: 40410187 DOI: 10.1038/s41514-025-00237-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 05/12/2025] [Indexed: 05/25/2025]
Abstract
Frailty is an age-related geriatric syndrome. We performed a longitudinal study of aging female (n = 40) and male (n = 47) C57BL/6NIA mice, measured frailty index and derived metabolomics data from plasma. We identify age-related differentially abundant metabolites, determine frailty-related metabolites, and generate frailty features, both in the whole cohort and sex-stratified subgroups. Using the features, we perform an association study and build a metabolomics-based frailty clock. We find that frailty-related metabolites are enriched for amino acid metabolism and metabolism of cofactors and vitamins, include ergothioneine, tryptophan and alpha-ketoglutarate, and present sex dimorphism. We identify B vitamin metabolism related flavin-adenine dinucleotide and pyridoxate as female-specific frailty biomarkers, and lipid metabolism related sphingomyelins, glycerophosphoethanolamine and glycerophosphocholine as male-specific frailty biomarkers. These associations are confirmed in a validation cohort, with ergothioneine and perfluorooctanesulfonate identified as robust frailty biomarkers. Our results identify sex-specific metabolite frailty biomarkers, and shed light on potential mechanisms.
Collapse
Affiliation(s)
- Dantong Zhu
- Institute for Systems Biology, Seattle, WA, USA
| | - Judy Z Wu
- Institute for Systems Biology, Seattle, WA, USA
| | - Patrick T Griffin
- Blavatnik Institute, Department of Genetics, Paul F. Glenn Center for Biology of Aging Research at Harvard Medical School, Boston, MA, USA
| | | | - David A Sinclair
- Blavatnik Institute, Department of Genetics, Paul F. Glenn Center for Biology of Aging Research at Harvard Medical School, Boston, MA, USA
| | - Alice E Kane
- Institute for Systems Biology, Seattle, WA, USA.
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
2
|
Mei Z, Liu J, Bennett DA, Seyfried N, Wingo AP, Wingo TS. Unraveling sex differences in Alzheimer's disease and related endophenotypes with brain proteomes. Alzheimers Dement 2025; 21:e70206. [PMID: 40346727 PMCID: PMC12064417 DOI: 10.1002/alz.70206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 03/11/2025] [Accepted: 03/29/2025] [Indexed: 05/11/2025]
Abstract
INTRODUCTION Sex differences exist in Alzheimer's disease (AD), but the underlying mechanisms remain unclear. METHODS We examined brain proteomes profiled from the dorsolateral prefrontal cortex of 770 donors (66.2% female). RESULTS Proteome-wide differential expression analysis in males and females jointly identified many significant proteins for AD dementia (n = 1228), amyloid beta (n = 1183), tangles (n = 1309), and global cognitive trajectory (n = 2325) at a false discovery rate of <0.05. Sex-stratified analyses also identified many proteins associated with AD or its endophenotypes. Finally, we found 10 proteins with significant sex-by-trait interactions, including one in AD clinical diagnosis (MARCKS), seven in cognitive trajectories (TOGARAM1, PLCD3, SLC22A5, MTFR1L, DCUN1D5, S100A12, and TRIM46), and two in cerebral pathologies (PANK4 and SOS1). DISCUSSION The 10 proteins with sex interaction in AD cover a range of functions likely relevant for AD pathogenesis, including estrogen response, inflammation, and mitochondrial biology, and their specific roles in AD ought to be studied. Future work should test their potential as sex-specific AD biomarkers. HIGHLIGHTS At the phenotypic level, we found sex differences in baseline cognitive performance, cognitive trajectories, and AD hallmark pathologies. Proteome-wide differential expression analyses identified many brain proteins associated with AD and its endophenotypes in either sex alone or when considered together. We found 10 brain proteins with significant sex interactions in AD and its endophenotypes, which could be investigated as potential sex-specific biomarkers of AD.
Collapse
Affiliation(s)
- Zhen Mei
- Department of NeurologyUniversity of California, DavisSacramentoCaliforniaUSA
| | - Jiaqi Liu
- Department of PsychiatryUniversity of California, DavisSacramentoCaliforniaUSA
| | - David A Bennett
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
| | - Nicholas Seyfried
- Department of BiochemistryEmory University School of MedicineAtlantaGeorgiaUSA
| | - Aliza P. Wingo
- Department of PsychiatryUniversity of California, DavisSacramentoCaliforniaUSA
- Division of Mental HealthAtlanta VA Medical CenterDecaturGeorgiaUSA
| | - Thomas S. Wingo
- Department of NeurologyUniversity of California, DavisSacramentoCaliforniaUSA
- Alzheimer's Disease Research CenterUniversity of California, DavisSacramentoCaliforniaUSA
| |
Collapse
|
3
|
Jeong H, Pan Y, Akhter F, Volkow ND, Zhu D, Du C. Evidence of cortical vascular impairments in early stage of Alzheimer's transgenic mice: Optical imaging. J Cereb Blood Flow Metab 2025; 45:960-976. [PMID: 39696904 PMCID: PMC12035375 DOI: 10.1177/0271678x241304893] [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: 12/21/2023] [Revised: 08/20/2024] [Accepted: 11/11/2024] [Indexed: 12/20/2024]
Abstract
Alzheimer's disease (AD), a neurodegenerative disorder with progressive cognitive decline, remains clinically challenging with limited understanding of etiology and interventions. Clinical studies have reported vascular defects prior to other pathological manifestations of AD, leading to the "Vascular Hypothesis" for the disorder. However, in vivo assessments of cerebral vasculature in AD rodent models have been constrained by limited spatiotemporal resolution or field of view of conventional imaging. We herein employed two in vivo imaging technologies, Dual-Wavelength Imaging and Optical Coherence Doppler Tomography, to evaluate cerebrovascular reactivity (CVR) to vasoconstrictive cocaine and vasodilatory hypercapnia challenges and to detect resting 3D cerebral blood flow (CBF) in living transgenic AD mice at capillary resolution. Results showed that CVR to cocaine and hypercapnia was significantly attenuated in 7-10 months old AD mice vs controls, indicating reduced vascular flexibility and reactivity. Additionally, in the AD mice, arterial CBF velocities were slower and the microvascular density in cortex was decreased compared to controls. These results reveal significant vascular impairments including reduced CVR and resting CBF in early-staged AD mice. Hence, this cutting-edge in vivo optical imaging offers an innovative venue for detecting early neurovascular dysfunction in AD brain with translational potential.
Collapse
Affiliation(s)
- Hyomin Jeong
- Department of Biomedical Engineering, State University of New York at Stony Brook, Stony Brook, NY, USA
| | - Yingtian Pan
- Department of Biomedical Engineering, State University of New York at Stony Brook, Stony Brook, NY, USA
| | - Firoz Akhter
- Department of Biomedical Engineering, State University of New York at Stony Brook, Stony Brook, NY, USA
| | - Nora D Volkow
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Donghui Zhu
- Department of Biomedical Engineering, State University of New York at Stony Brook, Stony Brook, NY, USA
| | - Congwu Du
- Department of Biomedical Engineering, State University of New York at Stony Brook, Stony Brook, NY, USA
| |
Collapse
|
4
|
Roberts H, Fang Y, Quinn K, Hill T, Peck MR, Bartke A, Hascup KN, Hascup ER. Lifespan of male and female APP/PS1 and APP NL-F/NL-F mouse models of Alzheimer's disease. J Alzheimers Dis 2025; 105:56-61. [PMID: 40138433 DOI: 10.1177/13872877251325878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Alzheimer's disease (AD) disproportionately affects women, yet most preclinical research studies are male-centric. We performed lifespan analyses of male and female AD mouse models (APP/PS1 and APPNL-F/NL-F) and their shared genetic background control (C57BL/6). Survival curves support significant sex differences between genotypes. Minimal longevity revealed increased age in male APP/PS1, and decreased age in male APPNL-F/NL-F mice. Maximal longevity revealed an increased average age in males. Furthermore, median lifespan differed between sex and genotype. This study supports dimorphic survival in two mouse models of AD, emphasizing the need to examine mechanisms and treatments in both sexes.
Collapse
Affiliation(s)
- Hannah Roberts
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer's Research and Treatment, Departments of Neurology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Yimin Fang
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer's Research and Treatment, Departments of Neurology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Kathleen Quinn
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer's Research and Treatment, Departments of Neurology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Tiarra Hill
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer's Research and Treatment, Departments of Neurology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Mackenzie R Peck
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer's Research and Treatment, Departments of Neurology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Andrzej Bartke
- Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL, USA
- Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Kevin N Hascup
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer's Research and Treatment, Departments of Neurology, Southern Illinois University School of Medicine, Springfield, IL, USA
- Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL, USA
- Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Erin R Hascup
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer's Research and Treatment, Departments of Neurology, Southern Illinois University School of Medicine, Springfield, IL, USA
- Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA
| |
Collapse
|
5
|
Knudtzon S, Nordengen K, Pålhaugen L, Gísladóttir B, Jarholm J, Bråthen G, Skogseth RE, Waterloo K, Selnes P, Fladby T, Kirsebom BE. Sexual dimorphisms in innate immune activation markers in predementia Alzheimer's disease. Brain Commun 2025; 7:fcaf161. [PMID: 40322776 PMCID: PMC12046404 DOI: 10.1093/braincomms/fcaf161] [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: 10/11/2024] [Revised: 04/04/2025] [Accepted: 04/23/2025] [Indexed: 05/08/2025] Open
Abstract
Females have an increased risk of developing Alzheimer's disease (AD). The innate immune system plays a key role in AD pathology, and sex differences in innate immune responses may contribute to differences in disease risk and progression. This study investigated sex differences in innate immune responses among participants without cerebrospinal fluid (CSF) determined amyloid pathology [A-; cognitively normal (CN), n = 83] and those with amyloid pathology (A+, n = 202), further stratified into preclinical (CN with A+, n = 72) and mild cognitive impairment (MCI with A+, n = 130). Participants were drawn from the Norwegian Dementia Disease Initiation cohort (n = 285). We measured plasma glial fibrillary acidic protein (GFAP) and CSF concentrations of nine innate immune markers: soluble triggering receptor expressed on myeloid cells 2 (sTREM2), monocyte chemoattractant protein 1 (MCP-1), fractalkine, chitinase 3-like 1 (YKL-40), clusterin, interferon gamma (IFN-γ), interleukin-6 (IL-6), IL-10, and IL-18. Linear regression was used, adjusted for multiple comparisons using the false discovery rate. In A+ cases (n = 202, females = 105), females had lower MCP-1 (P < 0.01), IL-6 and IL-18 (both P < 0.05) than males, while no sex differences were observed in A- cases (n = 83, females = 39). Among A+ participants, no sex differences were observed in CN cases (n = 72, females = 37), but females (n = 68) with MCI had lower MCP-1 and IL-6 (both P < 0.05) than males (n = 62) with MCI. Moreover, A+ females exhibited stronger positive associations between sTREM2 and clusterin with CSF total tau (P < 0.001; P < 0.05) and Neurofilament light chain (P < 0.01; P < 0.01) than males. These findings suggest sex-specific differences in innate immune responses, which may contribute to disease progression in amyloid-positive individuals.
Collapse
Affiliation(s)
- Stephanie Knudtzon
- Department of Neurology, University Hospital of North Norway, 9038 Tromsø, Norway
- Department of Psychology, Faculty of Health Sciences, UiT The Arctic University of Norway, 9037 Tromsø, Norway
| | - Kaja Nordengen
- Department of Neurology, Akershus University Hospital, 1478 Lørenskog, Norway
- Institute of Clinical Medicine, University of Oslo, 0318 Oslo, Norway
| | - Lene Pålhaugen
- Department of Neurology, Akershus University Hospital, 1478 Lørenskog, Norway
- Institute of Clinical Medicine, University of Oslo, 0318 Oslo, Norway
| | - Berglind Gísladóttir
- Department of Neurology, Akershus University Hospital, 1478 Lørenskog, Norway
- Clinical Molecular Biology (EpiGen), Medical Division, Akershus University Hospital and University of Oslo, 1478 Lørenskog, Norway
| | - Jonas Jarholm
- Department of Neurology, Akershus University Hospital, 1478 Lørenskog, Norway
- Institute of Clinical Medicine, University of Oslo, 0318 Oslo, Norway
| | - Geir Bråthen
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Science, Norwegian University of Science and Technology, 7491 Trondheim, Norway
- Department of Neurology and Clinical Neurophysiology, Trondheim University Hospital, 7491 Trondheim, Norway
| | - Ragnhild Eide Skogseth
- Department of Geriatric Medicine and the Neuro-SysMed Centre, Haraldsplass Deaconess Hospital, 5021 Bergen, Norway
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway
| | - Knut Waterloo
- Department of Neurology, University Hospital of North Norway, 9038 Tromsø, Norway
- Department of Psychology, Faculty of Health Sciences, UiT The Arctic University of Norway, 9037 Tromsø, Norway
| | - Per Selnes
- Department of Neurology, Akershus University Hospital, 1478 Lørenskog, Norway
| | - Tormod Fladby
- Department of Neurology, Akershus University Hospital, 1478 Lørenskog, Norway
- Institute of Clinical Medicine, University of Oslo, 0318 Oslo, Norway
| | - Bjørn-Eivind Kirsebom
- Department of Neurology, University Hospital of North Norway, 9038 Tromsø, Norway
- Department of Psychology, Faculty of Health Sciences, UiT The Arctic University of Norway, 9037 Tromsø, Norway
- Department of Neurology, Akershus University Hospital, 1478 Lørenskog, Norway
- Institute of Clinical Medicine, University of Oslo, 0318 Oslo, Norway
| |
Collapse
|
6
|
Zhu D, Wu JZ, Griffin P, Samuelson BA, Sinclair DA, Kane AE. Metabolomics biomarkers of frailty: a longitudinal study of aging female and male mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.22.634160. [PMID: 39896479 PMCID: PMC11785134 DOI: 10.1101/2025.01.22.634160] [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: 02/04/2025]
Abstract
Frailty is an age-related geriatric syndrome, for which the mechanisms remain largely unknown. We performed a longitudinal study of aging female (n = 40) and male (n = 47) C57BL/6NIA mice, measured frailty index and derived metabolomics data from plasma samples. We identify differentially abundant metabolites related to aging, determine frailty related metabolites via a machine learning approach, and generate a union set of frailty features, both in the whole cohort and in sex-stratified subgroups. Using the features, we perform an association study and build a metabolomics-based frailty clock. We find that frailty related metabolites are enriched for amino acid metabolism and metabolism of cofactors and vitamins, include ergothioneine, tryptophan, and alpha-ketoglutarate, and present sex dimorphism. We identify B vitamin metabolism related flavin adenine dinucleotide and pyridoxate as female-specific frailty biomarkers, and lipid metabolism related sphingomyelins, glycerophosphoethanolamine and glycerophosphocholine as male-specific frailty biomarkers. These associations are confirmed in a validation cohort, with ergothioneine and perfluorooctanesulfonate identified as robust frailty biomarkers. In summary, our results identify sex-specific metabolite biomarkers of frailty in aging, and shed light on potential mechanisms involved in frailty.
Collapse
Affiliation(s)
- Dantong Zhu
- Institute for Systems Biology, Seattle, WA 98109, USA
| | - Judy Z. Wu
- Institute for Systems Biology, Seattle, WA 98109, USA
| | - Patrick Griffin
- Blavatnik Institute, Department of Genetics, Paul F. Glenn Center for Biology of Aging Research at Harvard Medical School, Boston, MA 02115, USA
| | | | - David A. Sinclair
- Blavatnik Institute, Department of Genetics, Paul F. Glenn Center for Biology of Aging Research at Harvard Medical School, Boston, MA 02115, USA
| | - Alice E. Kane
- Institute for Systems Biology, Seattle, WA 98109, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
7
|
Lathika Rajendrakumar A, Arbeev KG, Bagley O, Duan M, Yashin AI, Ukraintseva S, for the Alzheimer’s Disease Neuroimaging Initiative. APOE4 and infectious diseases jointly contribute to brain glucose hypometabolism, a biomarker of Alzheimer's pathology: New findings from the ADNI. PLoS One 2025; 20:e0316808. [PMID: 39774485 PMCID: PMC11706463 DOI: 10.1371/journal.pone.0316808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
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, Durham, North Carolina, United States of America
| | - Konstantin G. Arbeev
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, North Carolina, United States of America
| | - Olivia Bagley
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, North Carolina, United States of America
| | - Matt Duan
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, North Carolina, United States of America
| | - Anatoliy I. Yashin
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, North Carolina, United States of America
| | - Svetlana Ukraintseva
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, North Carolina, United States of America
| | | |
Collapse
|
8
|
Roberts H, Fang Y, Quinn K, Hill T, Peck MR, Bartke A, Hascup KN, Hascup ER. Lifespan of male and female APP/PS1 and APP NL-F/NL-F mouse models of Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.15.618508. [PMID: 39464050 PMCID: PMC11507819 DOI: 10.1101/2024.10.15.618508] [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: 10/29/2024]
Abstract
Alzheimer's disease (AD) disproportionately affects women, yet most preclinical research studies are male-centric. We performed lifespan analyses of male and female AD mouse models (APP/PS1 and APPNL-F/NL-F) and their shared genetic background control (C57BL/6). Survival curves support significant sex differences between within genotypes. Minimal longevity revealed increased age in male APP/PS1, and decreased age in APPNL-F/NL-F mice. Maximal longevity revealed an increased average age in males. Furthermore, median lifespan differed between sex and genotype. This study supports sexual dimorphic survival in two mouse models of AD, emphasizing the need to examine mechanisms and treatments in both sexes.
Collapse
Affiliation(s)
- Hannah Roberts
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Dept of Neurology; Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Yimin Fang
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Dept of Neurology; Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Kathleen Quinn
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Dept of Neurology; Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Tiarra Hill
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Dept of Neurology; Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Mackenzie R. Peck
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Dept of Neurology; Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Andrzej Bartke
- Dept of Internal Medicine; Southern Illinois University School of Medicine, Springfield, IL, USA
- Dept of Medical Microbiology, Immunology and Cell Biology; Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Kevin N. Hascup
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Dept of Neurology; Southern Illinois University School of Medicine, Springfield, IL, USA
- Dept of Medical Microbiology, Immunology and Cell Biology; Southern Illinois University School of Medicine, Springfield, IL, USA
- Dept of Pharmacology; Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Erin R. Hascup
- Neuroscience Institute, Dale and Deborah Smith Center for Alzheimer’s Research and Treatment, Dept of Neurology; Southern Illinois University School of Medicine, Springfield, IL, USA
- Dept of Pharmacology; Southern Illinois University School of Medicine, Springfield, IL, USA
| |
Collapse
|
9
|
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
|
10
|
Bischoff-Grethe A, Stoner SA, Riley EP, Moore EM. Subcortical volume in middle-aged adults with fetal alcohol spectrum disorders. Brain Commun 2024; 6:fcae273. [PMID: 39229493 PMCID: PMC11369821 DOI: 10.1093/braincomms/fcae273] [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: 01/06/2024] [Revised: 05/06/2024] [Accepted: 08/28/2024] [Indexed: 09/05/2024] Open
Abstract
Studies of youth and young adults with prenatal alcohol exposure (PAE) have most consistently reported reduced volumes of the corpus callosum, cerebellum and subcortical structures. However, it is unknown whether this continues into middle adulthood or if individuals with PAE may experience premature volumetric decline with aging. Forty-eight individuals with fetal alcohol spectrum disorders (FASD) and 28 healthy comparison participants aged 30 to 65 participated in a 3T MRI session that resulted in usable T1-weighted and T2-weighted structural images. Primary analyses included volumetric measurements of the caudate, putamen, pallidum, cerebellum and corpus callosum using FreeSurfer software. Analyses were conducted examining both raw volumetric measurements and subcortical volumes adjusted for overall intracranial volume (ICV). Models tested for main effects of age, sex and group, as well as interactions of group with age and group with sex. We found the main effects for group; all regions were significantly smaller in participants with FASD for models using raw volumes (P's < 0.001) as well as for models using volumes adjusted for ICV (P's < 0.046). Although there were no significant interactions of group with age, females with FASD had smaller corpus callosum volumes relative to both healthy comparison females and males with FASD (P's < 0.001). As seen in children and adolescents, adults aged 30 to 65 with FASD showed reduced volumes of subcortical structures relative to healthy comparison adults, suggesting persistent impact of PAE. Moreover, the observed volumetric reduction of the corpus callosum in females with FASD could suggest more rapid degeneration, which may have implications for cognition as these individuals continue to age.
Collapse
Affiliation(s)
| | - Susan A Stoner
- Department of Psychiatry and Behavioral Sciences, Fetal Alcohol and Drug Unit, University of Washington School of Medicine, Seattle, Washington 98105, USA
| | - Edward P Riley
- Department of Psychology, Center for Behavioral Teratology, San Diego State University, San Diego, CA, 92120, USA
| | - Eileen M Moore
- Department of Psychology, Center for Behavioral Teratology, San Diego State University, San Diego, CA, 92120, USA
| |
Collapse
|
11
|
Moon HJ, Luo Y, Chugh D, Zhao L. Human apolipoprotein E glycosylation and sialylation: from structure to function. Front Mol Neurosci 2024; 17:1399965. [PMID: 39169951 PMCID: PMC11335735 DOI: 10.3389/fnmol.2024.1399965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 06/28/2024] [Indexed: 08/23/2024] Open
Abstract
Human apolipoprotein E (ApoE) was first identified as a polymorphic gene in the 1970s; however, the genetic association of ApoE genotypes with late-onset sporadic Alzheimer's disease (sAD) was only discovered 20 years later. Since then, intensive research has been undertaken to understand the molecular effects of ApoE in the development of sAD. Despite three decades' worth of effort and over 10,000 papers published, the greatest mystery in the ApoE field remains: human ApoE isoforms differ by only one or two amino acid residues; what is responsible for their significantly distinct roles in the etiology of sAD, with ApoE4 conferring the greatest genetic risk for sAD whereas ApoE2 providing exceptional neuroprotection against sAD. Emerging research starts to point to a novel and compelling hypothesis that the sialoglycans posttranslationally appended to human ApoE may serve as a critical structural modifier that alters the biology of ApoE, leading to the opposing impacts of ApoE isoforms on sAD and likely in the peripheral systems as well. ApoE has been shown to be posttranslationally glycosylated in a species-, tissue-, and cell-specific manner. Human ApoE, particularly in brain tissue and cerebrospinal fluid (CSF), is highly glycosylated, and the glycan chains are exclusively attached via an O-linkage to serine or threonine residues. Moreover, studies have indicated that human ApoE glycans undergo sialic acid modification or sialylation, a structural alteration found to be more prominent in ApoE derived from the brain and CSF than plasma. However, whether the sialylation modification of human ApoE has a biological role is largely unexplored. Our group recently first reported that the three major isoforms of human ApoE in the brain undergo varying degrees of sialylation, with ApoE2 exhibiting the most abundant sialic acid modification, whereas ApoE4 is the least sialylated. Our findings further indicate that the sialic acid moiety on human ApoE glycans may serve as a critical modulator of the interaction of ApoE with amyloid β (Aβ) and downstream Aβ pathogenesis, a prominent pathologic feature in AD. In this review, we seek to provide a comprehensive summary of this exciting and rapidly evolving area of ApoE research, including the current state of knowledge and opportunities for future exploration.
Collapse
Affiliation(s)
- Hee-Jung Moon
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, United States
| | - Yan Luo
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, United States
| | - Diksha Chugh
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, United States
| | - Liqin Zhao
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, United States
- Neuroscience Graduate Program, University of Kansas, Lawrence, KS, United States
| |
Collapse
|
12
|
Jacob SM, Lee S, Kim SH, Sharkey KA, Pfeffer G, Nguyen MD. Brain-body mechanisms contribute to sexual dimorphism in amyotrophic lateral sclerosis. Nat Rev Neurol 2024; 20:475-494. [PMID: 38965379 DOI: 10.1038/s41582-024-00991-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2024] [Indexed: 07/06/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is the most common form of human motor neuron disease. It is characterized by the progressive degeneration of upper and lower motor neurons, leading to generalized motor weakness and, ultimately, respiratory paralysis and death within 3-5 years. The disease is shaped by genetics, age, sex and environmental stressors, but no cure or routine biomarkers exist for the disease. Male individuals have a higher propensity to develop ALS, and a different manifestation of the disease phenotype, than female individuals. However, the mechanisms underlying these sex differences remain a mystery. In this Review, we summarize the epidemiology of ALS, examine the sexually dimorphic presentation of the disease and highlight the genetic variants and molecular pathways that might contribute to sex differences in humans and animal models of ALS. We advance the idea that sexual dimorphism in ALS arises from the interactions between the CNS and peripheral organs, involving vascular, metabolic, endocrine, musculoskeletal and immune systems, which are strikingly different between male and female individuals. Finally, we review the response to treatments in ALS and discuss the potential to implement future personalized therapeutic strategies for the disease.
Collapse
Affiliation(s)
- Sarah M Jacob
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Sukyoung Lee
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Seung Hyun Kim
- Department of Neurology, Hanyang University Hospital, Seoul, South Korea
| | - Keith A Sharkey
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Gerald Pfeffer
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
| | - Minh Dang Nguyen
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
- Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
13
|
Singhaarachchi PH, Antal P, Calon F, Culmsee C, Delpech JC, Feldotto M, Geertsema J, Hoeksema EE, Korosi A, Layé S, McQualter J, de Rooij SR, Rummel C, Slayo M, Sominsky L, Spencer SJ. Aging, sex, metabolic and life experience factors: Contributions to neuro-inflammaging in Alzheimer's disease research. Neurosci Biobehav Rev 2024; 162:105724. [PMID: 38762130 DOI: 10.1016/j.neubiorev.2024.105724] [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: 02/28/2024] [Revised: 04/24/2024] [Accepted: 05/12/2024] [Indexed: 05/20/2024]
Abstract
Alzheimer's disease (AD) is prevalent around the world, yet our understanding of the disease is still very limited. Recent work suggests that the cornerstone of AD may include the inflammation that accompanies it. Failure of a normal pro-inflammatory immune response to resolve may lead to persistent central inflammation that contributes to unsuccessful clearance of amyloid-beta plaques as they form, neuronal death, and ultimately cognitive decline. Individual metabolic, and dietary (lipid) profiles can differentially regulate this inflammatory process with aging, obesity, poor diet, early life stress and other inflammatory factors contributing to a greater risk of developing AD. Here, we integrate evidence for the interface between these factors, and how they contribute to a pro-inflammatory brain milieu. In particular, we discuss the importance of appropriate polyunsaturated fatty acids (PUFA) in the diet for the metabolism of specialised pro-resolving mediators (SPMs); raising the possibility for dietary strategies to improve AD outlook.
Collapse
Affiliation(s)
| | - Peter Antal
- Department of Measurement and Information Systems, Budapest University of Technology and Economics, 1111, Hungary
| | - Frédéric Calon
- Faculty of Pharmacy, Centre de Recherche du CHU de Québec-Laval University, Quebec G1V0A6, Canada; International Associated Laboratory OptiNutriBrain-NutriNeuro, Bordeaux F-33000, France; INAF, Quebec G1V0A6, Canada
| | - Carsten Culmsee
- Institute of Pharmacology and Clinical Pharmacy, Philipps University of Marburg, Marburg D-35032, Germany; Center for Mind, Brain and Behavior-CMBB, Giessen, D-35392, Marburg D-35032, Germany
| | - Jean-Christophe Delpech
- International Associated Laboratory OptiNutriBrain-NutriNeuro, Bordeaux F-33000, France; Université de Bordeaux, INRAE, Bordeaux INP, NutriNeurO, UMR 1286, Bordeaux F-33000, France; INAF, Quebec G1V0A6, Canada
| | - Martin Feldotto
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University Giessen, Giessen D-35392, Germany
| | - Jorine Geertsema
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam 1018, the Netherlands
| | - Emmy E Hoeksema
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam 1018, the Netherlands
| | - Aniko Korosi
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam 1018, the Netherlands
| | - Sophie Layé
- International Associated Laboratory OptiNutriBrain-NutriNeuro, Bordeaux F-33000, France; Université de Bordeaux, INRAE, Bordeaux INP, NutriNeurO, UMR 1286, Bordeaux F-33000, France; INAF, Quebec G1V0A6, Canada
| | - Jonathan McQualter
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Melbourne, Victoria 3083, Australia
| | - Susanne R de Rooij
- Department of Epidemiology and Data Science, Amsterdam University Medical Centers, University of Amsterdam, 1018, the Netherlands
| | - Christoph Rummel
- Center for Mind, Brain and Behavior-CMBB, Giessen, D-35392, Marburg D-35032, Germany; Institute of Veterinary Physiology and Biochemistry, Justus Liebig University Giessen, Giessen D-35392, Germany
| | - Mary Slayo
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Melbourne, Victoria 3083, Australia; Center for Mind, Brain and Behavior-CMBB, Giessen, D-35392, Marburg D-35032, Germany; Institute of Veterinary Physiology and Biochemistry, Justus Liebig University Giessen, Giessen D-35392, Germany
| | - Luba Sominsky
- Barwon Health, Geelong, Victoria 3220, Australia; IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria 3217, Australia
| | - Sarah J Spencer
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Melbourne, Victoria 3083, Australia.
| |
Collapse
|
14
|
Gladding JM, Rafiei N, Mitchell CS, Begg DP. Excision of the endothelial blood-brain barrier insulin receptor does not alter spatial cognition in mice fed either a chow or high-fat diet. Neurobiol Learn Mem 2024; 212:107938. [PMID: 38772444 DOI: 10.1016/j.nlm.2024.107938] [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: 12/21/2023] [Revised: 05/02/2024] [Accepted: 05/15/2024] [Indexed: 05/23/2024]
Abstract
Insulin is transported across the blood-brain barrier (BBB) endothelium to regulate aspects of metabolism and cognition. Brain insulin resistance often results from high-fat diet (HFD) consumption and is thought to contribute to spatial cognition deficits. To target BBB insulin function, we used Cre-LoxP genetic excision of the insulin receptor (InsR) from endothelial cells in adult male mice. We hypothesized that this excision would impair spatial cognition, and that high-fat diet consumption would exacerbate these effects. Excision of the endothelial InsR did not impair performance in two spatial cognition tasks, the Y-Maze and Morris Water Maze, in tests held both before and after 14 weeks of access to high-fat (or chow control) diet. The HFD increased body weight gain and induced glucose intolerance but did not impair spatial cognition. Endothelial InsR excision tended to increase body weight and reduce sensitivity to peripheral insulin, but these metabolic effects were not associated with impairments to spatial cognition and did not interact with HFD exposure. Instead, all mice showed intact spatial cognitive performance regardless of whether they had been fed chow or a HFD, and whether the InsR had been excised or not. Overall, the results indicate that loss of the endothelial InsR does not impact spatial cognition, which is in line with pharmacological evidence that other mechanisms at the BBB facilitate insulin transport and allow it to exert its pro-cognitive effects.
Collapse
Affiliation(s)
- Joanne M Gladding
- School of Psychology, Faculty of Science, University of New South Wales, Australia.
| | - Neda Rafiei
- School of Psychology, Faculty of Science, University of New South Wales, Australia
| | - Caitlin S Mitchell
- School of Psychology, Faculty of Science, University of New South Wales, Australia
| | - Denovan P Begg
- School of Psychology, Faculty of Science, University of New South Wales, Australia
| |
Collapse
|
15
|
Lopez-Lee C, Torres ERS, Carling G, Gan L. Mechanisms of sex differences in Alzheimer's disease. Neuron 2024; 112:1208-1221. [PMID: 38402606 PMCID: PMC11076015 DOI: 10.1016/j.neuron.2024.01.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 11/01/2023] [Accepted: 01/23/2024] [Indexed: 02/27/2024]
Abstract
Alzheimer's disease (AD) and the mechanisms underlying its etiology and progression are complex and multifactorial. The higher AD risk in women may serve as a clue to better understand these complicated processes. In this review, we examine aspects of AD that demonstrate sex-dependent effects and delve into the potential biological mechanisms responsible, compiling findings from advanced technologies such as single-cell RNA sequencing, metabolomics, and multi-omics analyses. We review evidence that sex hormones and sex chromosomes interact with various disease mechanisms during aging, encompassing inflammation, metabolism, and autophagy, leading to unique characteristics in disease progression between men and women.
Collapse
Affiliation(s)
- Chloe Lopez-Lee
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA; Neuroscience Graduate Program, Weill Cornell Medicine, New York, NY, USA
| | - Eileen Ruth S Torres
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Gillian Carling
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA; Neuroscience Graduate Program, Weill Cornell Medicine, New York, NY, USA
| | - Li Gan
- Helen and Robert Appel Alzheimer's Disease Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA; Neuroscience Graduate Program, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
16
|
Cleland NRW, Potter GJ, Buck C, Quang D, Oldham D, Neal M, Saviola A, Niemeyer CS, Dobrinskikh E, Bruce KD. Altered metabolism and DAM-signatures in female brains and microglia with aging. Brain Res 2024; 1829:148772. [PMID: 38244754 PMCID: PMC12036313 DOI: 10.1016/j.brainres.2024.148772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 12/21/2023] [Accepted: 01/15/2024] [Indexed: 01/22/2024]
Abstract
Despite Alzheimer's disease (AD) disproportionately affecting women, the mechanisms remain elusive. In AD, microglia undergo 'metabolic reprogramming', which contributes to microglial dysfunction and AD pathology. However, how sex and age contribute to metabolic reprogramming in microglia is understudied. Here, we use metabolic imaging, transcriptomics, and metabolic assays to probe age- and sex-associated changes in brain and microglial metabolism. Glycolytic and oxidative metabolism in the whole brain was determined using Fluorescence Lifetime Imaging Microscopy (FLIM). Young female brains appeared less glycolytic than male brains, but with aging, the female brain became 'male-like.' Transcriptomic analysis revealed increased expression of disease-associated microglia (DAM) genes (e.g., ApoE, Trem2, LPL), and genes involved in glycolysis and oxidative metabolism in microglia from aged females compared to males. To determine whether estrogen can alter the expression of these genes, BV-2 microglia-like cell lines, which abundantly express DAM genes, were supplemented with 17β-estradiol (E2). E2 supplementation resulted in reduced expression of DAM genes, reduced lipid and cholesterol transport, and substrate-dependent changes in glycolysis and oxidative metabolism. Consistent with the notion that E2 may suppress DAM-associated factors, LPL activity was elevated in the brains of aged female mice. Similarly, DAM gene and protein expression was higher in monocyte-derived microglia-like (MDMi) cells derived from middle-aged females compared to age-matched males and was responsive to E2 supplementation. FLIM analysis of MDMi from young and middle-aged females revealed reduced oxidative metabolism and FAD+ with age. Overall, our findings show that altered metabolism defines age-associated changes in female microglia and suggest that estrogen may inhibit the expression and activity of DAM-associated factors, which may contribute to increased AD risk, especially in post-menopausal women.
Collapse
Affiliation(s)
- Nicholas R W Cleland
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Garrett J Potter
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Courtney Buck
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Daphne Quang
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Dean Oldham
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Mikaela Neal
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Anthony Saviola
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Christy S Niemeyer
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Evgenia Dobrinskikh
- Section of Neonatology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, USA
| | - Kimberley D Bruce
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
17
|
Friese S, Ranzini G, Tuchtenhagen M, Lossow K, Hertel B, Pohl G, Ebert F, Bornhorst J, Kipp AP, Schwerdtle T. Long-term suboptimal dietary trace element supply does not affect trace element homeostasis in murine cerebellum. Metallomics 2024; 16:mfae003. [PMID: 38299785 PMCID: PMC10873500 DOI: 10.1093/mtomcs/mfae003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 12/14/2023] [Indexed: 02/02/2024]
Abstract
The ageing process is associated with alterations of systemic trace element (TE) homeostasis increasing the risk, e.g. neurodegenerative diseases. Here, the impact of long-term modulation of dietary intake of copper, iron, selenium, and zinc was investigated in murine cerebellum. Four- and 40-wk-old mice of both sexes were supplied with different amounts of those TEs for 26 wk. In an adequate supply group, TE concentrations were in accordance with recommendations for laboratory mice while suboptimally supplied animals received only limited amounts of copper, iron, selenium, and zinc. An additional age-adjusted group was fed selenium and zinc in amounts exceeding recommendations. Cerebellar TE concentrations were measured by inductively coupled plasma-tandem mass spectrometry. Furthermore, the expression of genes involved in TE transport, DNA damage response, and DNA repair as well as selected markers of genomic stability [8-oxoguanine, incision efficiency toward 8-oxoguanine, 5-hydroxyuracil, and apurinic/apyrimidinic sites and global DNA (hydroxy)methylation] were analysed. Ageing resulted in a mild increase of iron and copper concentrations in the cerebellum, which was most pronounced in the suboptimally supplied groups. Thus, TE changes in the cerebellum were predominantly driven by age and less by nutritional intervention. Interestingly, deviation from adequate TE supply resulted in higher manganese concentrations of female mice even though the manganese supply itself was not modulated. Parameters of genomic stability were neither affected by age, sex, nor diet. Overall, this study revealed that suboptimal dietary TE supply does not substantially affect TE homeostasis in the murine cerebellum.
Collapse
Affiliation(s)
- Sharleen Friese
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
- TraceAge—DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, Germany
| | - Giovanna Ranzini
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
| | - Max Tuchtenhagen
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
- TraceAge—DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, Germany
| | - Kristina Lossow
- TraceAge—DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, Germany
- Nutritional Physiology, Institute of Nutritional Sciences, Friedrich Schiller University Jena, Dornburger Str. 24, 07743 Jena, Germany
| | - Barbara Hertel
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
| | - Gabriele Pohl
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
| | - Franziska Ebert
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
| | - Julia Bornhorst
- TraceAge—DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, Germany
- Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Gaußstraße 20, 42119 Wuppertal, Germany
| | - Anna Patricia Kipp
- TraceAge—DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, Germany
- Nutritional Physiology, Institute of Nutritional Sciences, Friedrich Schiller University Jena, Dornburger Str. 24, 07743 Jena, Germany
| | - Tanja Schwerdtle
- Department of Food Chemistry, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
- TraceAge—DFG Research Unit on Interactions of Essential Trace Elements in Healthy and Diseased Elderly (FOR 2558), Berlin-Potsdam-Jena-Wuppertal, Germany
- German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Str. 8-10, 10589 Berlin, Germany
| |
Collapse
|
18
|
Cleland NRW, Potter GJ, Buck C, Quang D, Oldham D, Neal M, Saviola A, Niemeyer CS, Dobrinskikh E, Bruce KD. Altered Metabolism and DAM-signatures in Female Brains and Microglia with Aging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.28.569104. [PMID: 38076915 PMCID: PMC10705419 DOI: 10.1101/2023.11.28.569104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
Despite Alzheimer's disease (AD) disproportionately affecting women, the mechanisms remain elusive. In AD, microglia undergo 'metabolic reprogramming', which contributes to microglial dysfunction and AD pathology. However, how sex and age contribute to metabolic reprogramming in microglia is understudied. Here, we use metabolic imaging, transcriptomics, and metabolic assays to probe age-and sex-associated changes in brain and microglial metabolism. Glycolytic and oxidative metabolism in the whole brain was determined using Fluorescence Lifetime Imaging Microscopy (FLIM). Young female brains appeared less glycolytic than male brains, but with aging, the female brain became 'male-like.' Transcriptomic analysis revealed increased expression of disease-associated microglia (DAM) genes (e.g., ApoE, Trem2, LPL), and genes involved in glycolysis and oxidative metabolism in microglia from aged females compared to males. To determine whether estrogen can alter the expression of these genes, BV-2 microglia-like cell lines, which abundantly express DAM genes, were supplemented with 17β-estradiol (E2). E2 supplementation resulted in reduced expression of DAM genes, reduced lipid and cholesterol transport, and substrate-dependent changes in glycolysis and oxidative metabolism. Consistent with the notion that E2 may suppress DAM-associated factors, LPL activity was elevated in the brains of aged female mice. Similarly, DAM gene and protein expression was higher in monocyte-derived microglia-like (MDMi) cells derived from middle-aged females compared to age-matched males and was responsive to E2 supplementation. FLIM analysis of MDMi from young and middle-aged females revealed reduced oxidative metabolism and FAD+ with age. Overall, our findings show that altered metabolism defines age-associated changes in female microglia and suggest that estrogen may inhibit the expression and activity of DAM-associated factors, which may contribute to increased AD risk, especially in post-menopausal women.
Collapse
Affiliation(s)
- Nicholas R W Cleland
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - Garrett J Potter
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - Courtney Buck
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - Daphne Quang
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - Dean Oldham
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - Mikaela Neal
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - Anthony Saviola
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - Christy S. Niemeyer
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Evgenia Dobrinskikh
- Section of Neonatology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, USA
| | - Kimberley D. Bruce
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| |
Collapse
|
19
|
Jeong H, Pan Y, Akhter F, Volkow ND, Zhu D, Du C. Impairment of cerebral vascular reactivity and resting blood flow in early-staged transgenic AD mice: in vivo optical imaging studies. RESEARCH SQUARE 2023:rs.3.rs-3579916. [PMID: 37987006 PMCID: PMC10659553 DOI: 10.21203/rs.3.rs-3579916/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Background Alzheimer's disease (AD) is a neurodegenerative disorder with progressive cognitive decline in aging individuals that poses a significant challenge to patients due to an incomplete understanding of its etiology and lack of effective interventions. While "the Amyloid Cascade Hypothesis," the abnormal accumulation of amyloid-β in the brain, has been the most prevalent theory for AD, mounting evidence from clinical and epidemiological studies suggest that defects in cerebral vessels and hypoperfusion appear prior to other pathological manifestations and might contribute to AD, leading to "the Vascular Hypothesis." However, assessment of structural and functional integrity of the cerebral vasculature in vivo in the brain from AD rodent models has been challenging owing to the limited spatiotemporal resolution of conventional imaging technologies. Methods We employed two in vivo imaging technologies, i.e., Dual-Wavelength Imaging (DWI) and Optical Coherence Tomography (OCT), to evaluate cerebrovascular reactivity (CVR; responsiveness of blood vessels to vasoconstriction as triggered by cocaine) in a relatively large field of view of the cortex in vivo, and 3D quantitative cerebrovascular blood flow (CBF) imaging in living transgenic AD mice at single vessel resolution. Results Our results showed significantly impaired CVR and reduced CBF in basal state in transgenic AD mice compared to non-transgenic littermates in an early stage of AD progression. Changes in total hemoglobin (Δ[HbT]) in response to vasoconstriction were significantly attenuated in AD mice, especially in arteries and tissue, and the recovery time of Δ[HbT] after vasoconstriction was shorter for AD than WT in all types of vessels and cortical tissue, thereby indicating hypoperfusion and reduced vascular flexibility. Additionally, our 3D OCT images revealed that CBF velocities in arteries were slower and that the microvascular network was severely disrupted in the brain of AD mice. Conclusions These results suggest significant vascular impairment in basal CBF and dynamic CVR in the neurovascular network in a rodent model of AD at an early stage of the disease. These cutting-edge in vivo optical imaging tools offer an innovative venue for detecting early neurovascular dysfunction in relation to AD pathology and pave the way for clinical translation of early diagnosis and elucidation of AD pathogenesis in the future.
Collapse
Affiliation(s)
- Hyomin Jeong
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794, USA
| | - Yingtian Pan
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794, USA
| | - Firoz Akhter
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794, USA
| | - Nora D. Volkow
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20857, USA
| | - Donghui Zhu
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794, USA
| | - Congwu Du
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794, USA
| |
Collapse
|
20
|
Balu D, Valencia-Olvera AC, Nguyen A, Patnam M, York J, Peri F, Neumann F, LaDu MJ, Tai LM. A small-molecule TLR4 antagonist reduced neuroinflammation in female E4FAD mice. Alzheimers Res Ther 2023; 15:181. [PMID: 37858252 PMCID: PMC10585767 DOI: 10.1186/s13195-023-01330-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 10/10/2023] [Indexed: 10/21/2023]
Abstract
BACKGROUND APOE genotype is the greatest genetic risk factor for sporadic Alzheimer's disease (AD). APOE4 increases AD risk up to 12-fold compared to APOE3, an effect that is greater in females. Evidence suggests that one-way APOE could modulate AD risk and progression through neuroinflammation. Indeed, APOE4 is associated with higher glial activation and cytokine levels in AD patients and mice. Therefore, identifying pathways that contribute to APOE4-associated neuroinflammation is an important approach for understanding and treating AD. Human and in vivo evidence suggests that TLR4, one of the key receptors involved in the innate immune system, could be involved in APOE-modulated neuroinflammation. Consistent with that idea, we previously demonstrated that the TLR4 antagonist IAXO-101 can reduce LPS- and Aβ-induced cytokine secretion in APOE4 glial cultures. Therefore, the goal of this study was to advance these findings and determine whether IAXO-101 can modulate neuroinflammation, Aβ pathology, and behavior in mice that express APOE4. METHODS We used mice that express five familial AD mutations and human APOE3 (E3FAD) or APOE4 (E4FAD). Female and male E4FAD mice and female E3FAD mice were treated with vehicle or IAXO-101 in two treatment paradigms: prevention from 4 to 6 months of age or reversal from 6 to 7 months of age. Learning and memory were assessed by modified Morris water maze. Aβ deposition, fibrillar amyloid deposition, astrogliosis, and microgliosis were assessed by immunohistochemistry. Soluble levels of Aβ and apoE, insoluble levels of apoE and Aβ, and IL-1β were measured by ELISA. RESULTS IAXO-101 treatment resulted in lower Iba-1 coverage, lower number of reactive microglia, and improved memory in female E4FAD mice in both prevention and reversal paradigms. IAXO-101-treated male E4FAD mice also had lower Iba-1 coverage and reactivity in the RVS paradigm, but there was no effect on behavior. There was also no effect of IAXO-101 treatment on neuroinflammation and behavior in female E3FAD mice. CONCLUSION Our data supports that TLR4 is a potential mechanistic therapeutic target for modulating neuroinflammation and cognition in APOE4 females.
Collapse
Affiliation(s)
- Deebika Balu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Ana C Valencia-Olvera
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Austin Nguyen
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Mehul Patnam
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Jason York
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Francesco Peri
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | | | - Mary Jo LaDu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Leon M Tai
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
21
|
Baumgartner NE, Biraud MC, Lucas EK. Sex differences in socioemotional behavior and changes in ventral hippocampal transcription across aging in C57Bl/6J mice. Neurobiol Aging 2023; 130:141-153. [PMID: 37524006 PMCID: PMC10629502 DOI: 10.1016/j.neurobiolaging.2023.05.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 05/01/2023] [Accepted: 05/02/2023] [Indexed: 08/02/2023]
Abstract
Socioemotional health is positively correlated with improved cognitive and physical aging. Despite known sex differences in socioemotional behaviors and the trajectory of aging, the interactive effects between sex and aging on socioemotional outcomes are poorly understood. We performed the most comprehensive assessment of sex differences in socioemotional behaviors in C57Bl/6J mice across aging to date. Compared to males, females exhibited decreased anxiety-like behavior and social preference but increased social recognition. With age, anxiety-like behavior, cued threat memory generalization, and social preference increased in both sexes. To investigate potential neural mechanisms underlying these behavioral changes, we analyzed transcriptional neuropathology markers in the ventral hippocampus and found age-related changes in genes related to activated microglia, angiogenesis, and cytokines. Sex differences emerged in the timing, direction, and magnitude of these changes, independent of reproductive senescence in aged females. Interestingly, female-specific upregulation of autophagy-related genes correlated with age-related behavioral changes selectively in females. These novel findings reveal critical sex differences in trajectories of ventral hippocampal aging that may contribute to sex- and age-related differences in socioemotional outcomes.
Collapse
Affiliation(s)
- Nina E Baumgartner
- Department of Psychiatry and Behavioral Neurobiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA; Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA
| | - Mandy C Biraud
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA
| | - Elizabeth K Lucas
- Department of Psychiatry and Behavioral Neurobiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA; Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA.
| |
Collapse
|
22
|
Raheel K, Deegan G, Di Giulio I, Cash D, Ilic K, Gnoni V, Chaudhuri KR, Drakatos P, Moran R, Rosenzweig I. Sex differences in alpha-synucleinopathies: a systematic review. Front Neurol 2023; 14:1204104. [PMID: 37545736 PMCID: PMC10398394 DOI: 10.3389/fneur.2023.1204104] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 06/13/2023] [Indexed: 08/08/2023] Open
Abstract
Background Past research indicates a higher prevalence, incidence, and severe clinical manifestations of alpha-synucleinopathies in men, leading to a suggestion of neuroprotective properties of female sex hormones (especially estrogen). The potential pathomechanisms of any such effect on alpha-synucleinopathies, however, are far from understood. With that aim, we undertook to systematically review, and to critically assess, contemporary evidence on sex and gender differences in alpha-synucleinopathies using a bench-to-bedside approach. Methods In this systematic review, studies investigating sex and gender differences in alpha-synucleinopathies (Rapid Eye Movement (REM) Behavior Disorder (RBD), Parkinson's Disease (PD), Dementia with Lewy Bodies (DLB), Multiple System Atrophy (MSA)) from 2012 to 2022 were identified using electronic database searches of PubMed, Embase and Ovid. Results One hundred sixty-two studies were included; 5 RBD, 6 MSA, 20 DLB and 131 PD studies. Overall, there is conclusive evidence to suggest sex-and gender-specific manifestation in demographics, biomarkers, genetics, clinical features, interventions, and quality of life in alpha-synucleinopathies. Only limited data exists on the effects of distinct sex hormones, with majority of studies concentrating on estrogen and its speculated neuroprotective effects. Conclusion Future studies disentangling the underlying sex-specific mechanisms of alpha-synucleinopathies are urgently needed in order to enable novel sex-specific therapeutics.
Collapse
Affiliation(s)
- Kausar Raheel
- Sleep and Brain Plasticity Centre, Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, United Kingdom
| | - Gemma Deegan
- Sleep and Brain Plasticity Centre, Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, United Kingdom
- BRAIN, Imaging Centre, CNS, King’s College London, London, United Kingdom
| | - Irene Di Giulio
- Sleep and Brain Plasticity Centre, Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, United Kingdom
- School of Basic and Medical Biosciences, Faculty of Life Science and Medicine, King’s College London, London, United Kingdom
| | - Diana Cash
- Sleep and Brain Plasticity Centre, Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, United Kingdom
- BRAIN, Imaging Centre, CNS, King’s College London, London, United Kingdom
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, United Kingdom
| | - Katarina Ilic
- Sleep and Brain Plasticity Centre, Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, United Kingdom
- BRAIN, Imaging Centre, CNS, King’s College London, London, United Kingdom
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, United Kingdom
| | - Valentina Gnoni
- Sleep and Brain Plasticity Centre, Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, United Kingdom
- Center for Neurodegenerative Diseases and the Aging Brain, University of Bari Aldo Moro, Lecce, Italy
| | - K. Ray Chaudhuri
- Movement Disorders Unit, King’s College Hospital and Department of Clinical and Basic Neurosciences, Institute of Psychiatry, Psychology and Neuroscience and Parkinson Foundation Centre of Excellence, King’s College London, London, United Kingdom
| | - Panagis Drakatos
- School of Basic and Medical Biosciences, Faculty of Life Science and Medicine, King’s College London, London, United Kingdom
- Sleep Disorders Centre, Guy’s and St Thomas’ NHS Foundation Trust, London, United Kingdom
| | - Rosalyn Moran
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, United Kingdom
| | - Ivana Rosenzweig
- Sleep and Brain Plasticity Centre, Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, United Kingdom
- Sleep Disorders Centre, Guy’s and St Thomas’ NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
23
|
Stickel AM, Tarraf W, González KA, Ivanovic V, Morlett Paredes A, Zeng D, Cai J, Isasi CR, Kaplan R, Lipton RB, Daviglus M, Testai FD, Lamar M, Gallo LC, Talavera GA, Gellman MD, Ramos AR, González HM, DeCarli C. Characterizing age- and sex-related differences in brain structure among middle-aged and older Hispanic/Latino adults in the study of Latinos- investigation of neurocognitive aging magnetic resonance imaging (SOL-INCA MRI). Neurobiol Aging 2023; 126:58-66. [PMID: 36933278 PMCID: PMC10363333 DOI: 10.1016/j.neurobiolaging.2023.02.007] [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: 08/29/2022] [Revised: 02/15/2023] [Accepted: 02/15/2023] [Indexed: 02/25/2023]
Abstract
Hispanic/Latino adults are a growing segment of the older U.S. population yet are underrepresented in brain aging research. We aimed to characterize brain aging among diverse Hispanic/Latino individuals. Hispanic/Latino individuals (unweighted n = 2273 ages 35-85 years; 56% female) from the Hispanic Community Health Study/Study of Latinos (HCHS/SOL) population-based study underwent magnetic resonance imaging (MRI) as part of the SOL- Investigation of Neurocognitive Aging MRI (SOL-INCA-MRI) ancillary study (2018-2022). We performed linear regressions to calculate age associations with brain volumes for each outcome (total (global) brain, hippocampal, lateral ventricle, total white matter hyperintensity (WMH), individual cortical lobar, and total cortical gray matter) and tested modification by sex. Older age was associated with smaller gray matter volumes and larger lateral ventricle and WMH volumes. Age-related differences in global brain volumes and gray matter volumes in specific regions (i.e., the hippocampus and temporal and occipital lobes) were less pronounced among women. Our findings warrant further investigation into sex-specific mechanisms of brain aging using longitudinal studies.
Collapse
Affiliation(s)
- Ariana M Stickel
- Department of Neurosciences and Shiley-Marcos Alzheimer's Disease Research Center, University of California, San Diego, La Jolla, CA, USA; Department of Psychology, San Diego State University, San Diego, CA, USA
| | - Wassim Tarraf
- Institute of Gerontology & Department of Healthcare Sciences, Wayne State University, Detroit, MI, USA
| | - Kevin A González
- Department of Neurosciences and Shiley-Marcos Alzheimer's Disease Research Center, University of California, San Diego, La Jolla, CA, USA
| | - Vladamir Ivanovic
- Department of Neurology and Center for Neuroscience, University of California at Davis, Davis, CA, USA
| | - Alejandra Morlett Paredes
- Department of Neurosciences and Shiley-Marcos Alzheimer's Disease Research Center, University of California, San Diego, La Jolla, CA, USA
| | - Donglin Zeng
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC, USA
| | - Jianwen Cai
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC, USA
| | - Carmen R Isasi
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine, The Bronx, NY, USA
| | - Robert Kaplan
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine, The Bronx, NY, USA
| | - Richard B Lipton
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine, The Bronx, NY, USA; Department of Neurology, Albert Einstein College of Medicine, The Bronx, NY, USA
| | - Martha Daviglus
- Institute for Minority Health Research, University of Illinois at Chicago, College of Medicine, Chicago, IL, USA
| | - Fernando D Testai
- Department of Neurology & Neurorehabilitation, University of Illinois at Chicago, College of Medicine, Chicago, IL, USA
| | - Melissa Lamar
- Institute for Minority Health Research, University of Illinois at Chicago, College of Medicine, Chicago, IL, USA; Rush Alzheimer's Disease Research Center, Rush University Medical Center, Chicago, IL, USA
| | - Linda C Gallo
- Department of Psychology, San Diego State University, San Diego, CA, USA
| | - Gregory A Talavera
- Department of Psychology, San Diego State University, San Diego, CA, USA
| | - Marc D Gellman
- Department of Psychology, University of Miami, Miami, FL, USA
| | - Alberto R Ramos
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Hector M González
- Department of Neurosciences and Shiley-Marcos Alzheimer's Disease Research Center, University of California, San Diego, La Jolla, CA, USA
| | - Charles DeCarli
- Department of Neurology and Center for Neuroscience, University of California at Davis, Davis, CA, USA.
| |
Collapse
|
24
|
Park JC, Lim H, Byun MS, Yi D, Byeon G, Jung G, Kim YK, Lee DY, Han SH, Mook-Jung I. Sex differences in the progression of glucose metabolism dysfunction in Alzheimer's disease. Exp Mol Med 2023; 55:1023-1032. [PMID: 37121979 PMCID: PMC10238450 DOI: 10.1038/s12276-023-00993-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 02/21/2023] [Accepted: 02/23/2023] [Indexed: 05/02/2023] Open
Abstract
Alzheimer's disease (AD) is a common neurodegenerative disease characterized by amyloid plaques and impaired brain metabolism. Because women have a higher prevalence of AD than men, sex differences are of great interest. Using cross-sectional and longitudinal data, we showed sex-dependent metabolic dysregulations in the brains of AD patients. Cohort 1 (South Korean, n = 181) underwent Pittsburgh compound B-PET, fluorodeoxyglucose-PET, magnetic resonance imaging, and blood biomarker (plasma tau and beta-amyloid 42 and 40) measurements at baseline and two-year follow-ups. Transcriptome analysis of data from Cohorts 2 and 3 (European, n = 78; Singaporean, n = 18) revealed sex differences in AD-related alterations in brain metabolism. In women (but not in men), all imaging indicators displayed consistent correlation curves with AD progression. At the two-year follow-up, clear brain metabolic impairment was revealed only in women, and the plasma beta-amyloid 42/40 ratio was a possible biomarker for brain metabolism in women. Furthermore, our transcriptome analysis revealed sex differences in transcriptomes and metabolism in the brains of AD patients as well as a molecular network of 25 female-specific glucose metabolic genes (FGGs). We discovered four key-attractor FGG genes (ALDOA, ENO2, PRKACB, and PPP2R5D) that were associated with amyloid/tau-related genes (APP, MAPT, BACE1, and BACE2). Furthermore, these genes successfully distinguished amyloid positivity in women. Understanding sex differences in the pathogenesis of AD and considering these differences will improve development of effective diagnostics and therapeutic treatments for AD.
Collapse
Affiliation(s)
- Jong-Chan Park
- Department of Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Hanbyeol Lim
- Department of Medicine, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Min Soo Byun
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, 03080, Republic of Korea
- Department of Psychiatry, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea
| | - Dahyun Yi
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, 03080, Republic of Korea
| | - Gihwan Byeon
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, 03080, Republic of Korea
| | - Gijung Jung
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, 03080, Republic of Korea
| | - Yu Kyeong Kim
- Department of Nuclear Medicine, SMG-SNU Boramae Medical Center, Seoul, 07061, Republic of Korea
| | - Dong Young Lee
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, 03080, Republic of Korea
- Department of Psychiatry, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea
- Institute of Human Behavioral Medicine, Medical Research Center, Seoul National University, Seoul, 03080, Republic of Korea
| | - Sun-Ho Han
- Department of Biochemistry and Biomedical Sciences, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea.
- Korea Dementia Research Center, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea.
- Ilchun Genomic Medicine Institute (GMI), College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea.
| | - Inhee Mook-Jung
- Department of Biochemistry and Biomedical Sciences, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea.
- Korea Dementia Research Center, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea.
- Convergence Dementia Research Center, College of Medicine, Seoul National University, Seoul, 03080, Republic of Korea.
| |
Collapse
|
25
|
Liu FC, Cheng ML, Lo CJ, Hsu WC, Lin G, Lin HT. Exploring the aging process of cognitively healthy adults by analyzing cerebrospinal fluid metabolomics using liquid chromatography-tandem mass spectrometry. BMC Geriatr 2023; 23:217. [PMID: 37020298 PMCID: PMC10077689 DOI: 10.1186/s12877-023-03939-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 03/29/2023] [Indexed: 04/07/2023] Open
Abstract
BACKGROUND During biological aging, significant metabolic dysregulation in the central nervous system may lead to cognitive decline and neurodegeneration. However, the metabolomics of the aging process in cerebrospinal fluid (CSF) has not been thoroughly explored. METHODS In this cohort study of CSF metabolomics using liquid chromatography-mass spectrometry (LC-MS), fasting CSF samples collected from 92 cognitively unimpaired adults aged 20-87 years without obesity or diabetes were analyzed. RESULTS We identified 37 metabolites in these CSF samples with significant positive correlations with aging, including cysteine, pantothenic acid, 5-hydroxyindoleacetic acid (5-HIAA), aspartic acid, and glutamate; and two metabolites with negative correlations, asparagine and glycerophosphocholine. The combined alterations of asparagine, cysteine, glycerophosphocholine, pantothenic acid, sucrose, and 5-HIAA showed a superior correlation with aging (AUC = 0.982). These age-correlated changes in CSF metabolites might reflect blood-brain barrier breakdown, neuroinflammation, and mitochondrial dysfunction in the aging brain. We also found sex differences in CSF metabolites with higher levels of taurine and 5-HIAA in women using propensity-matched comparison. CONCLUSIONS Our LC-MS metabolomics of the aging process in a Taiwanese population revealed several significantly altered CSF metabolites during aging and between the sexes. These metabolic alterations in CSF might provide clues for healthy brain aging and deserve further exploration.
Collapse
Affiliation(s)
- Fu-Chao Liu
- Department of Anesthesiology, Chang Gung Memorial Hospital, 5 Fu-Shin Street, Kwei-Shan, Taoyuan, 333, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan
| | - Mei-Ling Cheng
- Metabolomics Core Laboratory, Healthy Aging Research Center, Chang Gung University, Taoyuan, 333, Taiwan
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan
- Clinical Metabolomics Core Laboratory, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chi-Jen Lo
- Metabolomics Core Laboratory, Healthy Aging Research Center, Chang Gung University, Taoyuan, 333, Taiwan
| | - Wen-Chuin Hsu
- College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan
- Department of Neurology, Chang Gung Memorial Hospital, Taoyuan, 333, Taiwan
| | - Gigin Lin
- Clinical Metabolomics Core Laboratory, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of Medical Imaging and Intervention, Imaging Core Lab, Chang Gung Memorial Hospital, Taoyuan, 333, Taiwan
| | - Huan-Tang Lin
- Department of Anesthesiology, Chang Gung Memorial Hospital, 5 Fu-Shin Street, Kwei-Shan, Taoyuan, 333, Taiwan.
- College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan.
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan.
| |
Collapse
|
26
|
Strefeler A, Jan M, Quadroni M, Teav T, Rosenberg N, Chatton JY, Guex N, Gallart-Ayala H, Ivanisevic J. Molecular insights into sex-specific metabolic alterations in Alzheimer's mouse brain using multi-omics approach. Alzheimers Res Ther 2023; 15:8. [PMID: 36624525 PMCID: PMC9827669 DOI: 10.1186/s13195-023-01162-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 01/02/2023] [Indexed: 01/11/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is a progressive neurodegenerative disorder that is characterized by altered cellular metabolism in the brain. Several of these alterations have been found to be exacerbated in females, known to be disproportionately affected by AD. We aimed to unravel metabolic alterations in AD at the metabolic pathway level and evaluate whether they are sex-specific through integrative metabolomic, lipidomic, and proteomic analysis of mouse brain tissue. METHODS We analyzed male and female triple-transgenic mouse whole brain tissue by untargeted mass spectrometry-based methods to obtain a molecular signature consisting of polar metabolite, complex lipid, and protein data. These data were analyzed using multi-omics factor analysis. Pathway-level alterations were identified through joint pathway enrichment analysis or by separately evaluating lipid ontology and known proteins related to lipid metabolism. RESULTS Our analysis revealed significant AD-associated and in part sex-specific alterations across the molecular signature. Sex-dependent alterations were identified in GABA synthesis, arginine biosynthesis, and in alanine, aspartate, and glutamate metabolism. AD-associated alterations involving lipids were also found in the fatty acid elongation pathway and lysophospholipid metabolism, with a significant sex-specific effect for the latter. CONCLUSIONS Through multi-omics analysis, we report AD-associated and sex-specific metabolic alterations in the AD brain involving lysophospholipid and amino acid metabolism. These findings contribute to the characterization of the AD phenotype at the molecular level while considering the effect of sex, an overlooked yet determinant metabolic variable.
Collapse
Affiliation(s)
- Abigail Strefeler
- grid.9851.50000 0001 2165 4204Metabolomics Unit, Faculty of Biology and Medicine, University de Lausanne, Lausanne, Switzerland
| | - Maxime Jan
- grid.9851.50000 0001 2165 4204Bioinformatics Competence Center, Faculty of Biology and Medicine, University de Lausanne, Lausanne, Switzerland
| | - Manfredo Quadroni
- grid.9851.50000 0001 2165 4204Protein Analysis Facility, Faculty of Biology and Medicine, University de Lausanne, Lausanne, Switzerland
| | - Tony Teav
- grid.9851.50000 0001 2165 4204Metabolomics Unit, Faculty of Biology and Medicine, University de Lausanne, Lausanne, Switzerland
| | - Nadia Rosenberg
- grid.9851.50000 0001 2165 4204Department of Fundamental Neurosciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Jean-Yves Chatton
- grid.9851.50000 0001 2165 4204Department of Fundamental Neurosciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Nicolas Guex
- grid.9851.50000 0001 2165 4204Bioinformatics Competence Center, Faculty of Biology and Medicine, University de Lausanne, Lausanne, Switzerland
| | - Hector Gallart-Ayala
- grid.9851.50000 0001 2165 4204Metabolomics Unit, Faculty of Biology and Medicine, University de Lausanne, Lausanne, Switzerland
| | - Julijana Ivanisevic
- grid.9851.50000 0001 2165 4204Metabolomics Unit, Faculty of Biology and Medicine, University de Lausanne, Lausanne, Switzerland
| |
Collapse
|
27
|
Pontrello CG, McWhirt JM, Glabe CG, Brewer GJ. Age-Related Oxidative Redox and Metabolic Changes Precede Intraneuronal Amyloid-β Accumulation and Plaque Deposition in a Transgenic Alzheimer's Disease Mouse Model. J Alzheimers Dis 2022; 90:1501-1521. [PMID: 36278355 PMCID: PMC9789488 DOI: 10.3233/jad-220824] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
BACKGROUND Many identified mechanisms could be upstream of the prominent amyloid-β (Aβ) plaques in Alzheimer's disease (AD). OBJECTIVE To profile the progression of pathology in AD. METHODS We monitored metabolic signaling, redox stress, intraneuronal amyloid-β (iAβ) accumulation, and extracellular plaque deposition in the brains of 3xTg-AD mice across the lifespan. RESULTS Intracellular accumulation of aggregated Aβ in the CA1 pyramidal cells at 9 months preceded extracellular plaques that first presented in the CA1 at 16 months of age. In biochemical assays, brain glutathione (GSH) declined with age in both 3xTg-AD and non-transgenic controls, but the decline was accelerated in 3xTg-AD brains from 2 to 4 months. The decline in GSH correlated exponentially with the rise in iAβ. Integrated metabolic signaling as the ratio of phospho-Akt (pAkt) to total Akt (tAkt) in the PI3kinase and mTOR pathway declined at 6, 9, and 12 months, before rising at 16 and 20 months. These pAkt/tAkt ratios correlated with both iAβ and GSH levels in a U-shaped relationship. Selective vulnerability of age-related AD-genotype-specific pAkt changes was greatest in the CA1 pyramidal cell layer. To demonstrate redox causation, iAβ accumulation was lowered in cultured middle-age adult 3xTg-AD neurons by treatment of the oxidized redox state in the neurons with exogenous cysteine. CONCLUSION The order of pathologic progression in the 3xTg-AD mouse was loss of GSH (oxidative redox shift) followed by a pAkt/tAkt metabolic shift in CA1, iAβ accumulation in CA1, and extracellular Aβ deposition. Upstream targets may prove strategically more effective for therapy before irreversible changes.
Collapse
Affiliation(s)
- Crystal G. Pontrello
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA, USA,
Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| | - Joshua M. McWhirt
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| | - Charles G. Glabe
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA, USA,
Center for Neurobiology of Learning and Memory, University of California Irvine, Irvine, CA, USA,
MIND Institute, University of California Irvine, Irvine, CA, USA
| | - Gregory J. Brewer
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA,
Center for Neurobiology of Learning and Memory, University of California Irvine, Irvine, CA, USA,
MIND Institute, University of California Irvine, Irvine, CA, USA,Correspondence to: Gregory J. Brewer, Department of Biomedical Engineering, University of California Irvine, Irvine, CA 92697, USA. Tel.: +1 217 502 4511; E-mail:
| |
Collapse
|
28
|
Mesquita PHC, Osburn SC, Godwin JS, Roberts MD, Kavazis AN. Effects of aging and long-term physical activity on mitochondrial physiology and redox state of the cortex and cerebellum of female rats. Physiol Rep 2022; 10:e15542. [PMID: 36543327 PMCID: PMC9771693 DOI: 10.14814/phy2.15542] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 11/09/2022] [Accepted: 11/21/2022] [Indexed: 06/17/2023] Open
Abstract
We investigated the effects of aging and long-term physical activity on markers of mitochondrial function and dynamics in the cortex and cerebellum of female rats. Additionally, we interrogated markers of oxidative damage and antioxidants. Thirty-four female Lewis rats were separated into three groups. A young group (YNG, n = 10) was euthanized at 6 months of age. Two other groups were aged to 15 months and included a physical activity group (MA-PA, n = 12) and a sedentary group (MA-SED, n = 12). There were no age effects for any of the variables investigated, except for SOD2 protein levels in the cortex (+6.5%, p = 0.012). Long-term physical activity increased mitochondrial complex IV activity in the cortex compared to YNG (+85%, p = 0.016) and MA-SED (+82%, p = 0.023) and decreased carbonyl levels in the cortex compared to YNG (-12.49%, p = 0.034). Our results suggest that the mitochondrial network and redox state of the brain of females may be more resilient to the aging process than initially thought. Further, voluntary wheel running had minimal beneficial effects on brain markers of oxidative damage and mitochondrial physiology.
Collapse
Affiliation(s)
| | | | | | - Michael D. Roberts
- School of KinesiologyAuburn UniversityAuburnAlabamaUSA
- Edward Via College of Osteopathic MedicineAuburnAlabamaUSA
| | | |
Collapse
|
29
|
Chitu V, Gökhan Ş, Stanley ER. Modeling CSF-1 receptor deficiency diseases - how close are we? FEBS J 2022; 289:5049-5073. [PMID: 34145972 PMCID: PMC8684558 DOI: 10.1111/febs.16085] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/17/2021] [Accepted: 06/18/2021] [Indexed: 12/11/2022]
Abstract
The role of colony-stimulating factor-1 receptor (CSF-1R) in macrophage and organismal development has been extensively studied in mouse. Within the last decade, mutations in the CSF1R have been shown to cause rare diseases of both pediatric (Brain Abnormalities, Neurodegeneration, and Dysosteosclerosis, OMIM #618476) and adult (CSF1R-related leukoencephalopathy, OMIM #221820) onset. Here we review the genetics, penetrance, and histopathological features of these diseases and discuss to what extent the animal models of Csf1r deficiency currently available provide systems in which to study the underlying mechanisms involved.
Collapse
Affiliation(s)
- Violeta Chitu
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, N.Y. 10461, USA
| | - Şölen Gökhan
- Institute for Brain Disorders and Neural Regeneration, Department of Neurology, Albert Einstein College of Medicine, Bronx, N.Y. 10461, USA
| | - E. Richard Stanley
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, N.Y. 10461, USA
| |
Collapse
|
30
|
Winek K, Tzur Y, Soreq H. Biological underpinnings of sex differences in neurological disorders. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2022; 164:27-67. [PMID: 36038206 DOI: 10.1016/bs.irn.2022.06.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The importance of sex differences in neurological disorders has been increasingly acknowledged in recent clinical and basic research studies, but the complex biology and genetics underlying sex-linked biological heterogeneity and its brain-to-body impact remained incompletely understood. Men and women differ substantially in their susceptibility to certain neurological diseases, in the severity of symptoms, prognosis as well as the nature and efficacy of their response to treatments. The detailed mechanisms underlying these differences, especially at the molecular level, are being addressed in many studies but leave a lot to be further revealed. Here, we provide an overview of recent advances in our understanding of how sex differences in the brain and brain-body signaling contribute to neurological disorders and further present some future prospects entailed in terms of diagnostics and therapeutics.
Collapse
Affiliation(s)
- Katarzyna Winek
- The Edmond & Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel; The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yonat Tzur
- The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Hermona Soreq
- The Edmond & Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel; The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
31
|
Guimarães P, Serranho P, Martins J, Moreira PI, Ambrósio AF, Castelo-Branco M, Bernardes R. Retinal Aging in 3× Tg-AD Mice Model of Alzheimer's Disease. Front Aging Neurosci 2022; 14:832195. [PMID: 35783138 PMCID: PMC9244797 DOI: 10.3389/fnagi.2022.832195] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 05/16/2022] [Indexed: 11/13/2022] Open
Abstract
The retina, as part of the central nervous system (CNS), can be the perfect target for in vivo, in situ, and noninvasive neuropathology diagnosis and assessment of therapeutic efficacy. It has long been established that several age-related brain changes are more pronounced in Alzheimer's disease (AD). Nevertheless, in the retina such link is still under-explored. This study investigates the differences in the aging of the CNS through the retina of 3× Tg-AD and wild-type mice. A dedicated optical coherence tomograph imaged mice's retinas for 16 months. Two neural networks were developed to model independently each group's ages and were then applied to an independent set containing images from both groups. Our analysis shows a mean absolute error of 0.875±1.1 × 10−2 and 1.112±1.4 × 10−2 months, depending on training group. Our deep learning approach appears to be a reliable retinal OCT aging marker. We show that retina aging is distinct in the two classes: the presence of the three mutated human genes in the mouse genome has an impact on the aging of the retina. For mice over 4 months-old, transgenic mice consistently present a negative retina age-gap when compared to wild-type mice, regardless of training set. This appears to contradict AD observations in the brain. However, the ‘black-box” nature of deep-learning implies that one cannot infer reasoning. We can only speculate that some healthy age-dependent neural adaptations may be altered in transgenic animals.
Collapse
Affiliation(s)
- Pedro Guimarães
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Institute for Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, Coimbra, Portugal
- *Correspondence: Pedro Guimarães
| | - Pedro Serranho
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Institute for Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, Coimbra, Portugal
- Department of Sciences and Technology, Universidade Aberta, Lisbon, Portugal
| | - João Martins
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Institute for Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, Coimbra, Portugal
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine (FMUC), University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Faculty of Medicine (FMUC), University of Coimbra, Coimbra, Portugal
| | - Paula I. Moreira
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Faculty of Medicine (FMUC), University of Coimbra, Coimbra, Portugal
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - António Francisco Ambrósio
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine (FMUC), University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Faculty of Medicine (FMUC), University of Coimbra, Coimbra, Portugal
| | - Miguel Castelo-Branco
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Institute for Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Faculty of Medicine (FMUC), University of Coimbra, Coimbra, Portugal
| | - Rui Bernardes
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Institute for Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Faculty of Medicine (FMUC), University of Coimbra, Coimbra, Portugal
| |
Collapse
|
32
|
Klonarakis M, De Vos M, Woo E, Ralph L, Thacker JS, Gil-Mohapel J. The three sisters of fate: Genetics, pathophysiology and outcomes of animal models of neurodegenerative diseases. Neurosci Biobehav Rev 2022; 135:104541. [DOI: 10.1016/j.neubiorev.2022.104541] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 11/28/2021] [Accepted: 01/13/2022] [Indexed: 02/07/2023]
|
33
|
Guo L, Zhong MB, Zhang L, Zhang B, Cai D. Sex Differences in Alzheimer's Disease: Insights From the Multiomics Landscape. Biol Psychiatry 2022; 91:61-71. [PMID: 33896621 PMCID: PMC8996342 DOI: 10.1016/j.biopsych.2021.02.968] [Citation(s) in RCA: 104] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 02/22/2021] [Accepted: 02/22/2021] [Indexed: 01/03/2023]
Abstract
Alzheimer's disease (AD) has complex etiologies, and the impact of sex on AD varies over the course of disease development. The literature provides some evidence of sex-specific contributions to AD. However, molecular mechanisms of sex-biased differences in AD remain elusive. Multiomics data in tandem with systems biology approaches offer a new avenue to dissect sex-stratified molecular mechanisms of AD and to develop sex-specific diagnostic and therapeutic strategies for AD. Single-cell transcriptomic datasets and cell deconvolution of bulk tissue transcriptomic data provide additional insights into brain cell type-specific impact on sex-biased differences in AD. In this review, we summarize the impact of sex chromosomes and sex hormones on AD, the impact of sex-biased differences during AD development, and the interplay between sex and a major AD genetic risk factor, the APOE ε4 genotype, through the multiomics landscape. Several sex-biased molecular pathways such as neuroinflammation and bioenergetic metabolism have been identified. The importance of sex chromosome and sex hormones, as well as the associated pathways in AD pathogenesis, is further strengthened by findings from omics studies. Future research efforts should integrate the multiomics data from different brain regions and different cell types using systems biology approaches, and leverage the knowledge into a holistic examination of sex differences in AD. Advances in systems biology technologies and increasingly available large-scale multiomics datasets will facilitate future studies dissecting such complex signaling mechanisms to better understand AD pathogenesis in both sexes, with the ultimate goals of developing efficacious sex- and APOE-stratified preventive and therapeutic interventions for AD.
Collapse
Affiliation(s)
- Lei Guo
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York; Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Margaret B Zhong
- Department of Neuroscience, Barnard College of Columbia University, New York, New York
| | - Larry Zhang
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York; Research and Development Service, James J. Peters VA Medical Center, Bronx, New York
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York; Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, New York.
| | - Dongming Cai
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York; Alzheimer Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, New York; Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, New York; Research and Development Service, James J. Peters VA Medical Center, Bronx, New York.
| |
Collapse
|
34
|
Lin HT, Cheng ML, Lo CJ, Hsu WC, Lin G, Liu FC. 1H NMR metabolomic profiling of human cerebrospinal fluid in aging process. Am J Transl Res 2021; 13:12495-12508. [PMID: 34956468 PMCID: PMC8661231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 09/21/2021] [Indexed: 06/14/2023]
Abstract
The molecular process of biological aging might be accompanied by significant metabolic derangement, especially in the central nervous system (CNS), since the brain has an enormous energy demand. However, the metabolic signature of the aging process in cerebrospinal fluid (CSF) has not been thoroughly investigated, especially in the Asian population. In this prospective cohort study on CSF metabolomics using proton nuclear magnetic resonance (NMR) spectroscopy, fasting CSF samples from 75 cognitively unimpaired patients aged 20-92 years without diabetes or obesity, undergoing spinal anesthesia for elective surgery were analyzed. Several metabolites in CSF samples were identified as having a significant association with the aging process in cerebral circulation; among the metabolites, the levels of alanine, citrate, creatinine, lactate, leucine, tyrosine, and valine significantly increased in old patients compared to those in young patients. The combined CSF metabolite alterations in citrate, lactate, leucine, tyrosine, and valine had a superior correlation with the aging process in all age groups. In conclusion, our pilot study of aging CSF metabolomics in the Taiwanese population presents significantly altered CSF metabolites with potential relevance to the aging process. These metabolic alterations in CSF samples might imply increasing anaerobic glycolysis, mitochondrial dysfunction, and decreasing glucose utilization in cerebral circulation in aged patients.
Collapse
Affiliation(s)
- Huan-Tang Lin
- Department of Anesthesiology, Chang Gung Memorial HospitalTaoyuan 333, Taiwan
- College of Medicine, Chang Gung UniversityTaoyuan 333, Taiwan
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung UniversityTaoyuan 333, Taiwan
| | - Mei-Ling Cheng
- Metabolomics Core Laboratory, Healthy Aging Research Center, Chang Gung UniversityTaoyuan 333, Taiwan
- Department of Biomedical Sciences, College of Medicine, Chang Gung UniversityTaoyuan 333, Taiwan
- Clinical Metabolomics Core Laboratory, Chang Gung Memorial HospitalTaoyuan, Taiwan
| | - Chi-Jen Lo
- Metabolomics Core Laboratory, Healthy Aging Research Center, Chang Gung UniversityTaoyuan 333, Taiwan
| | - Wen-Chuin Hsu
- College of Medicine, Chang Gung UniversityTaoyuan 333, Taiwan
- Department of Neurology, Chang Gung Memorial HospitalTaoyuan 333, Taiwan
| | - Gigin Lin
- Department of Medical Imaging and Intervention, Institute for Radiological Research, Chang Gung Memorial HospitalTaoyuan 333, Taiwan
- Clinical Phenome Center, Chang Gung Memorial HospitalTaoyuan 333, Taiwan
| | - Fu-Chao Liu
- Department of Anesthesiology, Chang Gung Memorial HospitalTaoyuan 333, Taiwan
- College of Medicine, Chang Gung UniversityTaoyuan 333, Taiwan
| |
Collapse
|
35
|
Beckman D, Morrison JH. Towards developing a rhesus monkey model of early Alzheimer's disease focusing on women's health. Am J Primatol 2021; 83:e23289. [PMID: 34056733 DOI: 10.1002/ajp.23289] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 05/11/2021] [Accepted: 05/16/2021] [Indexed: 12/22/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of elderly dementia, affecting nearly 50 million people worldwide, with two-thirds of the cases in the USA in women. Despite considerable investment, this prevalence is expected to increase further in the coming decades, based on the projected demographics of the population. Currently, most of the preclinical AD studies rely on transgenic mice carrying mutations associated with the early onset familiar form of AD, although the vast majority of cases are sporadic. A prevailing current hypothesis is that the cascade of events leading to AD starts with the accumulation of small soluble oligomers of the Aβ peptide (AβOs) that target and disrupt synapses. Taking advantage of the high translational power of rhesus monkeys due to their physiological and genetic similarities to humans, we recently developed a female rhesus monkey model of early AD pathogenesis based on exogenous administration AβOs. Here we review and discuss how soluble oligomers of Aβ can target vulnerable spines in the neocortex and hippocampus of female middle-aged monkeys and induce neuroinflammatory responses, similar to what is known to occur in the human brain. Developing a rhesus monkey model of early AD focusing on women's health is critical for the understanding of how hormonal changes during menopause transition affect brain health and ultimately may contribute to AD neurodegeneration.
Collapse
Affiliation(s)
- Danielle Beckman
- California National Primate Research Center, UC Davis, Davis, California, USA
| | - John H Morrison
- California National Primate Research Center, UC Davis, Davis, California, USA
- Department of Neurology, School of Medicine, UC Davis, Davis, California, USA
| |
Collapse
|
36
|
Vitali F, Branigan GL, Brinton RD. Preventing Alzheimer's disease within reach by 2025: Targeted-risk-AD-prevention (TRAP) strategy. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2021; 7:e12190. [PMID: 34584937 PMCID: PMC8451031 DOI: 10.1002/trc2.12190] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 04/20/2021] [Indexed: 01/15/2023]
Abstract
INTRODUCTION Alzheimer's disease (AD) is a progressive neurodegenerative disease that currently affects 6.2 million people in the United States and is projected to impact 12.7 million worldwide in 2050 with no effective disease-modifying therapeutic or cure. In 2011 as part of the National Alzheimer's Project Act, the National Plan to Address Alzheimer's Disease was signed into law which proposed to effectively prevent AD by 2025, which is rapidly approaching. The preclinical phase of AD can begin 20 years prior to diagnosis, which provides an extended window for preventive measures that would exert a transformative impact on incidence and prevalence of AD. METHODS A novel combination of text-mining and natural language processing strategies to identify (1) AD risk factors, (2) therapeutics that can target risk factor pathways, and (3) studies supporting therapeutics in the PubMed database was conducted. To classify the literature relevant to AD preventive strategies, a relevance score (RS) based on STRING (search tool for the retrieval of interacting genes/proteins) score for protein-protein interactions and a confidence score (CS) on Bayesian inference were developed. To address mechanism of action, network analysis of protein targets for effective drugs was conducted. Collectively, the analytic approach, referred to as a targeted-risk-AD-prevention (TRAP) strategy, led to a ranked list of candidate therapeutics to reduce AD risk. RESULTS Based on TRAP mining of 9625 publications, 364 AD risk factors were identified. Based on risk factor indications, 629 Food and Drug Administration-approved drugs were identified. Computation of ranking scores enabled identification of 46 relevant high confidence (RS & CS > 0.7) drugs associated with reduced AD risk. Within these candidate therapeutics, 16 had more than one clinical study supporting AD risk reduction. Top-ranked therapeutics with high confidence emerged within lipid-lowering, anti-inflammatory, hormone, and metabolic-related drug classes. DISCUSSION Outcomes of our novel bioinformatic strategy support therapeutic targeting of biological mechanisms and pathways underlying relevant AD risk factors with high confidence. Early interventions that target pathways associated with increased risk of AD have the potential to support the goal of effectively preventing AD by 2025.
Collapse
Affiliation(s)
- Francesca Vitali
- Center for Innovation in Brain ScienceUniversity of ArizonaTucsonArizonaUSA
- Department of Neurology, College of MedicineUniversity of ArizonaTucsonArizonaUSA
- Center for Biomedical Informatics and Biostatistics, College of MedicineUniversity of ArizonaTucsonArizonaUSA
| | - Gregory L. Branigan
- Center for Innovation in Brain ScienceUniversity of ArizonaTucsonArizonaUSA
- Department of Pharmacology, College of MedicineUniversity of ArizonaTucsonArizonaUSA
- MD‐PhD training program, College of MedicineUniversity of ArizonaTucsonArizonaUSA
| | - Roberta Diaz Brinton
- Center for Innovation in Brain ScienceUniversity of ArizonaTucsonArizonaUSA
- Department of Neurology, College of MedicineUniversity of ArizonaTucsonArizonaUSA
- Department of Pharmacology, College of MedicineUniversity of ArizonaTucsonArizonaUSA
| |
Collapse
|
37
|
Bagit A, Hayward GC, MacPherson REK. Exercise and estrogen: common pathways in Alzheimer's disease pathology. Am J Physiol Endocrinol Metab 2021; 321:E164-E168. [PMID: 34056921 PMCID: PMC8321825 DOI: 10.1152/ajpendo.00008.2021] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease that is characterized by progressive declines in cognitive function. Current epidemiological data indicate significant sex-linked disparities, where females have a higher risk of developing AD compared with male counterparts. This disparity necessitates further investigations to uncover the pathological and molecular factors influencing these sex differences. Although the underlying pathways behind this observed disparity remain elusive, recent research points to menopausal estrogen loss as a potential factor. Estrogen holds a significant role in amyloid precursor protein (APP) processing and overall neuronal health through the regulation of brain-derived neurotrophic factor (BDNF), a factor that is also reduced in postmenopausal women. BDNF is a known contributor to neuronal health and its reduced expression is typically linked to AD disorders. Exercise is known to increase BDNF and may provide an accessible activity for postmenopausal women to reduce their risk of AD. This review aims to discuss the relationship between estrogen, exercise, and BDNF in AD pathology.
Collapse
Affiliation(s)
- Ahmed Bagit
- Department of Health Sciences, Brock University, St. Catharines, Ontario, Canada
| | - Grant C Hayward
- Faculty of Medicine, Ottawa University, Ottawa, Ontario, Canada
| | - Rebecca E K MacPherson
- Department of Health Sciences, Brock University, St. Catharines, Ontario, Canada
- Centre for Neuroscience, Brock University, St. Catharines, Ontario, Canada
| |
Collapse
|
38
|
Yang X, Zhi J, Leng H, Chen Y, Gao H, Ma J, Ji J, Hu Q. The piperine derivative HJ105 inhibits Aβ 1-42-induced neuroinflammation and oxidative damage via the Keap1-Nrf2-TXNIP axis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 87:153571. [PMID: 33994056 DOI: 10.1016/j.phymed.2021.153571] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 04/10/2021] [Accepted: 04/12/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND Piperine is a great lead compound, as a phytopharmaceutical with reported neuroprotective effects in neurodegenerative diseases. HJ105, a piperine derivative with high affinity to Keap1 receptor, attracts increasing attention in Alzheimer's disease (AD) treatment. PURPOSE This work mainly aimed to study HJ105's therapeutic effects on Aβ1-42-associated AD and the underpinning mechanisms. METHODS In the in vivo part, a rat model of AD was established by bilateral intra-hippocampal administration of aggregated Aβ1-42, followed by a month of intragastric HJ105 or donepezil administration. Spatial and learning memories were detected by the Morris water maze assay, passive avoidance learning as well as Y-maze test. The morphology of hippocampal neurons was assessed by hematoxylin-eosin (H&E) staining. In addition, the amounts of the IL-1β and TNF-α were obtained with specific ELISA kits. More importantly, apoptosis-related proteins and factors involved in Nrf2/TXNIP/NLPR3 pathways were detected by Western blot, while the interaction between Keap1 and Nrf2 was assessed by co-immunoprecipitation. In the in vitro part, human neuroblastoma (SH-SY5Y) cells were applied to evaluate the role of HJ105 on Aβ1-42-induced neuronal damage. RESULTS Treatment of HJ105 not only reversed memory impairment, but also protected neurons in the hippocampus by inhibiting Bax/Bcl2 ratio increase. HJ105 decreased TXNIP expression, suppressing NLRP3 inflammasome activation in the hippocampus, which in turn counteracted the upregulation of IL-1β and TNF-α. Notably, HJ105 exerted an inhibitory effect on Keap1-Nrf2 interaction and upregulated nuclear Nrf2, which conversely increased the expression levels of superoxide dismutase, catalase and glutathione peroxidase and downregulated malondialdehyde. Additionally, neurotoxicity induced by Aβ1-42 in SH-SY5Y cells was alleviated by HJ105. CONCLUSION Overall, HJ105 exerts neuroprotective effects in SH-SY5Y cells induced by Aβ1-42 as well as in experimental rats with AD by decreasing apoptosis, oxidative stress and neuroinflammation, partly via suppression of Keap1-Nrf2 complex generation. HJ105 might represent a promising compound for AD treatment.
Collapse
Affiliation(s)
- Xiping Yang
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China
| | - Jingke Zhi
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China
| | - Haifeng Leng
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China
| | - Yu Chen
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China
| | - Haoran Gao
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China; School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, PR China
| | - Jinming Ma
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, Jiangsu Ocean University, Lianyungang 222005, PR China
| | - Jing Ji
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, Jiangsu Ocean University, Lianyungang 222005, PR China.
| | - Qinghua Hu
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China.
| |
Collapse
|
39
|
Foret JT, Dekhtyar M, Cole JH, Gourley DD, Caillaud M, Tanaka H, Haley AP. Network Modeling Sex Differences in Brain Integrity and Metabolic Health. Front Aging Neurosci 2021; 13:691691. [PMID: 34267647 PMCID: PMC8275835 DOI: 10.3389/fnagi.2021.691691] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 05/27/2021] [Indexed: 01/14/2023] Open
Abstract
Hypothesis-driven studies have demonstrated that sex moderates many of the relationships between brain health and cardiometabolic disease, which impacts risk for later-life cognitive decline. In the present study, we sought to further our understanding of the associations between multiple markers of brain integrity and cardiovascular risk in a midlife sample of 266 individuals by using network analysis, a technique specifically designed to examine complex associations among multiple systems at once. Separate network models were constructed for male and female participants to investigate sex differences in the biomarkers of interest, selected based on evidence linking them with risk for late-life cognitive decline: all components of metabolic syndrome (obesity, hypertension, dyslipidemia, and hyperglycemia); neuroimaging-derived brain-predicted age minus chronological age; ratio of white matter hyperintensities to whole brain volume; seed-based resting state functional connectivity in the Default Mode Network, and ratios of N-acetyl aspartate, glutamate and myo-inositol to creatine, measured through proton magnetic resonance spectroscopy. Males had a sparse network (87.2% edges = 0) relative to females (69.2% edges = 0), indicating fewer relationships between measures of cardiometabolic risk and brain integrity. The edges in the female network provide meaningful information about potential mechanisms between brain integrity and cardiometabolic health. Additionally, Apolipoprotein ϵ4 (ApoE ϵ4) status and waist circumference emerged as central nodes in the female model. Our study demonstrates that network analysis is a promising technique for examining relationships between risk factors for cognitive decline in a midlife population and that investigating sex differences may help optimize risk prediction and tailor individualized treatments in the future.
Collapse
Affiliation(s)
- Janelle T. Foret
- Department of Psychology, The University of Texas at Austin, Austin, TX, United States
| | - Maria Dekhtyar
- Department of Psychology, The University of Texas at Austin, Austin, TX, United States
| | - James H. Cole
- Department of Computer Science, Centre for Medical Image Computing, University College London, London, United Kingdom
- Dementia Research Centre, Institute of Neurology, University College London, London, United Kingdom
| | - Drew D. Gourley
- Department of Kinesiology and Health Education, The University of Texas at Austin, Austin, TX, United States
| | - Marie Caillaud
- Department of Psychology, The University of Texas at Austin, Austin, TX, United States
| | - Hirofumi Tanaka
- Department of Kinesiology and Health Education, The University of Texas at Austin, Austin, TX, United States
| | - Andreana P. Haley
- Department of Psychology, The University of Texas at Austin, Austin, TX, United States
- Biomedical Imaging Center, The University of Texas at Austin, Austin, TX, United States
| |
Collapse
|
40
|
Lee JH, Cho SH, Jang EH, Kim SA. Sex-specific Changes in Brain Estrogen Metabolism Induced by Acute Trimethyltin Exposure. In Vivo 2021; 35:793-797. [PMID: 33622871 DOI: 10.21873/invivo.12319] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/10/2021] [Accepted: 01/11/2021] [Indexed: 12/23/2022]
Abstract
BACKGROUND/AIM In this study, we investigated sex-specific effects of acute exposure to trimethyltin, a known neurotoxicant on metabolic steroids. MATERIALS AND METHODS We administered intraperitoneally 2.3 mg/kg trimethyltin to 4-week-old male mice and measured the levels of metabolic steroids 24 h after treatment. We also measured mRNA and protein levels of cytochrome P450 1B1 using real-time polymerase chain reaction and western blotting. RESULTS Cortisol levels in the cortex increased in both sexes following acute trimethyltin exposure. The estradiol levels decreased, and the 4-hydroxyestradiol levels increased only in females. We also observed increased cytochrome P450 1B1 mRNA and protein levels only in the female cortex. CONCLUSION Acute trimethyltin exposure induces distinct sex-specific metabolic changes in the brain before significant sexual maturation.
Collapse
Affiliation(s)
- Jung Ho Lee
- Department of Pharmacology, School of Medicine, Eulji University, Daejeon, Republic of Korea
| | - Sung-Hee Cho
- Chemical Analysis Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon, Republic of Korea
| | - Eun Hye Jang
- Department of Pharmacology, School of Medicine, Eulji University, Daejeon, Republic of Korea
| | - Soon Ae Kim
- Department of Pharmacology, School of Medicine, Eulji University, Daejeon, Republic of Korea;
| |
Collapse
|
41
|
A Negative Energy Balance Is Associated with Metabolic Dysfunctions in the Hypothalamus of a Humanized Preclinical Model of Alzheimer's Disease, the 5XFAD Mouse. Int J Mol Sci 2021; 22:ijms22105365. [PMID: 34065168 PMCID: PMC8161294 DOI: 10.3390/ijms22105365] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/15/2021] [Accepted: 05/16/2021] [Indexed: 12/19/2022] Open
Abstract
Increasing evidence links metabolic disorders with neurodegenerative processes including Alzheimer’s disease (AD). Late AD is associated with amyloid (Aβ) plaque accumulation, neuroinflammation, and central insulin resistance. Here, a humanized AD model, the 5xFAD mouse model, was used to further explore food intake, energy expenditure, neuroinflammation, and neuroendocrine signaling in the hypothalamus. Experiments were performed on 6-month-old male and female full transgenic (Tg5xFAD/5xFAD), heterozygous (Tg5xFAD/-), and non-transgenic (Non-Tg) littermates. Although histological analysis showed absence of Aβ plaques in the hypothalamus of 5xFAD mice, this brain region displayed increased protein levels of GFAP and IBA1 in both Tg5xFAD/- and Tg5xFAD/5xFAD mice and increased expression of IL-1β in Tg5xFAD/5xFAD mice, suggesting neuroinflammation. This condition was accompanied by decreased body weight, food intake, and energy expenditure in both Tg5xFAD/- and Tg5xFAD/5xFAD mice. Negative energy balance was associated with altered circulating levels of insulin, GLP-1, GIP, ghrelin, and resistin; decreased insulin and leptin hypothalamic signaling; dysregulation in main metabolic sensors (phosphorylated IRS1, STAT5, AMPK, mTOR, ERK2); and neuropeptides controlling energy balance (NPY, AgRP, orexin, MCH). These results suggest that glial activation and metabolic dysfunctions in the hypothalamus of a mouse model of AD likely result in negative energy balance, which may contribute to AD pathogenesis development.
Collapse
|
42
|
Microglial heterogeneity in aging and Alzheimer's disease: Is sex relevant? J Pharmacol Sci 2021; 146:169-181. [PMID: 34030799 DOI: 10.1016/j.jphs.2021.03.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 02/24/2021] [Accepted: 03/22/2021] [Indexed: 02/08/2023] Open
Abstract
Neurodegenerative diseases and their associated cognitive decline are known to be more prevalent during aging. Recent evidence has uncovered the role of microglia, the immunocompetent cells of the brain, in dysfunctions linked to neurodegenerative diseases such as is Alzheimer's disease (AD). Similar to other pathologies, AD is shown to be sex-biased, with females being more at risk compared to males. While the mechanisms driving this prevalence are still unclear, emerging data suggest the sex differences present in microglia throughout life might lead to different responses of these cells in both health and disease. Furthermore, microglial cells have recently been recognized as a deeply heterogeneous population, with multiple subsets and/or phenotypes stemming from diverse parameters such as age, sex or state of health. Therefore, this review discusses microglial heterogeneity during aging in both basal conditions and AD with a focus on existing sex differences in this process.
Collapse
|
43
|
Bryan J, Mandan A, Kamat G, Gottschalk WK, Badea A, Adams KJ, Thompson JW, Colton CA, Mukherjee S, Lutz MW. Likelihood ratio statistics for gene set enrichment in Alzheimer's disease pathways. Alzheimers Dement 2021; 17:561-573. [PMID: 33480182 PMCID: PMC8044005 DOI: 10.1002/alz.12223] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION The study of Alzheimer's disease (AD) has revealed biological pathways with implications for disease neuropathology and pathophysiology. These pathway-level effects may also be mediated by individual characteristics or covariates such as age or sex. Evaluation of AD biological pathways in the context of interactions with these covariates is critical to the understanding of AD as well as the development of model systems used to study the disease. METHODS Gene set enrichment methods are powerful tools used to interpret gene-level statistics at the level of biological pathways. We introduce a method for quantifying gene set enrichment using likelihood ratio-derived test statistics (gsLRT), which accounts for sample covariates like age and sex. We then use our method to test for age and sex interactions with protein expression levels in AD and to compare the pathway results between human and mouse species. RESULTS Our method, based on nested logistic regressions is competitive with the existing standard for gene set testing in the context of linear models and complex experimental design. The gene sets we identify as having a significant association with AD-both with and without additional covariate interactions-are validated by previous studies. Differences between gsLRT results on mouse and human datasets are observed. DISCUSSION Characterizing biological pathways involved in AD builds on the important work involving single gene drivers. Our gene set enrichment method finds pathways that are significantly related to AD while accounting for covariates that may be relevant to disease development. The method highlights commonalities and differences between human AD and mouse models, which may inform the development of higher fidelity models for the study of AD.
Collapse
Affiliation(s)
- Jordan Bryan
- Department of Statistical Science, Duke University, Durham, NC 27708, USA
| | - Arpita Mandan
- Department of Statistical Science, Duke University, Durham, NC 27708, USA
| | - Gauri Kamat
- Department of Statistical Science, Duke University, Durham, NC 27708, USA
| | | | - Alexandra Badea
- Department of Neurology, Duke University, Durham, NC 27708, USA
| | - Kendra J. Adams
- Department of Neurology, Duke University, Durham, NC 27708, USA
| | | | - Carol A. Colton
- Department of Neurology, Duke University, Durham, NC 27708, USA
| | - Sayan Mukherjee
- Department of Statistical Science, Duke University, Durham, NC 27708, USA
- Departments of Mathematics, Computer Science, and Biostatistics & Bioinformatics, Duke University, Durham, NC 27708, USA
| | - Michael W. Lutz
- Department of Neurology, Duke University, Durham, NC 27708, USA
| |
Collapse
|
44
|
Weber CM, Clyne AM. Sex differences in the blood-brain barrier and neurodegenerative diseases. APL Bioeng 2021; 5:011509. [PMID: 33758788 PMCID: PMC7968933 DOI: 10.1063/5.0035610] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 02/03/2021] [Indexed: 02/06/2023] Open
Abstract
The number of people diagnosed with neurodegenerative diseases is on the rise. Many of these diseases, including Alzheimer's disease, Parkinson's disease, multiple sclerosis, and motor neuron disease, demonstrate clear sexual dimorphisms. While sex as a biological variable must now be included in animal studies, sex is rarely included in in vitro models of human neurodegenerative disease. In this Review, we describe these sex-related differences in neurodegenerative diseases and the blood-brain barrier (BBB), whose dysfunction is linked to neurodegenerative disease development and progression. We explain potential mechanisms by which sex and sex hormones affect BBB integrity. Finally, we summarize current in vitro BBB bioengineered models and highlight their potential to study sex differences in BBB integrity and neurodegenerative disease.
Collapse
Affiliation(s)
- Callie M. Weber
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, USA
| | - Alisa Morss Clyne
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland 20742, USA
| |
Collapse
|
45
|
Hayward GC, Baranowski BJ, Marko DM, MacPherson REK. Examining the effects of ovarian hormone loss and diet-induced obesity on Alzheimer's disease markers of amyloid-β production and degradation. J Neurophysiol 2021; 125:1068-1078. [PMID: 33534663 DOI: 10.1152/jn.00489.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
After menopause, women experience declines in ovarian sex hormones, an event that has recently been associated with increased amyloid-β peptides, a main feature of Alzheimer's disease. Diet-induced insulin resistance also increases amyloid-β peptides; however, whether this process is exacerbated with ovarian sex hormone loss remains unknown. Female C57BL6/J mice received either bilateral ovariectomy (OVX; n = 20) or remained intact (n = 20) at 24 wk of age and were placed on either a low- or high-fat diet (LFD, n = 10 for OVX and intact; HFD, n = 10 for OVX and intact) for 10 wk. Independently, OVX led to increases in the amyloidogenic marker, soluble amyloid precursor protein β (sAPPβ). The HFD in combination with OVX led to lower insulin degrading enzyme (IDE) protein content and activity in the prefrontal cortex, indicative of decreased amyloid-β degradation; however, no differences in amyloid-β content were observed. Data from this study provide novel evidence of independent effects of peripheral insulin resistance and ovarian sex hormone loss in decreasing brain markers of amyloid-β degradation. Furthermore, findings indicate how the loss of ovarian sex hormones can promote the formation of amyloidogenic APP cleavage products, independent of diet-induced insulin resistance.NEW & NOTEWORTHY This study provides novel insight into the effect of peripheral insulin resistance and ovarian hormone loss in decreasing brain markers of amyloid-β degradation. Results demonstrate that ovarian hormone loss through ovariectomy increased the amyloidogenic marker, sAPPβ, while the high-fat diet in combination with ovariectomy led to lower IDE protein content and activity in the prefrontal cortex, indicative of decreased amyloid-β degradation. These original results provide important information for future targets in early AD pathogenesis.
Collapse
Affiliation(s)
- Grant C Hayward
- Department of Health Sciences, Brock University, St Catharines, Ontario, Canada.,Centre for Neuroscience, Brock University, St. Catharines, Ontario, Canada.,Faculty of Medicine, Ottawa University, Ottawa, Ontario, Canada
| | - Bradley J Baranowski
- Department of Health Sciences, Brock University, St Catharines, Ontario, Canada.,Centre for Neuroscience, Brock University, St. Catharines, Ontario, Canada
| | - Daniel M Marko
- Department of Health Sciences, Brock University, St Catharines, Ontario, Canada.,Centre for Neuroscience, Brock University, St. Catharines, Ontario, Canada
| | - Rebecca E K MacPherson
- Department of Health Sciences, Brock University, St Catharines, Ontario, Canada.,Centre for Neuroscience, Brock University, St. Catharines, Ontario, Canada
| |
Collapse
|
46
|
Bayliak MM, Sorochynska OM, Kuzniak OV, Gospodaryov DV, Demianchuk OI, Vasylyk YV, Mosiichuk NM, Storey KB, Garaschuk O, Lushchak VI. Middle age as a turning point in mouse cerebral cortex energy and redox metabolism: Modulation by every-other-day fasting. Exp Gerontol 2020; 145:111182. [PMID: 33290862 DOI: 10.1016/j.exger.2020.111182] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 10/19/2020] [Accepted: 12/01/2020] [Indexed: 12/17/2022]
Abstract
Normal brain aging is accompanied by intensification of free radical processes and compromised bioenergetics. Caloric restriction is expected to counteract these changes but the underlying protective mechanisms remain poorly understood. The present work aimed to investigate the intensity of oxidative stress and energy metabolism in the cerebral cortex comparing mice of different ages as well as comparing mice given one of two regimens of food availability: ad libitum versus every-other-day fasting (EODF). Levels of oxidative stress markers, ketone bodies, glycolytic intermediates, mitochondrial respiration, and activities of antioxidant and glycolytic enzymes were assessed in cortex from 6-, 12- and 18-month old C57BL/6J mice. The greatest increase in oxidative stress markers and the sharpest decline in key glycolytic enzyme activities was observed in mice upon the transition from young (6 months) to middle (12 months) age, with smaller changes occurring upon transition to old-age (18 months). Brain mitochondrial respiration showed no significant changes with age. A decrease in the activities of key glycolytic enzymes was accompanied by an increase in the activity of glucose-6-phosphate dehydrogenase suggesting that during normal brain aging glucose metabolism is altered to lower glycolytic activity and increase dependence on the pentose-phosphate pathway. Interestingly, levels of ketone bodies and antioxidant capacity showed a greater decrease in the brain cortex of females as compared with males. The EODF regimen further suppressed glycolytic enzyme activities in the cortex of old mice, and partially enhanced oxygen consumption and respiratory control in the cortex of middle aged and old males. Thus, in the mammalian cortex the major aging-induced metabolic changes are already seen in middle age and are slightly alleviated by an intermittent fasting mode of feeding.
Collapse
Affiliation(s)
- Maria M Bayliak
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenko Str., Ivano-Frankivsk 76018, Ukraine
| | - Oksana M Sorochynska
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenko Str., Ivano-Frankivsk 76018, Ukraine
| | - Oksana V Kuzniak
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenko Str., Ivano-Frankivsk 76018, Ukraine
| | - Dmytro V Gospodaryov
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenko Str., Ivano-Frankivsk 76018, Ukraine
| | - Oleh I Demianchuk
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenko Str., Ivano-Frankivsk 76018, Ukraine
| | - Yulia V Vasylyk
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenko Str., Ivano-Frankivsk 76018, Ukraine
| | - Nadia M Mosiichuk
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenko Str., Ivano-Frankivsk 76018, Ukraine
| | - Kenneth B Storey
- Department of Biology, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario K1S 5B6, Canada
| | - Olga Garaschuk
- Department of Neurophysiology, University of Tübingen, 72074 Tübingen, Germany
| | - Volodymyr I Lushchak
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenko Str., Ivano-Frankivsk 76018, Ukraine.
| |
Collapse
|
47
|
Shin J, Pelletier S, Richer L, Pike GB, Gaudet D, Paus T, Pausova Z. Adiposity-related insulin resistance and thickness of the cerebral cortex in middle-aged adults. J Neuroendocrinol 2020; 32:e12921. [PMID: 33340164 PMCID: PMC8132297 DOI: 10.1111/jne.12921] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 11/13/2020] [Accepted: 11/13/2020] [Indexed: 12/14/2022]
Abstract
The thickness of the cerebral cortex decreases with ageing. Recent research suggests that obesity and type 2 diabetes mellitus may accelerate this cortical thinning, and that obesity-related insulin resistance may be a shared mechanistic pathway. Ageing of the cerebral cortex demonstrates sex-specific trajectories, with a gradual shift towards accelerated thinning beginning in midlife. Here, we investigated whether adiposity-related insulin resistance is associated with lower thickness of the human cerebral cortex in a community-based sample of middle-aged adults. We studied 533 adult participants (36-65 years) from the Saguenay Youth Study. Adiposity was assessed with bioimpedance, and insulin resistance was evaluated from a fasting blood sample with the homeostatic model assessment of insulin resistance (HOMA-IR). Associations between adiposity-related insulin resistance (adiposity/IR) and cortical thickness were assessed with linear models, separately in males and females younger or older than 50 years. Potential biological underpinnings were investigated with virtual histology. Adiposity/IR was associated with lower cortical thickness in females older than 50 years but not in males or younger females. The strength of the association varied across the cerebral cortex, with regions of the lateral frontal and parietal cortices and the superior temporal cortex demonstrating most pronounced thinning. Based on virtual histology, adiposity/IR-related cortical thinning may involve neurones, astrocytes, oligodendrocytes and ependymal cells acting so that they lower the cortical potential for synaptogenesis, formation of dendritic spines, production of extracellular matrix and myelination. Adiposity-related insulin resistance is associated with lower cortical thickness in middle-aged women older than 50 years. This aspect of thinning may involve neuronal and glial cells in a way that lowers the capacity of the cerebral cortex for neuronal plasticity and maintenance of myelination.
Collapse
Affiliation(s)
- Jean Shin
- The Hospital for Sick Children, University of Toronto, Toronto, Canada
- Departments of Physiology and Nutritional Sciences, University of Toronto, Toronto, Canada
| | - Stephanie Pelletier
- The Hospital for Sick Children, University of Toronto, Toronto, Canada
- Departments of Physiology and Nutritional Sciences, University of Toronto, Toronto, Canada
| | - Louis Richer
- Department of Health Sciences, Université du Québec à Chicoutimi, Chicoutimi, Canada
| | - G. Bruce Pike
- Department of Radiology and Clinical Neurosciences, University of Calgary, Calgary, Canada
| | - Daniel Gaudet
- Lipidology Unit, Community Genomic Medicine Centre and ECOGENE-21, Department of Medicine, Université de Montréal, Saguenay, Canada
| | - Tomas Paus
- Bloorview Research Institute, Holland Bloorview Kids Rehabilitation Hospital, Toronto, Canada
- Departments of Psychology and Psychiatry, University of Toronto, Toronto, Canada
| | - Zdenka Pausova
- The Hospital for Sick Children, University of Toronto, Toronto, Canada
- Departments of Physiology and Nutritional Sciences, University of Toronto, Toronto, Canada
| |
Collapse
|
48
|
Mishra A, Shang Y, Wang Y, Bacon ER, Yin F, Brinton RD. Dynamic Neuroimmune Profile during Mid-life Aging in the Female Brain and Implications for Alzheimer Risk. iScience 2020; 23:101829. [PMID: 33319170 PMCID: PMC7724165 DOI: 10.1016/j.isci.2020.101829] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 10/13/2020] [Accepted: 11/16/2020] [Indexed: 12/24/2022] Open
Abstract
Aging and endocrine transition states can significantly impact inflammation across organ systems. Neuroinflammation is well documented in Alzheimer disease (AD). Herein, we investigated neuroinflammation that emerges during mid-life aging, chronological and endocrinological, in the female brain as an early initiating mechanism driving AD risk later in life. Analyses were conducted in a translational rodent model of mid-life chronological and endocrinological aging followed by validation in transcriptomic profiles from women versus age-matched men. In the translational model, the neuroinflammatory profile of mid-life aging in females was endocrine and chronological state specific, dynamic, anatomically distributed, and persistent. Microarray dataset analyses of aging human hippocampus indicated a sex difference in neuroinflammatory profile in which women exhibited a profile comparable to the pattern discovered in our translational rodent model, whereas age-matched men exhibited a profile consistent with low neuroimmune activation. Translationally, these findings have implications for therapeutic interventions during mid-life to decrease late-onset AD risk.
Collapse
Affiliation(s)
- Aarti Mishra
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ 85719, USA
| | - Yuan Shang
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ 85719, USA
| | - Yiwei Wang
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ 85719, USA
| | - Eliza R Bacon
- Department of Medical Oncology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Fei Yin
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ 85719, USA
| | - Roberta D Brinton
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ 85719, USA
| |
Collapse
|
49
|
Wang YX, Xia ZH, Jiang X, Li LX, Wang HG, An D, Liu YQ. Genistein inhibits amyloid peptide 25-35-induced neuronal death by modulating estrogen receptors, choline acetyltransferase and glutamate receptors. Arch Biochem Biophys 2020; 693:108561. [PMID: 32857999 DOI: 10.1016/j.abb.2020.108561] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 07/31/2020] [Accepted: 08/22/2020] [Indexed: 01/06/2023]
Abstract
PURPOSE To explore genistein, the most active component of soy isoflavones, on viability, expression of estrogen receptor (ER) subtypes, choline acetyltransferase (ChAT), and glutamate receptor subunits in amyloid peptide 25-35-induced hippocampal neurons, providing valuable data and basic information for neuroprotective effect of genistein in Aβ25-35-induced neuronal injury. METHODS We established an in vitro model of Alzheimer's disease by exposing primary hippocampal neurons of newborn rats to amyloid peptide 25-35 (20 μM) for 24 h and observing the effects of genistein (10 μM, 3 h) on viability, expression of ER subtypes, ChAT, NMDA receptor subunit NR2B and AMPA receptor subunit GluR2 in Aβ25-35-induced hippocampal neurons. RESULTS We found that amyloid peptide 25-35 exposure reduced the viability of hippocampal neurons. Meanwhile, amyloid peptide 25-35 exposure decreased the expression of ER subtypes, ChAT and GluR2, and increased the expression of NR2B. Genistein at least partially reversed the effects of amyloid peptide 25-35 in hippocampal neurons. CONCLUSION Genistein could increase the expression of ChAT as a consequence of activating estrogen receptor subtypes, modulating the expression of NR2B and GluR2, and thereby ameliorating the status of hippocampal neurons and exerting neuroprotective effects against amyloid peptide 25-35. Our data suggest that genistein might represent a potential cell-targeted therapy which could be a promising approach to treating AD.
Collapse
Affiliation(s)
- Yu-Xiang Wang
- Department of Zoology and Developmental Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China; Department of Immunology and Pathogenic Biology, School of Basic Medical Sciences, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, China
| | - Zhen-Hong Xia
- Department of Zoology and Developmental Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Xue Jiang
- Department of Zoology and Developmental Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Li-Xia Li
- Department of Zoology and Developmental Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Hong-Gang Wang
- Department of Zoology and Developmental Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Di An
- Department of Zoology and Developmental Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Yan-Qiang Liu
- Department of Zoology and Developmental Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
50
|
Hill LK, Hoang DM, Chiriboga LA, Wisniewski T, Sadowski MJ, Wadghiri YZ. Detection of Cerebrovascular Loss in the Normal Aging C57BL/6 Mouse Brain Using in vivo Contrast-Enhanced Magnetic Resonance Angiography. Front Aging Neurosci 2020; 12:585218. [PMID: 33192479 PMCID: PMC7606987 DOI: 10.3389/fnagi.2020.585218] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 09/11/2020] [Indexed: 12/28/2022] Open
Abstract
Microvascular rarefaction, or the decrease in vascular density, has been described in the cerebrovasculature of aging humans, rats, and, more recently, mice in the presence and absence of age-dependent diseases. Given the wide use of mice in modeling age-dependent human diseases of the cerebrovasculature, visualization, and quantification of the global murine cerebrovasculature is necessary for establishing the baseline changes that occur with aging. To provide in vivo whole-brain imaging of the cerebrovasculature in aging C57BL/6 mice longitudinally, contrast-enhanced magnetic resonance angiography (CE-MRA) was employed using a house-made gadolinium-bearing micellar blood pool agent. Enhancement in the vascular space permitted quantification of the detectable, or apparent, cerebral blood volume (aCBV), which was analyzed over 2 years of aging and compared to histological analysis of the cerebrovascular density. A significant loss in the aCBV was detected by CE-MRA over the aging period. Histological analysis via vessel-probing immunohistochemistry confirmed a significant loss in the cerebrovascular density over the same 2-year aging period, validating the CE-MRA findings. While these techniques use widely different methods of assessment and spatial resolutions, their comparable findings in detected vascular loss corroborate the growing body of literature describing vascular rarefaction aging. These findings suggest that such age-dependent changes can contribute to cerebrovascular and neurodegenerative diseases, which are modeled using wild-type and transgenic laboratory rodents.
Collapse
Affiliation(s)
- Lindsay K. Hill
- Department of Chemical and Biomolecular Engineering, NYU Tandon School of Engineering, Brooklyn, NY, United States
- Department of Radiology, Center for Advanced Imaging Innovation and Research (CAI2R), NYU Grossman School of Medicine, New York, NY, United States
- Department of Radiology, Bernard and Irene Schwartz Center for Biomedical Imaging, NYU Grossman School of Medicine, New York, NY, United States
- Department of Biomedical Engineering, SUNY Downstate Medical Center, Brooklyn, NY, United States
| | - Dung Minh Hoang
- Department of Radiology, Center for Advanced Imaging Innovation and Research (CAI2R), NYU Grossman School of Medicine, New York, NY, United States
- Department of Radiology, Bernard and Irene Schwartz Center for Biomedical Imaging, NYU Grossman School of Medicine, New York, NY, United States
| | - Luis A. Chiriboga
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, United States
| | - Thomas Wisniewski
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, United States
- Department of Neurology, NYU Grossman School of Medicine, New York, NY, United States
- Department of Psychiatry, NYU Grossman School of Medicine, New York, NY, United States
| | - Martin J. Sadowski
- Department of Neurology, NYU Grossman School of Medicine, New York, NY, United States
- Department of Psychiatry, NYU Grossman School of Medicine, New York, NY, United States
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY, United States
| | - Youssef Z. Wadghiri
- Department of Radiology, Center for Advanced Imaging Innovation and Research (CAI2R), NYU Grossman School of Medicine, New York, NY, United States
- Department of Radiology, Bernard and Irene Schwartz Center for Biomedical Imaging, NYU Grossman School of Medicine, New York, NY, United States
| |
Collapse
|